Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
TETRAHYDROISOQUINOLINE HETEROBIFUNCTIONAL BCL-XL DEGRADERS
Document Type and Number:
WIPO Patent Application WO/2023/215482
Kind Code:
A1
Abstract:
This disclosure provides compounds of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof that induce degradation of a BCL-XL protein. These compounds are useful, for example, for treating a cancer in a subject (e.g., a human). This disclosure also provides compositions containing the compounds provided herein as well as methods of using and making the same.

Inventors:
HORN EVAN J (US)
HANSEN JOSHUA D (US)
ALEXANDER MATTHEW D (US)
HUANG FEI (US)
NAGY MARK A (US)
Application Number:
PCT/US2023/021010
Publication Date:
November 09, 2023
Filing Date:
May 04, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
TREELINE BIOSCIENCES INC (US)
International Classes:
C07D417/14; A61K31/4725; A61K47/50; A61P35/00; C07D451/06; C07D487/08
Domestic Patent References:
WO2013055897A12013-04-18
WO2021146536A12021-07-22
WO2023044046A12023-03-23
WO2023083194A12023-05-19
WO2017184995A12017-10-26
WO2019144117A12019-07-25
WO2020163823A22020-08-13
WO2021078301A12021-04-29
WO2021146536A12021-07-22
WO2021007307A12021-01-14
WO2021222114A12021-11-04
WO2022169780A12022-08-11
WO2023044046A12023-03-23
Foreign References:
US199762634579P
US194562634545P
US195662634497P
US196262634298P
US198362633987P
US195362633392P
US20070027135A12007-02-01
US20100305122A12010-12-02
US20070027135A12007-02-01
US20130096120A12013-04-18
Other References:
TROUP ROBERT I. ET AL: "Current strategies for the design of PROTAC linkers: a critical review", EXPLORATION OF TARGETED ANTI-TUMOR THERAPY, vol. 1, no. 5, 30 October 2020 (2020-10-30), XP055828975, Retrieved from the Internet [retrieved on 20230623], DOI: 10.37349/etat.2020.00018
ADAMSCORY, CELL DEATH & DIFFERENTIATION, vol. 25, no. 1, 2018, pages 27 - 36
CHAMBERLAINHAMANN, NATURE CHEMICAL BIOLOGY, vol. 15, no. 10, 2019, pages 937 - 944
HE, YONGHAN ET AL., JOURNAL OF HEMATOLOGY & ONCOLOGY, vol. 13, no. 1, 2020, pages 1 - 13
WU ET AL., NATURE STRUCTURAL & MOLECULAR BIOLOGY, vol. 27, no. 7, 2020, pages 605 - 614
DONG ET AL., JOURNAL OF MEDICINALCHEMISTRY, vol. 64, no. 15, 2021, pages 10606 - 10620
YANG ET AL., TARGETED ONCOLOGY, vol. 16, no. 1, 2021, pages 1 - 12
LV ET AL., NATURE COMMUNICATIONS, vol. 12, no. 1, 2021, pages 6896
GUO ET AL., AGING (ALBANY NY), vol. 13, no. 15, 2021, pages 19750
PARK ET AL., PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 112, no. 40, 2015, pages 12492 - 12497
ZHANG, HAICHAO ET AL., MOLECULAR CANCER, vol. 14, no. 1, 2015, pages 1 - 9
BEROUKHIM ET AL., NATURE, vol. 463, no. 7283, 2010, pages 899 - 905
KHAN ET AL., NATURE MEDICINE, vol. 25, no. 12, 2019, pages 1938 - 1947
BALACHANDER ET AL., CLINICAL CANCER RESEARCH, vol. 26, no. 24, 2020, pages 6535 - 6549
PHILLIPS, D. C. ET AL., BLOOD CANCER JOURNAL, vol. 5, no. 11, 2015, pages e368 - e368
XIAO, YU ET AL., MOLECULAR CANCER THERAPEUTICS, vol. 14, no. 8, 2015, pages 1837 - 1847
SOUERS, ANDREW J. ET AL., NATURE MEDICINE, vol. 19, no. 2, 2013, pages 202 - 208
MOREAU, KEVIN ET AL., BRITISH JOURNALOF PHARMACOLOGY, vol. 177, no. 8, 2020, pages 1709 - 1718
CAMPBELLTAIT, OPEN BIOLOGY, vol. 8, no. 5, 2018, pages 180002
PULLARKAT ET AL., CANCER DISCOVERY, 2021
NEGIVOISIN-CHIRET, CHEMBIOCHEM, 2022
WILSON, WYNDHAM H ET AL., THE LANCET ONCOLOGY, vol. 11, no. 12, 2010, pages 1149 - 1159
KEITEL, ULRIKE ET AL., ONCOTARGET, vol. 5, no. 23, 2014, pages 11778
CHONGHAILETRIONA NI ET AL., CANCER DISCOVERY, vol. 4, no. 9, 2014, pages 1074 - 1087
ZAANAN, AZIZ ET AL., JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 290, no. 39, 2015, pages 23838 - 23849
SODERQUIST, RYAN S. ET AL., NATURE COMMUNICATIONS, vol. 9, no. 1, 2018, pages 1 - 13
STOVER, ELIZABETH H. ET AL., MOLECULAR CANCER RESEARCH, vol. 17, no. 11, 2019, pages 2281 - 2293
CONCORAN, R.B. ET AL., ANNALS OF ONCOLOGY, vol. 30, no. 5, 2019, pages v159 - v193
LAKHANI, NEHAL J. ET AL., JOURNAL OF CLINICAL ONCOLOGY, 2020, pages 3509 - 3509
GRUBB, TREG ET AL., CLINICAL CANCER RESEARCH, 2022
JOLY, FLORENCE ET AL., GYNECOLOGIC ONCOLOGY, vol. 165, no. 1, 2022, pages 30 - 39
NANJO, SHIGEKI ET AL., THE JOURNAL OF CLINICAL INVESTIGATION, 2022
CRESPO-GARCIA, SERGIO ET AL., CELL METABOLISM, vol. 33, no. 4, 2021, pages 818 - 832
HASSANJANNAH WALEDASHAY D. BHATWADEKAR, FRONTIERS IN PHARMACOLOGY, vol. 13, 2022, pages 896907
ZHU, Y. 1. ET AL., AGING CELL, vol. 14, no. 4, 2015, pages 644 - 658
ZHU, Y. I. ET AL., AGING CELL, vol. 15, no. 3, 2016, pages 428 - 435
CHANG, JIANHUI ET AL., NATURE MEDICINE, vol. 22, no. 1, 2016, pages 78 - 83
ZHU, YI ET AL., AGING (ALBANY NY), vol. 9, no. 3, 2017, pages 955
LAGARES, DAVID ET AL., SCIENCE TRANSLATIONAL MEDICINE, vol. 9, no. 420, 2017, pages 13765
PAN, JIN ET AL., INTERNATIONAL JOURNAL OF RADIATION ONCOLOGY BIOLOGY PHYSICS, vol. 99, no. 2, 2017, pages 353 - 361
BUSSIAN, TYLER J. ET AL., NATURE, vol. 562, no. 7728, 2018, pages 578 - 582
MONCSEK, ANJA ET AL., HEPATOLOGY, vol. 67, no. 1, 2018, pages 247 - 259
VAN WILLIGENBURG, HESTERPETER LJ DE KEIZERRON WF DE BRUIN, PHARMACOLOGICAL RESEARCH, vol. 130, 2018, pages 322 - 330
WALASZCZYK, ANNA ET AL., AGING CELL, vol. 18, no. 3, 2019, pages el2945
AGUAYO-MAZZUCATO, CRISTINA ET AL., CELL METABOLISM, vol. 30, no. 1, 2019, pages 129 - 142
SESSIONS, GARRETT A. ET AL., THEFASEB JOURNAL, vol. 33, no. 11, 2019, pages 12364
GERDES, ERIN O. WISSLER ET AL., INTERNATIONAL REVIEW OF NEUROBIOLOGY, vol. 155, 2020, pages 203 - 234
SASAKI, MOTOKOYASUNORI SATOYASUNI NAKANUMA, JOURNAL OF AUTOIMMUNITY, vol. 107, 2020, pages 102377
YABLUCHANSKIY, ANDRIY ET AL., GEROSCIENCE, vol. 42, 2020, pages 409 - 428
DOOKIIN, EMILY ET AL., AGING CELL, vol. 19, no. 10, 2020, pages e13249
JIA, KANGNI ET AL., JOURNAL OF CARDIOVASCULAR PHARMACOLOGY, vol. 76, no. 4, 2020, pages 452 - 460
SIERRA-RAMIREZ, ARANTZAZU ET AL., AGING (ALBANY NY), vol. 12, no. 12, 2020, pages 11337
YANG, HAO ET AL., AGING (ALBANY NY, vol. 12, no. 13, 2020, pages 12750
LAWRIE, ALLANSHEILAE. FRANCIS, THE JOURNAL OF CLINICAL INVESTIGATION, vol. 131, no. 11, 2021, pages 149721
PARAMOS-DE-CARVALHO, DIOGO ET AL., CELL REPORTS, vol. 36, no. 1, 2021, pages 109334
TARANTINI, STEFANO ET AL., GEROSCIENCE, vol. 43, no. 5, 2021, pages 2427 - 2440
PARK, JI HEE ET AL., THE BRITISH JOURNAL O DERMATOLOGY, 2021
FIELDER, EDWARD ET AL., ELIFE, vol. 11, 2022, pages e75492
SUZUKI, KEIJI ET AL., MUTATION RESEARCH/GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS, vol. 8, no. 6, 2022, pages 503448
HE, AN ET AL., AMERICAN JOURNAL OF TRANSPLANTATION, vol. 22, no. 11, 2022, pages 2529 - 2547
JOHNSON, LAURA A. ET AL., INFLAMMATORY BOWEL DISEASES, vol. 28, no. 2, 2022, pages 161 - 175
MIURA, YUGO ET AL., STEM CELL RESEARCH & THERAPY, vol. 13, no. 1, 2022, pages 222
COOLEY, JOSEPH C. ET AL., JCI INSIGHT, vol. 8, no. 3, 2023, pages el63762
WATANABE, YUSUKE ET AL., HEPATOLOGY RESEARCH, vol. 53, 2023, pages 460 - 472
TAKAYA, KENTO ET AL., REJUVENATION RESEARCH, vol. 26, no. 1, 2023, pages 9 - 20
Attorney, Agent or Firm:
KENDALL, John T. et al. (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS: 1. A compound of Formula (I): Formula (I) or a pharmaceutically acceptable salt thereof, wherein: Ring A is selected from the group consisting of: (a) C3-15 cycloalkylene or 3-15 membered heterocyclylene, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: Ra and Rb; and (b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 substituents independently selected from the group consisting of: Ra and Rb; R1 is selected from the group consisting of: (a) C(O)OH, (b) C(O)NRdRe, and (c) C(O)OC1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with 1-3 Rc; each R2, R3, R4, and R5 is independently selected from the group consisting of: halo, CN, C1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, C1-3 haloalkoxy, OH, and NRdRe; m2 is 0, 1, or 2; m3 and m4 are independently 0, 1, 2, or 3; m5 is 0, 1, 2, 3, or 4; L is –(LA)n1–, wherein LA and n1 are defined according to (AA) or (BB): n1 is an integer from 3 to 15; and each LA is independently selected from the group consisting of: LA1, LA3, and LA4, provided that 1-3 occurrences of LA is LA4; n1 is an integer from 0 to 20; and each LA is independently selected from the group consisting of: LA1 and LA3; each LA1 is independently selected from the group consisting of: -CH2-, -CHRL-, and - C(RL)2-; each LA3 is independently selected from the group consisting of: -N(Rd)-, -N(Rb)-, -O- , -S(O)0-2-, and C(=O); each LA4 is independently selected from the group consisting of: (a) C3-15 cycloalkylene or 3-15 membered heterocyclylene, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: Ra and Rb; and (b) C6-15 arylene or 5-15 membered heteroarylene, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: Ra and Rb; provided that L does not contain any N-O, O-O, N-N, N-S(O)0, or O-S(O)0-2 bonds; wherein each RL is independently selected from the group consisting of: halo; cyano; - OH; -C1-6 alkoxy; -C1-6 haloalkoxy; -NRdRe; C(=O)N(Rf)2; S(O)0-2(C1-6 alkyl); S(O)0-2(C1-6 haloalkyl); S(O)1-2N(Rf)2; -Rb; and C1-6 alkyl optionally substituted with 1-6 Rc; Ring C is selected from the group consisting of: ; c1 is 0, 1, 2, or 3; each RY is independently selected from the group consisting of: Ra and Rb; RaN is H or C1-6 alkyl optionally substituted with 1-3 Rc; Y1 and Y2 are independently N, CH, or CRY; yy represents the point of attachment to L; X is CH, C, or N; the is a single bond or a double bond; LC is selected from the group consisting of: a bond, -CH2-, -CHRa-, -C(Ra)2-, -N(Rd)-, and O, provided that when X is N, then LC is other than O; each Ra is independently selected from the group consisting of: (a) halo; (b) cyano; (c) -OH; (d) oxo; (e) -C1-6 alkoxy; (f) -C1-6 haloalkoxy; (g) -NRdRe; (h) C(=O)C1-6 alkyl; (i) C(=O)C1-6 haloalkyl; (j) C(=O)OH; (k) C(=O)OC1-6 alkyl; (l) C(=O)OC1-6 haloalkyl; (m) C(=O)N(Rf)2; (n) S(O)0-2(C1-6 alkyl); (o) S(O)0-2(C1-6 haloalkyl); (p) S(O)1-2N(Rf)2; and (q) C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted with 1-6 Rc; each Rb is independently selected from the group consisting of: -(Lb)b-Rb1 and -Rb1, wherein: each b is independently 1, 2, or 3; each -Lb is independently selected from the group consisting of: -O-; -N(H)-; -N(C1-3 alkyl)-; -S(O)0-2-; C(=O); and C1-3 alkylene; and each Rb1 is independently selected from the group consisting of: C3-10 cycloalkyl, 4-10 membered heterocyclyl, C6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 Rg; each Rc is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C1-6 haloalkoxy; -NRdRe; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC1-6 haloalkyl; C(=O)OH; C(=O)N(Rf)2; S(O)0-2(C1-6 alkyl); S(O)0-2(C1-6 haloalkyl); and S(O)1-2N(Rf)2; each Rd and Re is independently selected from the group consisting of: H; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC1-6 haloalkyl; C(=O)N(Rf)2; S(O)1- 2(C1-6 alkyl); S(O)1-2(C1-6 haloalkyl); S(O)1-2N(Rf)2; and C1-6 alkyl optionally substituted with 1-3 Rh; each Rf is independently selected from the group consisting of: H and C1-6 alkyl optionally substituted with 1-3 Rh; each Rg is independently selected from the group consisting of: Rh; C1-3 alkyl; and C1- 3 haloalkyl; and each Rh is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C1-6 haloalkoxy; -NH2, -N(H)(C1-3 alkyl); and -N(C1-3 alkyl)2. 2. The compound of claim 1, wherein Ring A is a phenylene optionally substituted with 1-3 Ra. 3. The compound of claims 1 or 2, wherein Ring , wherein aa represents the point of attachment to L; or Ring A is , wherein aa represents the point of attachment to L; or Ring , wherein aa represents the point of attachment to L. 4. The compound of claim 1, wherein Ring A is 5-6 membered heteroarylene optionally substituted with 1-3 Ra. 5. The compound of any one of claims 1-4, wherein one Ra present on Ring A is C1-3 alkyl optionally substituted with 1-3 F; or one Ra present on Ring A is methyl or CF3. 6. The compound of any one of claims 1-5, wherein R1 is C(O)OH. 7. The compound of any one of claims 1-6, wherein m2 is 0. 8. The compound of any one of claims 1-7, wherein m3 is 0. 9. The compound of any one of claims 1-8, wherein m4 is 0. 10. The compound of any one of claims 1-9, wherein m5 is 0. 11. The compound of any one of claims 1-6, wherein m2 is 0; m3 is 0; m4 is 0; and m5 is 0. 12. The compound of any one of claims 1-11, wherein Ring ; or Ring Ring . 13. The compound of claim 12, wherein c1 is 0. 14. The compound of any one of claims 12 or 13, wherein RaN is C1-3 alkyl; or RaN is methyl. 15. The compound of any one of claims 12-14, wherein LC is a bond. 16. The compound of any one of claims 12-15, wherein X is CH. 17. The compound of any one of claims 1-12, wherein the . 18. The compound of any one of claims 1-17, wherein L is – ; and LA and n1 are defined according to (AA). 19. The compound of any one of claims 1-18, wherein n1 is an integer from 3 to 5; or n1 is an integer from 5 to 9; or n1 is 6, 7, or 8; or n1 is an integer from 9 to 12. 20. The compound of any one of claims 1-19, wherein 1-2 occurrences of LA is LA4; or one occurrence of LA is LA4; or two occurrences of LA are LA4.

21. The compound of any one of claims 1-20, wherein each LA4 is independently selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 Ra; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 Ra. 22. The compound of any one of claims 1-21, wherein 1-4 occurrences of LA is LA3; or 1-3 occurrences of LA is LA3. 23. The compound of any one of claims 1-22, wherein 0-1 occurrence of LA3 is C(=O); and each remaining occurrence of LA3 is independently selected from the group consisting of: -O-; -N(H)-; and -N(C1-3 alkyl)-. 24. The compound of any one of claims 1-23, wherein 2-7 occurrences of LA is LA1; or 2-5 occurrences of LA is LA1. 25. The compound of any one of claims 1-24, wherein 0-2 occurrences of LA1 is - CHRL- or -C(RL)2-; and each remaining occurrence of LA1 is -CH2-. 26. The compound of any one of claims 1-25, wherein each occurrence of LA1 is - CH2-; or . one occurrence of LA1 is -CHRL- or -C(RL)2-; and each remaining occurrence of LA1 is -CH2-. 27. The compound of any one of claims 1-26, wherein each RL is independently selected from the group consisting of: -F and -C1-3 alkyl optionally substituted with 1-3 F. 28. The compound of any one of claims 1-18, wherein L is –(LA)n1-, and LA and n1 are defined according to (AA): n1 is an integer from 5 to 9; 1-2 occurrences of LA is LA4; 2-7 occurrences of LA are LA1; and 1-3 occurrences of LA is LA3.

29. The compound of any one of claims 1-18, wherein L is selected from the group consisting of: (ii) –(LA3)0-2-(LA1)0-5-LA4-(LA1)0-5-LA4-bb; (iii) –(LA3)0-2-(LA1)0-5-LA4-(LA1)0-5-LA3-LA4-bb; and (iv) –(LA3)0-1-(LA1)2-3-LA3-LA4-(LA1)0-5-(LA3)1-2-bb; provided that L contains 1-7 LA1; and wherein bb represents the point of attachment to Ring C. 30. The compound of any one of claims 1-18, wherein L is: –(LA3)0-2-(LA1)2-9-(LA3)0-1-LA4-bb, wherein bb represents the point of attachment to Ring C. 31. The compound of claim 28-30, wherein each LA4 is independently selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 Ra; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 Ra. 32. The compound of any one of claims 28-31, wherein 0-1 occurrence of LA1 is - CHRL- or -C(RL)2-; and each remaining occurrence of LA1 is -CH2-. 33. The compound of any one of claims 28-32, wherein 0-1 occurrence of LA3 is C(=O); and each remaining occurrence of LA3 is independently selected from the group consisting of: -O-; -N(H)-; and -N(C1-3 alkyl)-. 34. The compound of any one of claims 1-18, wherein L is: –(LA3)0-2-(LA1)0-5-LA4-(LA1)0-5-(LA3)1-2- bb; provided that L contains 1-7 LA1; and wherein bb represents the point of attachment to Ring C. 35. The compound of any one of claims 1-18 or 34, wherein L is: –(LA3)0-1-(LA1)0-5-LA4-(LA1)0-5-(LA3)2- bb; provided that L contains 1-5 LA1; and wherein bb represents the point of attachment to Ring C. 36. The compound of any one of claims 1-18 or 34-35, wherein L is: –(LA3)0-1-(LA1)0-5-LA4-(LA1)0-5-C(=O)-N(Rd)-bb; provided that L contains 1-5 LA1; and wherein bb represents the point of attachment to Ring C. 37. The compound of any one of claims 34-36, wherein L is a divalent group of Formula (L-1): Formula (L-1) wherein: a3 is 0 or 1; LA3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5; LA1a and LA1b are independently selected from the group consisting of: -CH2-, -CHRL- , and -C(RL)2-; LA4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 Ra; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 Ra; and bb represents the point of attachment to Ring C. 38. The compound of claim 37, wherein a3 is 1. 39. The compound of claims 37 or 38, wherein LA3 is -O-. 40. The compound of claim 37, wherein a3 is 0.

41. The compound of any one of claims 37-40, wherein a1a + a1b is 3 or 4; or a1a + a1b is 2 or 5. 42. The compound of any one of claims 38-40, wherein a1a + a1b is 2 to 5. 43. The compound of any one of claims 1-18 or 34, wherein L is: –(LA3)0-1-(LA1)0-5-LA4-(LA1)0-5-LA3-bb; provided that L contains 2-7 LA1; and wherein bb represents the point of attachment to Ring C. 44. The compound of claims 34 or 43, wherein L is a divalent group of Formula (L- 2): wherein: a3a is 0 or 1; LA3a and LA3b are independently selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 7; LA1a and LA1b are independently selected from the group consisting of: -CH2-, -CHRL- , and -C(RL)2-; LA4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 Ra; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 Ra; and bb represents the point of attachment to Ring C. 45. The compound of claim 44, wherein a3a is 1. 46. The compound of claims 44 or 45, wherein LA3a is -O-.

47. The compound of any one of claims 44-46, wherein LA3b is -N(H)- or –N(C1-3 alkyl)-. 48. The compound of any one of claims 44-46, wherein LA3b is -O-. 49. The compound of any one of claims 44-48, wherein a1a + a1b is 2, 3, or 4. 50. The compound of any one of claims 44-48, wherein a1a + a1b is 5 or 6. 51. The compound of any one of claims 1-18, wherein L is: –(LA3)0-1-(LA1)0-5-LA4-(LA1)0-5-LA4-bb; or –(LA3)0-1-(LA1)0-5-LA4-(LA1)0-5-LA3-LA4-bb; provided that L contains 2-7 LA1; and wherein bb represents the point of attachment to Ring C. 52. The compound of claim 51, wherein L is: –(LA3)0-1-(LA1)0-5-LA4-(LA1)0-5-(piperazinylene)-bb; –(LA3)0-1-(LA1)0-5-LA4-(LA1)0-5-(piperidinylene)-bb; or –(LA3)0-1-(LA1)0-5-LA4-(LA1)0-5-C(=O)-(piperazinylene)-bb; provided that L contains 2-5 LA1; wherein the piperazinylene and piperidinylene are each optionally substituted with 1-3 Ra, and wherein bb represents the point of attachment to Ring C. 53. The compound of claim 51, wherein L is a divalent group of Formula (L-3), (L- 3a), (L-3b),or (L-4): Formula (L-3a)

Formula (L-4) wherein: a3 is 0 or 1; LA3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; LA1a and LA1b are independently selected from the group consisting of: -CH2-, -CHRL- , and -C(RL)2-; LA4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 Ra; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 Ra; n2 and n3 are independently 0, 1, or 2; m8 is 0, 1, or 2; LA4b is 7-10 membered bicyclic nitrogen-containing heterocyclylene optionally substituted with 1-3 Ra; and bb represents the point of attachment to Ring C. 54. The compound of claim 53, wherein a3 is 1. 55. The compound of claims 53 or 54, wherein LA3 is -O-. 56. The compound of any one of claims 53-55, wherein a1a + a1b is 3 or 4. 57. The compound of any one of claims 53-55, wherein a1a + a1b is 2.

58. The compound of any one of claims 37-42, 44-50, or 53-57, wherein each occurrence of LA1a and LA1b is -CH2-. 59. The compound of any one of claims 37-42, 44-50, or 53-57, wherein one occurrence of LA1a is -CHRL- or -C(RL)2-; each remaining occurrence of LA1a is -CH2-; and each occurrence of LA1b is -CH2-. 60. The compound of any one of claims 37-42, 44-50, or 53-57, wherein one occurrence of LA1b is -CHRL- or -C(RL)2-; each remaining occurrence of LA1b is -CH2-; and each occurrence of LA1a is -CH2-. 61. The compound of any one of claims 34-60, wherein LA4 is 4-10 membered heterocyclylene optionally substituted with 1-3 Ra. 62. The compound of any one of claims 34-60, wherein LA4 is 4-10 membered nitrogen-containing heterocyclylene optionally substituted with 1-3 Ra. 63. The compound of any one of claims 34-62, wherein LA4 is selected from the optionally substituted with 1-3 Ra at one or more ring carbon atoms, wherein cc represents the point of attachment to LA1b. 64. The compound of claims 62 or 63, wherein each Ra present on LA4 is independently F or C1-3 alkyl optionally substituted with 1-3 F. 65. The compound of any one of claims 34-61, wherein LA4 is C3-10 cycloalkylene optionally substituted with 1-3 Ra. 66. The compound of any one of claims 34-61 or 65, wherein LA4 is 1,4- cyclohexylene optionally substituted with 1-3 Ra.

67. The compound of any one of claims 34-60, wherein LA4 is phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 Ra. 68. The compound of any one of claims 35-60 or 67, wherein LA4 is: (a) phenylene optionally substituted with 1-3 Ra; (b) 1,2-phenylene optionally substituted with 1-3 Ra; or (c) 1,4-phenylene optionally substituted with 1-3 Ra. 69. The compound of any one of claims 1-18 or 30, wherein L is: –(LA3)0-1-(LA1)2-9-(LA3)0-1-(piperazinylene)-bb, wherein bb represents the point of attachment to Ring C. 70. The compound of claim 69, wherein L is a divalent group of Formula (L-6): Formula (L-6) wherein: LA3a is selected from the group consisting of: -O-, -N(H)-, and -N(C1-3 alkyl)-; a3a is 0 or 1; a1 is an integer from 2 to 11; a3c is 0 or 1; and bb represents the point of attachment to Ring C. 71. The compound of claim 70, wherein a3a is 1. 72. The compound of claims 70 or 71, wherein LA3a is -O-. 73. The compound of any one of claims 70-72, wherein 0-1 occurrence of LA1 is - CHRL- or -C(RL)2-; and each remaining occurrence of LA1 is -CH2-. 74. The compound of any one of claims 1-18 or 34-37, wherein L is selected from the group consisting of:

wherein bb represents the point of attachment to Ring C. 75. The compound of any one of claims 1-18 or 34-37, wherein L is selected from the group consisting of:

wherein bb represents the point of attachment to Ring C. 76. The compound of any one of claims 1-18, 34, or 43-44, wherein L is , , or , wherein bb represents the point of attachment to Ring C. 77. The compound of any one of claims 1-18 or 51-53, wherein L is selected from the group consisting of: wherein bb represents the point of attachment to Ring C.

78. The compound of any one of claims 1-18 or 51-53, wherein L is selected from the group consisting of: wherein bb represents the point of attachment to Ring C. 79. The compound of claim 1, wherein the compound of Formula (I) is a compound of Formula (I-A): Formula (I-A) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; LA3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5; LA1a and LA1b are independently selected from the group consisting of: -CH2-, -CHRL- , and -C(RL)2-; and LA4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 Ra; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 Ra; or the compound of Formula (I) is a compound of Formula (I-B): Formula (I-B) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; LA3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5; LA1a and LA1b are independently selected from the group consisting of: -CH2-, -CHRL- , and -C(RL)2-; and LA4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 Ra; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 Ra; or the compound of Formula (I) is a compound of Formula (I-C) or (I-D): Formula (I-D) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3a is 0 or 1; LA3a and LA3b are independently selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 7; LA1a and LA1b are independently selected from the group consisting of: -CH2-, -CHRL- , and -C(RL)2-; and LA4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 Ra; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 Ra; or the compound of Formula (I) is a compound of Formula (I-E): Formula (I-E) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; LA3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; LA1a and LA1b are independently selected from the group consisting of: -CH2-, -CHRL- , and -C(RL)2-; and LA4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 Ra; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 Ra; and m8 is 0, 1, or 2; or the compound of Formula (I) is a compound of Formula (I-Ea):

Formula (I-Ea) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; LA3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; LA1a and LA1b are independently selected from the group consisting of: -CH2-, -CHRL- , and -C(RL)2-; and LA4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 Ra; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 Ra; n2 and n3 are independently 0, 1, or 2; and m8 is 0, 1, or 2; or the compound of Formula (I) is a compound of Formula (I-Eb): Formula (I-Eb) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; LA3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; LA1a and LA1b are independently selected from the group consisting of: -CH2-, -CHRL- , and -C(RL)2-; and LA4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 Ra; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 Ra; n2 and n3 are independently 0, 1, or 2; and m8 is 0, 1, or 2; or the compound of Formula (I) is a compound of Formula (I-F): Formula (I-F) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; LA3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; LA1a and LA1b are independently selected from the group consisting of: -CH2-, -CHRL- , and -C(RL)2-; and LA4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 Ra; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 Ra; and m8 is 0, 1, or 2; or the compound of Formula (I) is a compound of Formula (I-G): Formula (I-G) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; LA3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; LA1a and LA1b are independently selected from the group consisting of: -CH2-, -CHRL- , and -C(RL)2-; and LA4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 Ra; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 Ra; and m8 is 0, 1, or 2; or the compound of Formula (I) is a compound of Formula (I-H):

Formula (I-H) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; LA3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; LA1a and LA1b are independently selected from the group consisting of: -CH2-, -CHRL- , and -C(RL)2-; and LA4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 Ra; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 Ra; and m8 is 0, 1, or 2. 80. A compound selected from the group consisting of the compounds in Table C1, or a pharmaceutically acceptable salt thereof. 81. The compound of claim 80, wherein the compound does not exhibit Ymin (6 h) ≥ 70% and/or Ymin (24 h) ≥ 70% under conditions described in Example B1. 82. A pharmaceutical composition comprising a compound as claimed in any one of claims 1-81, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.

83. A method for treating a cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of any one of claims 1-81, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 82. 84. The method of claim 83, wherein the method comprises administering an additional therapy or therapeutic agent. 85. The method of claim 84, wherein the additional therapy or therapeutic agent is an ALK inhibitor, a BCL-2 inhibitor, a BCR-Abl inhibitor, a BRaf inhibitor, a CDK2 inhibitor, CDK 4/6 inhibitor, a CDK7 inhibitor, a CDK9 inhibitor, an EGFR inhibitor, an anti-EGFR antibody or an anti-EGFR antibody-drug conjugate, an ERK inhibitor, a FGFR1 inhibitor, a FGFR2 inhibitor, a FGFR3 inhibitor, a FGFR4 inhibitor, a HER2 inhibitor, an anti-HER2 antibody or an anti-HER2 antibody-drug conjugate, a JAK inhibitor, a KRas inhibitor, a MEK inhibitor, a MET inhibitor, a PARP inhibitor, an LSD1 inhibitor, a BET inhibitor, a telomerase inhibitor, a TORC1/2 inhibitor, chemotherapy, radiotherapy, or a combination thereof. 86. The method of claim 85, wherein the additional therapy or therapeutic agent is a JAK inhibitor. 87. The method of claim 86, wherein the JAK inhibitor is fedratinib (e.g., fedratinib dihydrochloride monohydrate), momelotinib (e.g., momelotinib dihydrochloride), pacritinib (e.g., pacritinib citrate), ruxolitinib (e.g., ruxolitinib phosphate), or a combination thereof. 88. The method of any one of claims 85-87, wherein the cancer is breast cancer, colorectal cancer, bile duct cancer, colorectal cancer, gastrointestinal stromal tumor, pancreatic cancer, bladder cancer, kidney cancer, cervical cancer, ovarian cancer, uterine cancer, head and neck cancer, hematological cancer, lung cancer, skin cancer, or a combination thereof. 89. The method of claim 88, wherein the hematological cancer is acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), follicular lymphoma (FL), small lymphocytic lymphoma (SLL), essential thrombocythemia, polycythemia vera, myelofibrosis, or a combination thereof. 90. The method of claim 89, wherein the hematological cancer is essential thrombocythemia, polycythemia vera, myelofibrosis, or a combination thereof. 91. The method of any one of claims 89-90, wherein the hematological cancer has a JAK2 mutation (e.g., JAK2 V617F). 92. A compound of Formula (SI-A): Formula (SI-A) or salts thereof, wherein: each R2 and R4 is independently selected from: halo, CN, C1-3 alkyl, C1-3 haloalkyl, C1- 3 alkoxy, C1-3 haloalkoxy, OH, and NRdRe; m2 is 0, 1, or 2; m4 is 0, 1, 2, or 3; m6 is 0 or 1; Re1 is H or C1-6 alkyl optionally substituted with 1-3 -F; Re2 is H or C1-6 alkyl optionally substituted with 1-3 -F; a3 is 0 or 1; LA3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; LA1a and LA1b are independently selected from the group consisting of: -CH2-, -CHRL- , and -C(RL)2-; LA4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 Ra; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 Ra; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5; each RL is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C1-6 haloalkoxy; -NRdRe; C(=O)N(Rf)2; S(O)0-2(C1-6 alkyl); S(O)0-2(C1-6 haloalkyl); S(O)1-2N(Rf)2; -Rb; and C1-6 alkyl optionally substituted with 1-6 Rc; each Ra is independently selected from the group consisting of: each Rb is independently selected from the group consisting of: -(Lb)b-Rb1 and -Rb1, wherein: each b is independently 1, 2, or 3; each -Lb is independently selected from the group consisting of: -O-; -N(H)-; -N(C1-3 alkyl)-; -S(O)0-2-; C(=O); and C1-3 alkylene; and each Rb1 is independently selected from the group consisting of: C3-10 cycloalkyl, 4-10 membered heterocyclyl, C6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 Rg; each Rc is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C1-6 haloalkoxy; -NRdRe; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC1-6 haloalkyl; C(=O)OH; C(=O)N(Rf)2; S(O)0-2(C1-6 alkyl); S(O)0-2(C1-6 haloalkyl); and S(O)1-2N(Rf)2; each Rd and Re is independently selected from the group consisting of: H; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC1-6 haloalkyl; C(=O)N(Rf)2; S(O)1- 2(C1-6 alkyl); S(O)1-2(C1-6 haloalkyl); S(O)1-2N(Rf)2; and C1-6 alkyl optionally substituted with 1-3 Rh; each Rf is independently selected from the group consisting of: H and C1-6 alkyl optionally substituted with 1-3 Rh; and each Rh is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C1-6 haloalkoxy; -NH2, -N(H)(C1-3 alkyl); and -N(C1-3 alkyl)2. 93. A compound of Formula (I-E): Formula (SI-E) or salts thereof, wherein: each R2 and R4 is independently selected from: H, C1-3 alkyl, and halo; a3 is 0 or 1; m2 is 0, 1, or 2; m4 is 0, 1, 2, or 3; m6 is 0 or 1; Re1 is H or C1-6 alkyl optionally substituted with 1-3 -F; LA3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; LA1a and LA1b are independently selected from the group consisting of: -CH2-, -CHRL- , and -C(RL)2-; LA4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 Ra; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 Ra; a1a is an integer from 0 to 5; a1d is an integer from 0 to 4, provided that a1a + a1d is from 1 to 4; each RL is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C1-6 haloalkoxy; -NRdRe; C(=O)N(Rf)2; S(O)0-2(C1-6 alkyl); S(O)0-2(C1-6 haloalkyl); S(O)1-2N(Rf)2; -Rb; and C1-6 alkyl optionally substituted with 1-6 Rc; each Ra is independently selected from the group consisting of: each Rb is independently selected from the group consisting of: -(Lb)b-Rb1 and -Rb1, wherein: each b is independently 1, 2, or 3; each -Lb is independently selected from the group consisting of: -O-; -N(H)-; -N(C1-3 alkyl)-; -S(O)0-2-; C(=O); and C1-3 alkylene; each Rb1 is independently selected from the group consisting of: C3-10 cycloalkyl, 4-10 membered heterocyclyl, C6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 Rg; each Rc is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C1-6 haloalkoxy; -NRdRe; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC1-6 haloalkyl; C(=O)OH; C(=O)N(Rf)2; S(O)0-2(C1-6 alkyl); S(O)0-2(C1-6 haloalkyl); and S(O)1-2N(Rf)2; each Rd and Re is independently selected from the group consisting of: H; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC1-6 haloalkyl; C(=O)N(Rf)2; S(O)1- 2(C1-6 alkyl); S(O)1-2(C1-6 haloalkyl); S(O)1-2N(Rf)2; and C1-6 alkyl optionally substituted with 1-3 Rh; each Rf is independently selected from the group consisting of: H and C1-6 alkyl optionally substituted with 1-3 Rh; each Rg is independently selected from the group consisting of: Rh; C1-3 alkyl; and C1- 3 haloalkyl; and each Rh is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C1-6 haloalkoxy; -NH2, -N(H)(C1-3 alkyl); and -N(C1-3 alkyl)2. 94. A compound of any one of Formulas (I-Ind-1), (I-Ind-2), or (I-Ind-3): or salts thereof, wherein: each RY is independently selected from the group consisting of: Ra and Rb; c1 is 0, 1, 2, or 3; n2 and n3 are independently 0, 1, or 2; RaN is H or C1-6 alkyl optionally substituted with 1-3 Rc; m8 is 0 to 4; each Ra is independently selected from the group consisting of: each Rb is independently selected from the group consisting of: -(Lb)b-Rb1 and -Rb1, wherein: each b is independently 1, 2, or 3; each -Lb is independently selected from the group consisting of: -O-; -N(H)-; -N(C1-3 alkyl)-; -S(O)0-2-; C(=O); and C1-3 alkylene; each Rb1 is independently selected from the group consisting of: C3-10 cycloalkyl, 4-10 membered heterocyclyl, C6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 Rg; each Rc is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C1-6 haloalkoxy; -NRdRe; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC1-6 haloalkyl; C(=O)OH; C(=O)N(Rf)2; S(O)0-2(C1-6 alkyl); S(O)0-2(C1-6 haloalkyl); and S(O)1-2N(Rf)2; each Rd and Re is independently selected from the group consisting of: H; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC1-6 haloalkyl; C(=O)N(Rf)2; S(O)1- 2(C1-6 alkyl); S(O)1-2(C1-6 haloalkyl); S(O)1-2N(Rf)2; and C1-6 alkyl optionally substituted with 1-3 Rh; each Rf is independently selected from the group consisting of: H and C1-6 alkyl optionally substituted with 1-3 Rh; each Rg is independently selected from the group consisting of: Rh; C1-3 alkyl; and C1- 3 haloalkyl; and each Rh is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C1-6 haloalkoxy; -NH2, -N(H)(C1-3 alkyl); and -N(C1-3 alkyl)2. 95. Any of the compounds, compositions, combinations, pharmaceutical compositions, methods, uses, or processes as provided herein.

Description:
Tetrahydroisoquinoline Heterobifunctional BCL-XL Degraders CROSS-REFERENCE TO RELATED APPLICATIONS This application claims priority to U.S. Provisional Application Serial Nos.63/457,997, filed April 7, 2023; 63/454,545, filed March 24, 2023; 63/449,756, filed March 3, 2023; 63/429,862, filed December 2, 2022; 63/398,783, filed August 17, 2022; and 63/339,253, filed May 6, 2022; each of which is incorporated by reference in its entirety herein. TECHNICAL FIELD This disclosure provides compounds of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or pharmaceutically acceptable salts thereof, that induce degradation of a BCL-XL protein. These compounds are useful, for example, for treating cancer in a subject (e.g., a human). This disclosure also provides compositions containing the compounds provided herein as well as methods of using and making the same. BACKGROUND The BCL-2 family of proteins is involved in the regulation of cell apoptosis and includes proteins that are pro-apoptosis, pro-survival, and BH3-only. At a high level, the balance of binding of BH3-only proteins to the pro-apoptosis and pro-survival members of the BCL-2 family can determine whether a cell will undergo apoptosis. The protein BCL-XL, encoded by the BCL2L1 gene, is a pro-survival member of the BCL-2 family. In many cancers, it can be desirable to initiate apoptosis of tumor cells, which may be achieved by decreasing the amount of pro-survival protein (e.g., BCL-XL) available to compete for BH3-only protein binding. SUMMARY This disclosure provides compounds of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or pharmaceutically acceptable salts thereof, that induce degradation of a BCL-XL protein. These compounds are useful, for example, for treating a cancer in a subject (e.g., a human). This disclosure also provides compositions containing the compounds provided herein as well as methods of using and making the same. Provided herein are compounds of Formula (I) or (II): Formula (II) or pharmaceutically acceptable salts thereof, wherein: Ring A is selected from the group consisting of: (a) C 3-15 cycloalkylene or 3-15 membered heterocyclylene, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; and (b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 substituents independently selected from the group consisting of: R a and R b ; L T1 is a bond or C1-3 alkylene optionally substituted with 1-3 substituents independently selected from the group consisting of: oxo and R c , wherein one CH2 unit of the C1-3 alkylene is optionally replaced with -O- or -N(R d )-; A* is selected from the group consisting of: (a) C3-15 cycloalkyl or 3-15 membered heterocyclyl, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; (b) C6-15 aryl or 5-15 membered heteroaryl, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; and (c) H; R 1 is selected from the group consisting of: (a) C(O)OH, (b) C(O)NR d R e , and (c) C(O)OC1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with 1-3 R c ; each R 2 , R 3 , R 4 , and R 5 is independently selected from the group consisting of: halo, CN, C1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, C1-3 haloalkoxy, OH, and NR d R e ; m2 is 0, 1, or 2; m3 and m4 are independently 0, 1, 2, or 3; m5 is 0, 1, 2, 3, or 4; L is –(L A )n1–, wherein L A and n1 are defined according to (AA) or (BB): n1 is an integer from 3 to 15; and each L A is independently selected from the group consisting of: L A1 , L A3 , and L A4 , provided that 1-3 occurrences of L A is L A4 ; (BB) n1 is an integer from 0 to 20; and each L A is independently selected from the group consisting of: L A1 and L A3 ; each L A1 is independently selected from the group consisting of: -CH2-, -CHR L -, and - C(R L )2-; each L A3 is independently selected from the group consisting of: -N(R d )-, -N(R b )-, -O- , -S(O)0-2-, and C(=O); each L A4 is independently selected from the group consisting of: (a) C3-15 cycloalkylene or 3-15 membered heterocyclylene, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; and (b) C 6-15 arylene or 5-15 membered heteroarylene, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; provided that L does not contain any N-O, O-O, N-N, N-S(O)0, or O-S(O)0-2 bonds; wherein each R L is independently selected from the group consisting of: halo; cyano; - OH; -C 1-6 alkoxy; -C 1-6 haloalkoxy; -NR d R e ; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); S(O)1-2N(R f )2; -R b ; and C1-6 alkyl optionally substituted with 1-6 R c ; Ring C is selected from the group consisting of: ; c1 is 0, 1, 2, or 3; each R Y is independently selected from the group consisting of: R a and R b ; R aN is H or C1-6 alkyl optionally substituted with 1-3 R c ; Y 1 and Y 2 are independently N, CH, or CR Y ; yy represents the point of attachment to L; X is CH, C, or N; the single bond or a double bond; L C is selected from the group consisting of: a bond, -CH 2 -, -CHR a -, -C(R a ) 2 -, -N(R d )-, and O, provided that when X is N, then L C is other than O; each R a is independently selected from the group consisting of: (a) halo; (b) cyano; (c) -OH; (d) oxo; (e) -C1-6 alkoxy; (f) -C 1-6 haloalkoxy; (g) -NR d R e ; (h) C(=O)C1-6 alkyl; (i) C(=O)C1-6 haloalkyl; (j) C(=O)OH; (k) C(=O)OC1-6 alkyl; (l) C(=O)OC1-6 haloalkyl; (m) C(=O)N(R f ) 2 ; (n) S(O) 0-2 (C 1-6 alkyl); (o) S(O)0-2(C1-6 haloalkyl); (p) S(O) 1-2 N(R f ) 2 ; and (q) C 1-6 alkyl, C 2-6 alkenyl, or C 2-6 alkynyl, each optionally substituted with 1-6 R c ; each R b is independently selected from the group consisting of: -(L b )b-R b1 and -R b1 , wherein: each b is independently 1, 2, or 3; each -L b is independently selected from the group consisting of: -O-; -N(H)-; -N(C1-3 alkyl)-; -S(O) 0-2 -; C(=O); and C 1-3 alkylene; and each R b1 is independently selected from the group consisting of: C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 R g ; each R c is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C1-6 haloalkoxy; -NR d R e ; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)OH; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); and S(O)1-2N(R f )2; each R d and R e is independently selected from the group consisting of: H; C(=O)C1-6 alkyl; C(=O)C 1-6 haloalkyl; C(=O)OC 1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)N(R f ) 2 ; S(O) 1- 2(C1-6 alkyl); S(O)1-2(C1-6 haloalkyl); S(O)1-2N(R f )2; and C1-6 alkyl optionally substituted with 1-3 R h ; each R f is independently selected from the group consisting of: H and C 1-6 alkyl optionally substituted with 1-3 R h ; each R g is independently selected from the group consisting of: R h ; C1-3 alkyl; and C1- 3 haloalkyl; and each R h is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C1-6 haloalkoxy; -NH2, -N(H)(C1-3 alkyl); and -N(C1-3 alkyl)2. Also provided herein is a pharmaceutical composition comprising a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier. Provided herein is a method for treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein. Also provided herein is a BCL-X L protein non-covalently bound with a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof. Also provided herein is a ternary complex comprising a BCL-XL protein, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I- F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, and a CRBN protein, or a portion thereof. To facilitate understanding of the disclosure set forth herein, a number of additional terms are provided. Generally, the nomenclature used herein and the laboratory procedures in organic chemistry, medicinal chemistry, and pharmacology described are those well-known and commonly employed in the art. Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. Each of the patents, applications, published applications, and other publications that are mentioned throughout the specification and the attached appendices are incorporated herein by reference in their entireties. The details of one or more embodiments of the invention are set forth in the accompanying drawings and the description below. Other features and advantages of the invention will be apparent from the description and drawings, and from the claims. DETAILED DESCRIPTION This disclosure provides compounds of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or pharmaceutically acceptable salts thereof, that induce degradation of a BCL-XL (also referred to as Bcl-xL herein) protein. These compounds are useful, for example, for treating a cancer. This disclosure also provides compositions containing the compounds provided herein as well as methods of using and making the same. Without being bound by any particular theory, it is believed that in healthy cells, pro- apoptotic effectors such as BAX and BAK can move between the cytosol and the mitochondrial outer membrane (MOM), on which Voltage Dependent Anion Channel 2 (VDAC2) can act as a receptor. Pro-survival BCL-2 family members (e.g., BCL-2, BCL-XL, and MCL-1) can retrotranslocate BAX back to the cytosol. A BH3-only protein (e.g., BIM) can engage a rear site on a pro-apoptotic effector (e.g., BAX or BAK) and release the C-terminal transmembrane domain (α9) of the effector, enabling binding to the MOM. Binding of BIM to the canonical BH3 binding groove of BAX or BAK releases the N-terminus and α1 of BAX or BAK, and subsequent unfolding of the “latch” domain releases BIM from the BH3 binding groove. If a pro-survival BCL-2 family member then binds to BAX or BAK, apoptotic signaling is generally halted. However, if BAX or BAK are allowed to dimerize, then oligomerize, the MOM can be permeabilized, leading to apoptosis. An abundance of pro-survival BCL-2 family members is sometimes thought to “prime” cells for death (e.g., via cytotoxic therapies, including BH3 mimetics). It is believed that oncogenic mutations and stresses can cause an upregulation in BH3-only proteins, thus placing a selective pressure on cancer cells for upregulation of the pro-survival BCL-2 family proteins. Thus, with more BH3-only proteins around, the cells are believed to be more sensitive to further manipulations of the BCL-2 family balance. See, e.g., Adams and Cory, Cell Death & Differentiation 25.1 (2018): 27-36. Compounds that induce degradation of a target protein are sometimes referred to as heterobifunctional compounds, PROTACs, or degraders. Such compounds generally include a moiety that binds to the target protein and a moiety that binds to a ubiquitin E3 ligase (sometimes referred to as an E3 ligase or simply an E3), these two moieties being optionally separated by a linker. To induce degradation, heterobifunctional compounds are believed to induce formation of a ternary complex between the target protein, the compound, and an E3 ligase. Formation of the ternary complex is then followed by ubiquitination of the target protein and degradation of the ubiquitinated target protein by a proteosome. Several E3 ligases have been used as the partner E3 ligase for heterobifunctional degraders. Herein, the cereblon (CRBN) E3 ligase (also referred to herein as a CRBN protein) is used. A degradation approach for a target protein can have potential advantages compared to, for example, small molecule inhibition of the target protein. One potential advantage is that the duration of effect of a heterobifunctional compound is generally based on the resynthesis rate of the target protein. Another potential advantage is that many heterobifunctional compounds are believed to be released from the ubiquitinated target protein-E3 ligase complex and made available for formation of further ternary complexes; this is sometimes referred to as “catalytic” turnover of the heterobifunctional compound. Degradation of a target protein can also be advantageous over small molecule inhibition in some cases, as degradation can impair a scaffolding function of a target protein, whereas a small molecule might not. It is also generally believed that for formation of a ternary complex, high affinity to the target protein is not always required. Heterobifunctional compounds are further described in, for example, International Publication Nos. WO 2017/184995; WO 2019/144117; WO 2020/163823; WO 2021/078301; WO 2021/146536; WO 2021/007307; WO 2021/222114; WO 2022/169780; WO 2023/044046; Chamberlain and Hamann, Nature Chemical Biology 15.10 (2019): 937-944; Li and Song, Journal of Hematology & Oncology 13 (2020): 1-14; Wu, et al. Nature Structural & Molecular Biology 27.7 (2020): 605-614; Dong, et al., Journal of Medicinal Chemistry 64.15 (2021): 10606-10620; Yang, et al., Targeted Oncology 16.1 (2021): 1-12; Lv, et al., Nature Communications 12.1 (2021): 6896. Compound Embodiments Provided herein are compounds of Formula (I) or (II): Formula (II) or pharmaceutically acceptable salts thereof, wherein: Ring A is selected from the group consisting of: (a) C 3-15 cycloalkylene or 3-15 membered heterocyclylene, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; and (b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 substituents independently selected from the group consisting of: R a and R b ; L T1 is a bond or C1-3 alkylene optionally substituted with 1-3 substituents independently selected from the group consisting of: oxo and R c , wherein one CH2 unit of the C1-3 alkylene is optionally replaced with -O- or -N(R d )-; A* is selected from the group consisting of: (a) C3-15 cycloalkyl or 3-15 membered heterocyclyl, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; (b) C6-15 aryl or 5-15 membered heteroaryl, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; and (c) H; R 1 is selected from the group consisting of: (a) C(O)OH, (b) C(O)NR d R e , and (c) C(O)OC1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with 1-3 R c ; each R 2 , R 3 , R 4 , and R 5 is independently selected from the group consisting of: halo, CN, C1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, C1-3 haloalkoxy, OH, and NR d R e ; m2 is 0, 1, or 2; m3 and m4 are independently 0, 1, 2, or 3; m5 is 0, 1, 2, 3, or 4; L is –(L A ) n1 –, wherein L A and n1 are defined according to (AA) or (BB): (AA) n1 is an integer from 3 to 15; and each L A is independently selected from the group consisting of: L A1 , L A3 , and L A4 , provided that 1-3 occurrences of L A is L A4 ; n1 is an integer from 0 to 20; and each L A is independently selected from the group consisting of: L A1 and L A3 ; each L A1 is independently selected from the group consisting of: -CH2-, -CHR L -, and - C(R L )2-; each L A3 is independently selected from the group consisting of: -N(R d )-, -N(R b )-, -O- , -S(O) 0-2 -, and C(=O); each L A4 is independently selected from the group consisting of: (a) C 3-15 cycloalkylene or 3-15 membered heterocyclylene, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; and (b) C 6-15 arylene or 5-15 membered heteroarylene, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; provided that L does not contain any N-O, O-O, N-N, N-S(O) 0 , or O-S(O) 0-2 bonds; wherein each R L is independently selected from the group consisting of: halo; cyano; - OH; -C 1-6 alkoxy; -C 1-6 haloalkoxy; -NR d R e ; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); S(O)1-2N(R f )2; -R b ; and C1-6 alkyl optionally substituted with 1-6 R c ; Ring C is selected from the group consisting of: ; c1 is 0, 1, 2, or 3; each R Y is independently selected from the group consisting of: R a and R b ; R aN is H or C1-6 alkyl optionally substituted with 1-3 R c ; Y 1 and Y 2 are independently N, CH, or CR Y ; yy represents the point of attachment to L; X is CH, C, or N; the is a single bond or a double bond; L C is selected from the group consisting of: a bond, -CH 2 -, -CHR a -, -C(R a ) 2 -, -N(R d )-, and O, provided that when X is N, then L C is other than O; each R a is independently selected from the group consisting of: (a) halo; (b) cyano; (c) -OH; (d) oxo; (e) -C1-6 alkoxy; (f) -C1-6 haloalkoxy; (g) -NR d R e ; (h) C(=O)C1-6 alkyl; (i) C(=O)C1-6 haloalkyl; (j) C(=O)OH; (k) C(=O)OC1-6 alkyl; (l) C(=O)OC1-6 haloalkyl; (m) C(=O)N(R f ) 2 ; (n) S(O) 0-2 (C 1-6 alkyl); (o) S(O)0-2(C1-6 haloalkyl); (p) S(O) 1-2 N(R f ) 2 ; and (q) C 1-6 alkyl, C 2-6 alkenyl, or C 2-6 alkynyl, each optionally substituted with 1-6 R c ; each R b is independently selected from the group consisting of: -(L b )b-R b1 and -R b1 , wherein: each b is independently 1, 2, or 3; each -L b is independently selected from the group consisting of: -O-; -N(H)-; -N(C1-3 alkyl)-; -S(O) 0-2 -; C(=O); and C 1-3 alkylene; and each R b1 is independently selected from the group consisting of: C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 R g ; each R c is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C1-6 haloalkoxy; -NR d R e ; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)OH; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); and S(O)1-2N(R f )2; each R d and R e is independently selected from the group consisting of: H; C(=O)C1-6 alkyl; C(=O)C 1-6 haloalkyl; C(=O)OC 1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)N(R f ) 2 ; S(O) 1- 2 (C 1-6 alkyl); S(O) 1-2 (C 1-6 haloalkyl); S(O) 1-2 N(R f ) 2 ; and C 1-6 alkyl optionally substituted with 1-3 R h ; each R f is independently selected from the group consisting of: H and C1-6 alkyl optionally substituted with 1-3 R h ; each R g is independently selected from the group consisting of: R h ; C1-3 alkyl; and C1- 3 haloalkyl; and each R h is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C 1-6 haloalkoxy; -NH 2 , -N(H)(C 1-3 alkyl); and -N(C 1-3 alkyl) 2 . In some embodiments, the compounds are compounds of Formula (I), or pharmaceutically acceptable salts thereof. In some embodiments, the compounds are compounds of Formula (II), or pharmaceutically acceptable salts thereof. In some embodiments, Ring A is phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 substituents independently selected from the group consisting of: R a and R b . In some embodiments, Ring A is phenylene optionally substituted with 1-3 substituents independently selected from the group consisting of: R a and R b . In some embodiments, Ring A is a phenylene optionally substituted with 1-3 R a . In some embodiments, Ring A is phenylene (e.g., 1,3-phenylene or 1,4-phenylene). In some embodiments, Ring , wherein aa represents the point of attachment to L or L T1 . wherein aa represents the point of attachment to L or L T1 . In some embodiments, Ring A is , wherein aa represents the point of attachment to L or L T1 . aa represents the point of attachment to L or L T1 . In some embodiments, Ring A is 5-6 membered heteroarylene optionally substituted with 1-3 R a . In some embodiments, Ring A is 5-6 membered heteroarylene optionally substituted with 1-2 R a . In some embodiments, Ring A is 5-membered heteroarylene optionally substituted with 1-2 R a . In some embodiments, Ring A is pyrazolylene optionally substituted with 1-2 R a . For example, Ring A can be selected from the group consisting of: , wherein aa represents the point of attachment to L or L T1 . In some embodiments, Ring A is C3-10 cycloalkylene optionally substituted with 1-6 R a . In some embodiments, Ring A is C4-6 cycloalkylene optionally substituted with 1-3 R a . In some embodiments, Ring A is cyclohexylene optionally substituted with 1-3 R a . For example, Ring A can be 1,4-cyclohexylene. In some embodiments, one R a present on Ring A is C1-3 alkyl optionally substituted with 1-3 F. In some embodiments, one R a present on Ring A is methyl or CF 3 . In some embodiments, R 1 is C(O)OH. In some embodiments, R 1 is C(O)OC1-6 alkyl. In some embodiments, R 1 is C(O)NHR e . In some embodiments, R 1 is C(O)OH; and Ring , wherein aa represents the point of attachment to L or L T1 . In some embodiments, Ring , wherein aa represents the point of attachment to L or L T1 . For example, Ring A can be wherein aa represents the point of attachment to L or L T1 . In some embodiments, Ring , wherein aa represents the point of attachment to L or L T1 . For example, Ring A can wherein aa represents the point of attachment to L or L T1 . In some embodiments, R 1 is C(O)OH; and Ring A is , wherein aa represents the point of attachment to L or L T1 . In some embodiments, R 1 is C(O)OH or C(O)OC 1-6 alkyl; and Ring A is selected from the group consisting of: , wherein R a1 is C1-3 alkyl optionally substituted with 1-3 F; and aa represents the point of attachment to L or L T1 . In some embodiments, L T1 is C1-3 alkylene. For example, L T1 can be -CH2-. In some embodiments, A* is H. In some embodiments, A* is C 3-15 cycloalkyl or 3-15 membered heterocyclyl, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b . In some embodiments, A* is C 3-15 cycloalkyl optionally substituted with 1-3 R a . For example, A* can be adamantyl optionally substituted with 1-3 R a . In some embodiments, m2 is 0. In some embodiments, m3 is 0. In some embodiments, m4 is 0. In some embodiments, m5 is 0. In some embodiments, m2 is 0; m3 is 0; m4 is 0; and m5 is 0. In some embodiments, m2 is 0; m3 is 0; m4 is 0; and m5 is 0; R 1 is C(O)OH or C(O)OC 1-6 alkyl; and Ring A is selected from the group consisting of: , wherein R a1 is C 1-3 alkyl optionally substituted with 1-3 F; and aa represents the point of attachment to L or L T1 . In some embodiments, m2 is 0; m3 is 0; m4 is 0; and m5 is 0; R 1 is C(O)OH or C(O)OC 1-6 alkyl; and attachment to L or L T1 . In some embodiments, Ring . some embodiments, Ring C is In some embodiments, Ring , wherein c1 is 0 or 1; and R a2 is selected from the group consisting of: halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 -F. In some embodiments, Ring C is , wherein R aN is C1-3 alkyl (e.g., methyl); c1 is 0 or 1; and R a2 is selected from the group consisting of: halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 -F. In some , In some embodiments, Ring C is . In some embodiments, Ring C is In some embodiments, c1 is 0. In some embodiments, c1 is 1. In some embodiments, c1 is 1; and R Y is R a . In some embodiments, R Y is -F, C1-3 alkyl, or C1-3 alkyl substituted with 1-3 -F. In some embodiments, R aN is C 1-3 alkyl. For example, R aN can be methyl. In some embodiments, L C is a bond or -N(R d )-. In some embodiments, L C is a bond or –N(H)-. For example, L C can be a bond. For example, L C can be -NH-. In some embodiments, X is CH. In some embodiments, L is –(L A )n1-; and L A and n1 are defined according to (AA). In some embodiments, n1 is an integer from 3 to 5. In some embodiments, n1 is an integer from 5 to 9. In some embodiments, n1 is 6, 7, or 8. In some embodiments, n1 is an integer from 9 to 12. In some embodiments, L A and n1 are defined according to (AA); and 1-2 occurrences of L A is L A4 . In some embodiments, one occurrence of L A is L A4 . In some embodiments, two occurrences of L A are L A4 . In some embodiments, each L A4 is independently selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . In some embodiments, L A and n1 are defined according to (AA); and 1-4 occurrences of L A is L A3 . In some embodiments, 1-3 (e.g., 1-2 or 2-3) occurrences of L A is L A3 . In some embodiments, 0-1 occurrence of L A3 is C(=O); and each remaining occurrence of L A3 is independently selected from the group consisting of: -O-; -N(H)-; and -N(C1-3 alkyl)-. In some embodiments, L A and n1 are defined according to (AA); and 2-7 occurrences of L A are L A1 . In some embodiments, 2-5 occurrences of L A are L A1 . In some embodiments, 0-2 (e.g., 0-1) occurrences of L A1 are -CHR L - or -C(R L )2-; and each remaining occurrence of L A1 is -CH2-. In some embodiments, each occurrence of L A1 is -CH2-. In some embodiments, one occurrence of L A1 is -CHR L - or -C(R L ) 2 -; and each remaining occurrence of L A1 is -CH 2 - . In some embodiments, each R L is independently selected from the group consisting of: -F and -C1-3 alkyl optionally substituted with 1-3 F (e.g., CH3 or CF3). In some embodiments, L is –(L A )n1-, and L A and n1 are defined according to (AA), wherein n1 is an integer from 5 to 9; 1-2 occurrences of L A is L A4 ; 2-7 occurrences of L A are L A1 ; and 1-3 occurrences of L A is L A3 . In some embodiments, each L A4 is independently selected from the group consisting of: C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . In some embodiments, 0-1 occurrence of L A1 is -CHR L - or -C(R L ) 2 -; and each remaining occurrence of L A1 is -CH 2 -. In some embodiments, 0-1 occurrence of L A3 is C(=O); and each remaining occurrence of L A3 is independently selected from the group consisting of: -O-; -N(H)-; and -N(C1-3 alkyl)-. In some embodiments, L is selected from the group consisting of: (i) –(L A3 ) 0-2 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 -(L A3 ) 1-2 - bb ; (ii) –(L A3 )0-2-(L A1 )0-5-L A4 -(L A1 )0-5-L A4 -bb; (iii) –(L A3 ) 0-2 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 -L A3 -L A4 - bb ; and (iv) –(L A3 ) 0-1 -(L A1 ) 2-3 -L A3 -L A4 -(L A1 ) 0-5 -(L A3 ) 1-2 - bb ; provided that L contains 2-7 L A1 ; and wherein bb represents the point of attachment to Ring C. In some embodiments, each L A4 is independently selected from the group consisting of: C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . In some embodiments, 0-1 occurrence of L A1 is -CHR L - or -C(R L )2-; and each remaining occurrence of L A1 is -CH 2 -. In some embodiments, 0-1 occurrence of L A3 is C(=O); and each remaining occurrence of L A3 is independently selected from the group consisting of: -O-; -N(H)-; and -N(C1-3 alkyl)-. In some embodiments, L is –(L A3 ) 0-2 -(L A1 ) 2-9 -(L A3 ) 0-1 -L A4 - bb , wherein bb represents the point of attachment to Ring C. In some embodiments, L A4 is selected from the group consisting of: C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . In some embodiments, L A4 is 4-10 membered heterocyclylene (e.g., 5-6 membered heterocyclylene (e.g., 6-membered heterocyclylene (e.g., piperazinylene))) optionally substituted with 1-3 R a . In some embodiments, 0-1 occurrence of L A1 is -CHR L - or -C(R L )2-; and each remaining occurrence of L A1 is -CH2-. In some embodiments, 0-1 occurrence of L A3 is C(=O); and each remaining occurrence of L A3 is independently selected from the group consisting of: -O-; -N(H)-; and -N(C 1-3 alkyl)-. In some embodiments, L is: –(L A3 )0-2-(L A1 )0-5-L A4 -(L A1 )0-5-(L A3 )1-2- bb; provided that L contains 1-7 (e.g., 2-7) L A1 ; and wherein bb represents the point of attachment to Ring C. In some embodiments, L is: –(L A3 ) 0-1 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 -(L A3 ) 2 - bb ; provided that L contains 1-5 (e.g., 2-5) L A1 ; and wherein bb represents the point of attachment to Ring C. In some embodiments, L A4 is selected from the group consisting of: C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . In some embodiments, 0-1 occurrence of L A1 is -CHR L - or -C(R L )2-; and each remaining occurrence of L A1 is -CH 2 -. In some embodiments, 0-1 occurrence of L A3 is C(=O); and each remaining occurrence of L A3 is independently selected from the group consisting of: -O-; -N(H)-; and - N(C1-3 alkyl)-. In some embodiments, L is: –(L A3 ) 0-1 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 -C(=O)-N(R d )- bb ; provided that L contains 1-5 (e.g., 2-5) L A1 ; and wherein bb represents the point of attachment to Ring C. In some embodiments, L A4 is selected from the group consisting of: C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . For example, L A4 can be a 4-10 membered (e.g., nitrogen containing) heterocyclylene optionally substituted with 1-3 R a . In some embodiments, 0-1 occurrence of L A1 is -CHR L - or -C(R L ) 2 -; and each remaining occurrence of L A1 is -CH 2 -. For example, L can be selected from the group consisting of:

, wherein bb represents the point of attachment to Ring C. For example, L can be selected , , wherein bb represents the point of attachment to Ring C. consisting of: , wherein bb represents the point of attachment to Ring C. In some embodiments, L is a divalent group of Formula (L-1): Formula (L-1) wherein: a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5 (e.g., from 2 to 5); L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and bb represents the point of attachment to Ring C. In some embodiments of (L-1), a3 is 1. In some embodiments, L A3 is -O-. In some embodiments of (L-1), a3 is 1; and L A3 is -O-. In some embodiments of (L-1), a3 is 0. In some embodiments of (L-1), a1a + a1b is 3 or 4. For example, a1a + a1b can be 3. For example, a1a + a1b can be 4. In some embodiments of (L-1), a1a + a1b is 2 or 5. For example, a1a + a1b can be 5. For example, a1a + a1b can be 2. In some embodiments of (L-1), a1a + a1b is 1. In some embodiments of (L-1), each occurrence of L A1a and L A1b is -CH2-. In some embodiments of (L-1), one occurrence of L A1a is -CHR L - or -C(R L ) 2 -; each remaining occurrence of L A1a is -CH 2 -; and each occurrence of L A1b is -CH 2 -. In some embodiments of (L-1), one occurrence of L A1b is -CHR L - or -C(R L )2-; each remaining occurrence of L A1b is - CH2-; and each occurrence of L A1a is -CH2-. In some embodiments, L is: –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-L A3 -bb; provided that L contains 2-7 L A1 ; and wherein bb represents the point of attachment to Ring C. In some embodiments, L is: –(L A3 ) 0-1 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 -O- bb , –(L A3 ) 0-1 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 -N(H)- bb , or –(L A3 ) 0-1 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 -N(C 1-3 alkyl)- bb ; provided that L contains 2-7 L A1 ; and wherein bb represents the point of attachment to Ring C. In some embodiments, L A4 is selected from the group consisting of: C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . In some embodiments, 0-1 occurrence of L A1 is -CHR L - or -C(R L )2-; and each remaining occurrence of L A1 is -CH2-. For example, L In some embodiments, L is: –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-bb; provided that L contains 2-7 L A1 ; and wherein bb represents the point of attachment to Ring C. In some embodiments, L A4 is selected from the group consisting of: (a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and (b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . In some embodiments, 0-1 occurrence of L A1 is -CHR L - or -C(R L ) 2 -; and each remaining occurrence of the point of attachment to Ring C. In some embodiments, L is a divalent group of Formula (L-2) or (L-2a): Formula (L-2a) wherein: a3a is 0 or 1; L A3a and L A3b are independently selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 7; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and bb represents the point of attachment to Ring C. In some embodiments of (L-2) or (L-2a), a3a is 1. In some embodiments, L A3a is -O- . In some embodiments of (L-2) or (L-2a), a3a is 1 and L A3a is -O-. In some embodiments of (L-2), L A3b is -N(H)- or –N(C1-3 alkyl)-. In some embodiments of (L-2), L A3b is -O-. In some embodiments of (L-2) or (L-2a), a1a + a1b is 2, 3, or 4. For example, a1a + a1b can be 3. For example, a1a + a1b can be 4. In some embodiments, a1a is 3. In some embodiments of (L-2) or (L-2a), a1a + a1b is 5 or 6. In some embodiments of (L-2) or (L-2a), each occurrence of L A1a and L A1b is -CH2-. In some embodiments of (L-2) or (L-2a), one occurrence of L A1a is -CHR L - or -C(R L ) 2 -; each remaining occurrence of L A1a is -CH 2 -; and each occurrence of L A1b is -CH 2 -. In some embodiments of (L-2) or (L-2a), one occurrence of L A1b is -CHR L - or -C(R L )2-; each remaining occurrence of L A1b is -CH2-; and each occurrence of L A1a is -CH2-. In some embodiments, L is: –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-L A4 -bb; –(L A3 )0-1-(L A1 )0-5- L A4 -(L A1 )0-5-L A3 -L A4 -bb; or –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-L A4 -L A3 -bb, provided that L contains 2-7 L A1 ; and wherein bb represents the point of attachment to Ring C. In some embodiments, L is: –(L A3 ) 0-1 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 -(4-10 (e.g.,6) membered heterocyclylene)- bb; –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-L A3 -(4-10 (e.g.,6) membered heterocyclylene)-bb; or –(L A3 )0- 1 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 -L A3 -(4-10 (e.g.,6) membered heterocyclylene)-L A3 - bb , provided that L contains 2-7 L A1 ; wherein the 4-10 (e.g., 6) membered heterocyclylene is optionally substituted with 1-3 R a ; and wherein bb represents the point of attachment to Ring C. In some embodiments, L is: –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-(piperazinylene)-bb; – (L A3 ) 0-1 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 -(piperidinylene)- bb ; –(L A3 ) 0-1 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 - (pyrrolidinylene)-bb; –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-C(=O)-(piperazinylene)-bb; –(L A3 )0-1- (L A1 )0-5-L A4 -(L A1 )0-5-C(=O)-(piperidinylene)-bb; or –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5- (piperidinylene)-O- bb , provided that L contains 2-5 L A1 ; wherein the piperazinylene, piperidinylene, and pyrrolidinylene are each optionally substituted with 1-3 R a , and wherein bb represents the point of attachment to Ring C. In some embodiments, 0-1 occurrence of L A1 is -CHR L - or -C(R L )2-; and each remaining occurrence of L A1 is -CH2-. In some embodiments, L is: –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-(4-10 membered heterocyclylene)-bb; or –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-L A3 -(4-10 membered heterocyclylene)- bb ; provided that L contains 2-7 L A1 , further provided that the 4-10 membered heterocyclylene contains two ring nitrogen atoms and no additional ring heteroatoms, wherein the 4-10 membered heterocyclylene is optionally substituted with 1-3 R a ; and wherein bb represents the point of attachment to Ring C. In some embodiments, L is: –(L A3 ) 0-1 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 - (piperazinylene)-bb; or –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-C(=O)-(piperazinylene)-bb; provided that L contains 2-5 L A1 ; wherein the piperazinylene is optionally substituted with 1-3 R a ; and bb represents the point of attachment to Ring C. In some embodiments, L is: –(L A3 ) 0-1 -(L A1 ) 0- 5-L A4 -(L A1 )0-5-(piperazinylene)-bb; or –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-C(=O)-(piperazinylene)- bb; provided that L contains 2-5 L A1 ; and wherein bb represents the point of attachment to Ring C. In some embodiments, 0-1 occurrence of L A1 is -CHR L - or -C(R L ) 2 -; and each remaining occurrence of L A1 is -CH 2 -. , , , wherein bb represents the point of attachment to Ring C. For example, L can be selected from the group consisting of:

wherein bb represents the point of attachment to Ring C. In some embodiments, L is a divalent group of Formula (L-3) or (L-4): Formula (L-4) wherein: a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5 (e.g., from 2 to 5); L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; m8 is 0, 1, or 2; and bb represents the point of attachment to Ring C. In some embodiments of (L-3) or (L-4), m8 is 0. In some embodiments of (L-3) or (L- 4), m8 is 1. In some embodiments of (L-3) or (L-4), m8 is 2. In some embodiments of (L-3) or (L-4), a3 is 1. In some embodiments, L A3 is O. In some embodiments of (L-3) or (L-4), a3 is 1; and L A3 is -O-. In some embodiments of (L-3) or (L-4), a1a + a1b is 3 or 4. For example, a1a + a1b can be 3. For example, a1a + a1b can be 4. In some embodiments of (L-3) or (L-4), a1a + a1b is 5. In some embodiments of (L-3) or (L-4), a1a + a1b is 2. In some embodiments of (L-3) or (L-4), a1a + a1b is 2 to 5. In some embodiments of (L-3) or (L-4), each occurrence of L A1a and L A1b is -CH 2 -. In some embodiments of (L-3) or (L-4), one occurrence of L A1a is -CHR L - or -C(R L ) 2 -; each remaining occurrence of L A1a is -CH2-; and each occurrence of L A1b is -CH2-. In some embodiments of (L-3) or (L-4), one occurrence of L A1b is -CHR L - or -C(R L ) 2 -; each remaining occurrence of L A1b is -CH 2 -; and each occurrence of L A1a is -CH 2 -. In some embodiments, L is: –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-(4-10 (e.g., 6) membered heterocyclylene)- bb ; –(L A3 ) 0-1 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 -L A3 -(4-10 (e.g., 6) membered heterocyclylene)-bb; or –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-(4-10 (e.g., 6) membered heterocyclylene)-L A3 -bb; provided that L contains 2-7 L A1 , further provided that the 4-10 (e.g., 6) membered heterocyclylene contains one ring nitrogen atom and no additional ring heteroatoms, wherein the 4-10 membered heterocyclylene is optionally substituted with 1-3 R a ; and wherein bb represents the point of attachment to Ring C. In some embodiments, L is: – (L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-(piperidinylene)-bb; –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-C(=O)- (piperidinylene)- bb ; or –(L A3 ) 0-1 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 -(piperidinylene)-O- bb ; provided that L contains 2-5 L A1 ; wherein the piperidinylene is optionally substituted with 1-3 R a ; and bb represents the point of attachment to Ring C. In some embodiments, L is: –(L A3 )0-1-(L A1 )0-5- L A4 -(L A1 ) 0-5 -(piperidinylene)- bb ; provided that L contains 2-5 L A1 ; and wherein bb represents the point of attachment to Ring C. In some embodiments, 0-1 occurrence of L A1 is -CHR L - or -C(R L )2-; and each remaining occurrence of L A1 is -CH2-. For example, can be selected from the group consisting of: , , , and , wherein bb represents the point of attachment to Ring C. In some embodiments, L is: –(L A3 ) 0-1 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 -L A4 -L A3 - bb ; provided that L contains 2-7 L A1 ; and wherein bb represents the point of attachment to Ring C. In some embodiments, L is: –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-(4-10 (e.g., 6) membered heterocyclylene)- O- bb , provided that L contains 2-7 L A1 ; wherein the 4-10 (e.g., 6) membered heterocyclylene is optionally substituted with 1-3 R a ; and wherein bb represents the point of attachment to Ring C. In some embodiments, 0-1 occurrence of L A1 is -CHR L - or -C(R L )2-; and each remaining occurrence of L A1 is -CH 2 -. For example, L can , wherein bb represents the point of attachment to Ring C. In some embodiments, L is a divalent group of Formula (L-3a) or (L-3b): Formula (L-3b) wherein: a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; n2 and n3 are independently 0, 1, or 2; m8 is 0, 1, or 2; and bb represents the point of attachment to Ring C. In some embodiments, L is a divalent group of Formula (L-3c): Formula (L-3c) wherein: a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; L A4a is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; n2 and n3 are independently 0, 1, or 2; m8 is 0, 1, or 2; L A4b is 7-10 membered bicyclic nitrogen-containing heterocyclylene optionally substituted with 1-3 R a ; and bb represents the point of attachment to Ring C. In some embodiments of Formula (L-3c), L A4b is a 7-10 membered spirocyclic nitrogen-containing heterocyclylene optionally substituted with 1-3 R a . For example, L A4b can be . In some embodiments of Formula (L-3c), L A4a is a 4-10 membered heterocyclylene optionally substituted with 1-3 R a . In some embodiments, L is a divalent group of Formula (L-3d) or (L-4a): Formula (L-4a) wherein: a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; n2 and n3 are independently 0, 1, or 2; m8 is 0, 1, or 2; and bb represents the point of attachment to Ring C. In some embodiments of (L-3a), (L-3b), (L-3c), (L-3d), or (L-4a), a3 is 1. In some embodiments, L A3 is O. In some embodiments of (L-3a), a3 is 1; and L A3 is -O-. In some embodiments of (L-3a), (L-3b), (L-3c), (L-3d), or (L-4a), a1a + a1b is 3 or 4. For example, a1a + a1b can be 3. For example, a1a + a1b can be 4. In some embodiments of (L-3a), (L-3b), (L-3c), (L-3d), or (L-4a), a1a + a1b is 5. In some embodiments of (L-3a), (L-3b), (L-3c), (L-3d), or (L-4a), a1a + a1b is 2. In some embodiments of (L-3a), (L-3b), (L-3c), (L-3d), or (L-4a), each occurrence of L A1a and L A1b is -CH 2 -. In some embodiments of (L-3a), (L-3b), (L-3c), (L-3d), or (L-4a), one occurrence of L A1a is -CHR L - or -C(R L )2-; each remaining occurrence of L A1a is -CH2-; and each occurrence of L A1b is -CH2-. In some embodiments of (L-3a), (L-3b), (L-3c), (L-3d), or (L-4a), one occurrence of L A1b is -CHR L - or -C(R L ) 2 -; each remaining occurrence of L A1b is -CH 2 -; and each occurrence of L A1a is -CH 2 -. In some embodiments of (L-3a), (L-3b), or (L-4a), m8 is 0. In some embodiments of (L-3a), (L-3b), (L-3c), or (L-4a), m8 is 1. In some embodiments of (L-3a), (L-3b), (L-3c), or (L-4a), m8 is 2. In some embodiments of (L-3a), (L-3b), (L-3d), or (L-4a), n2 is 1; and n3 is 1. In some embodiments of (L-3a), (L-3b), (L-3d), or (L-4a), n2 is 0; and n3 is 1. In some embodiments, L is: –(L A3 )0-1-(L A1 )2-3-L A3 -L A4 -(L A1 )0-5-(L A3 )1-2-bb; provided that L contains 2-7 L A1 ; and wherein bb represents the point of attachment to Ring C. In some embodiments, L is: –(L A3 ) 0-1 -CH 2 CH 2 -O-L A4 -(L A1 ) 0-5 -(L A3 ) 1-2 - bb ; wherein bb represents the point of attachment to Ring C. In some embodiments, L A4 is selected from the group consisting of: C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . In some embodiments, 0-1 occurrence of L A1 is -CHR L - or -C(R L )2-; and each remaining occurrence of L A1 is -CH2-. For example, L can be , wherein bb represents the point of attachment to Ring C. In some embodiments, L is a divalent group of Formula (L-5): Formula (L-5) wherein: a1 is an integer from 0 to 3; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and bb represents the point of attachment to Ring C. In some embodiments of (L-5), a1 is 1. In some embodiments of (L-5), 0-1 occurrence of L A1 is -CHR L - or -C(R L )2-; and each remaining occurrence of L A1 is -CH 2 -. In some embodiments, L is: -(L A1 )1-5-L A3 -L A4 -(L A1 )0-5-(L A3 )1-2-bb; provided that L contains 2-7 L A1 ; and wherein bb represents the point of attachment to Ring C. In some embodiments, L is: -(L A1 ) 2-3 -O-L A4 -(L A1 ) 0-5 -C(=O)N(R d )- bb ; provided that L contains 2-7 L A1 ; and wherein bb represents the point of attachment to Ring C. In some embodiments, L A4 is 4- 10 membered nitrogen-containing heterocyclylene optionally substituted with 1-3 R a . In some embodiments, 0-1 occurrence of L A1 is -CHR L - or -C(R L ) 2 -; and each remaining occurrence of L A1 is -CH 2 -. For example, L can , wherein bb represents the point of attachment to Ring C. In some embodiments, L is a divalent group of Formula (L-5a): Formula (L-5a) wherein: a1a and a1b are independently 1, 2, or 3; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and bb represents the point of attachment to Ring C. In some embodiments of Formula (L-5a), a1a is 3. In some embodiments of Formula (L-5a), a1b is 1. In some embodiments of Formula (L-5a), each occurrence of L A1a and L A1b is -CH2-. In some embodiments of Formula (L-5a), one occurrence of L A1a is -CHR L - or -C(R L ) 2 -; each remaining occurrence of L A1a is -CH 2 -; and each occurrence of L A1b is -CH 2 -. In some embodiments of Formula (L-5a), one occurrence of L A1b is -CHR L - or -C(R L )2-; each remaining occurrence of L A1b is -CH2-; and each occurrence of L A1a is -CH2-. In some embodiments of (L-1), (L-2), (L-2a), (L-3), (L-3a), (L-3b), (L-3d), (L-4), (L- 4a), (L-5) or (L-5a), L A4 is 4-10 membered heterocyclylene optionally substituted with 1-3 R a . In some embodiments, L A4 is 4-10 membered nitrogen-containing heterocyclylene optionally substituted with 1-3 R a . In some embodiments of (L-1), (L-2), (L-2a), (L-3), (L-3a), (L-3b), (L-3d), (L-4), (L- 4a), (L-5) or (L-5a), L A4 is selected from the group consisting of: , , , each optionally substituted with 1-3 R a at one or more ring carbon atoms, wherein cc represents the point of attachment to L A1b or L A1 . In some embodiments, each R a present on L A4 is independently F or C 1-3 alkyl optionally substituted with 1-3 F. In some embodiments of (L-1), (L-2), (L-2a), (L-3), (L-3a), (L-3b), (L-3d), (L-4), (L- 4a), (L-5) or (L-5a), L A4 is C 3-10 cycloalkylene optionally substituted with 1-3 R a . For example, L A4 can be 1,4-cyclohexylene optionally substituted with 1-3 R a . In some embodiments of (L-1), (L-2), (L-2a), (L-3), (L-3a), (L-3b), (L-3d), (L-4), (L- 4a), (L-5) or (L-5a), L A4 is phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . In some embodiments, L A4 is phenylene optionally substituted with 1-3 R a . In some embodiments, L A4 is 1,2-phenylene optionally substituted with 1-3 R a . In some embodiments, L A4 is 1,4-phenylene optionally substituted with 1-3 R a . In some embodiments, L A4 is 1,3-phenylene optionally substituted with 1-3 R a . In some embodiments, L is –(L A3 ) 0-1 -(L A1 ) 2-9 -(L A3 ) 0-1 -(piperazinylene)- bb , wherein bb represents the point of attachment to Ring C. In some embodiments, 0-1 occurrence of L A1 is -CHR L - or -C(R L )2-; and each remaining occurrence of L A1 is -CH2-. In some embodiments, L is a divalent group of Formula (L-6): Formula (L-6) wherein: L A3a is selected from the group consisting of: -O-, -N(H)-, and -N(C 1-3 alkyl)-; a3a is 0 or 1; a1 is an integer from 2 to 11; a3c is 0 or 1; and bb represents the point of attachment to Ring C. In some embodiments of (L-6), a3a is 1. In some embodiments, L A3a is -O-. In some embodiments of (L-6), a3a is 1 and L A3a is -O-. In some embodiments of (L-6), 0-1 occurrence of L A1 is -CHR L - or -C(R L ) 2 -; and each remaining occurrence of L A1 is -CH2-. In some embodiments of (L-6), a1 is an integer from 2 to 5. In some embodiments of (L-6), a1 is an integer from 5 to 7 (e.g., 6). In some embodiments of (L-6), a1 is an integer from 7 to 9 (e.g., 8). In some embodiments of (L-6), a1 is an integer from 9 to 11. In some embodiments, L is selected from the group consisting of: .

wherein bb represents the point of attachment to Ring C. In some embodiments, L is selected from the group consisting of: wherein bb represents the point of attachment to Ring C. In some embodiments, L is selected from the group consisting of:

wherein bb represents the point of attachment to Ring C. In some embodiments, L is selected from the group consisting of: wherein bb represents the point of attachment to Ring C. , bb represents the point of attachment to Ring C. In some embodiments, L is selected from the group consisting of:

wherein bb represents the point of attachment to Ring C. In some embodiments, L is selected from the group consisting of: wherein bb represents the point of attachment to Ring C. In some embodiments, , wherein bb represents the point of attachment to Ring C. In some embodiments, , wherein bb represents the point of attachment to Ring C. In some embodiments, L is ; and L A and n1 are defined according to (BB). In some embodiments, n1 is an integer from 3 to 5. In some embodiments, n1 is an integer from 5 to 9. In some embodiments, n1 is an integer from 9 to 15. In some embodiments, 1-4 occurrences of L A is L A3 . In some embodiments, 1-3 occurrences of L A is L A3 . In some embodiments, 2-3 occurrences of L A are L A3 . In some embodiments, 0-1 occurrence of L A3 is C(=O); and each remaining occurrence of L A3 is independently selected from the group consisting of: -O-; -N(H)-; and -N(C1-3 alkyl)-. In some embodiments, 0-2 occurrences of L A1 is -CHR L - or -C(R L ) 2 -; and each remaining occurrence of L A1 is -CH 2 -. In some embodiments, L A and n1 are defined according to (BB); and n1 is an integer from 3 to 5. In some embodiments, L A and n1 are defined according to (BB); and n1 is an integer from 9 to 15. In some embodiments, L A and n1 are defined according to (BB); and L is –(L A3 )0-2- (L A1 ) 1-15 -(L A3 ) 0-2 - bb , wherein bb represents the point of attachment to Ring C. In some embodiments, L is –(L A3 ) 0-1 -(L A1 ) 1-15 -(L A3 ) 1-2 - bb , wherein bb represents the point of attachment to Ring C. In some embodiments, L contains 2 or 3 L A3 . In some embodiments, 0-1 occurrence of L A3 is C(=O); and each remaining occurrence of L A3 is independently selected from the group consisting of: -O-; -N(H)-; and -N(C 1-3 alkyl)-. In some embodiments, 0-2 occurrences of L A1 is -CHR L - or -C(R L ) 2 -; and each remaining occurrence of L A1 is -CH 2 -. In some embodiments, each occurrence of L A1 is -CH2-. In some embodiments, one occurrence of L A1 is -CHR L - or -C(R L ) 2 -; and each remaining occurrence of L A1 is -CH 2 -. In some embodiments, each R L is independently selected from the group consisting of: -F and -C 1-3 alkyl optionally substituted with 1-3 F. In some embodiments, L is a divalent group of Formula (L-7): Formula (L-7) wherein: L A3a is selected from the group consisting of: -O-, -N(H)-, and -N(C1-3 alkyl)-; a3a is 0 or 1; a1 is an integer from 1 to 15; each L A3b is independently selected from the group consisting of: C(=O), -O-, N(H)-, and -N(C1-3 alkyl)-; a3b is 1 or 2; and bb represents the point of attachment to Ring C. In some embodiments In some embodiments of (L-7), –(L A3b )a3b- is -N(H)- or -N(C1-3 alkyl)-. In some embodiments of (L-7), 0-2 occurrences of L A1 is -CHR L - or -C(R L ) 2 -; and each remaining occurrence of L A1 is -CH2-. In some embodiments, each occurrence of L A1 is -CH2- . In some embodiments, L is (L-1); Ring , wherein aa represents the point of attachment to L, wherein the R a present on Ring A is methyl or CF 3 ; and R 1 is C(O)OH or C(O)OC1-6 alkyl. In some embodiments, L is (L-3); Ring , wherein aa represents the point of attachment to L, wherein the R a present on Ring A is methyl or CF 3 ; and R 1 is C(O)OH or C(O)OC 1-6 alkyl. In some embodiments, L is (L-3a); Ring , wherein aa represents the point of attachment to L, wherein the R a present on Ring A is methyl or CF3; and R 1 is C(O)OH or C(O)OC1-6 alkyl. In some embodiments, L is (L-3b); Ring , wherein aa represents the point of attachment to L, wherein the R a present on Ring A is methyl or CF 3 ; and R 1 is C(O)OH or C(O)OC 1-6 alkyl. In some embodiments, Ring , wherein aa represents the point of attachment to L, wherein the R a present on Ring A is methyl or CF 3 ; R 1 is C(O)OH or C(O)OC 1-6 alkyl; and Ring , wherein R aN is C 1-3 alkyl (e.g., methyl); c1 is 0 or 1; and R a2 is selected from the group consisting of halo (e.g., - F) and C1-3 alkyl optionally substituted with 1-3 F. In some embodiments of Formula (I), L is (L-1); Ring A is , wherein aa represents the point of attachment to L, wherein the R a present on Ring A is methyl or CF 3 ; R 1 is C(O)OH or C(O)OC 1-6 alkyl; and Ring , wherein R aN is C1-3 alkyl (e.g., methyl); c1 is 0 or 1; and R a2 is selected from the group consisting of halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 F. In some embodiments of Formula (I), L is (L-3); Ring A is , , wherein aa represents the point of attachment to L, wherein the R a present on Ring A is methyl or CF3; R 1 is C(O)OH or C(O)OC1-6 alkyl; and Ring C is , wherein R aN is C 1-3 alkyl (e.g., methyl); c1 is 0 or 1; and R a2 is selected from the group consisting of halo (e.g., -F) and C1-3 alkyl optionally substituted with 1-3 F. In some embodiments of Formula (I), L is (L-3a); Ring A is , , wherein aa represents the point of attachment to L, wherein the R a present on Ring A is methyl or CF 3 ; R 1 is C(O)OH or C(O)OC 1-6 alkyl; and Ring C is , wherein R aN is C 1-3 alkyl (e.g., methyl); c1 is 0 or 1; and R a2 is selected from the group consisting of halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 F. In some embodiments of Formula (I), L is (L-3b); Ring A is , , wherein aa represents the point of attachment to L, wherein the R a present on Ring A is methyl or CF3; R 1 is C(O)OH or C(O)OC1-6 alkyl; and Ring C is , wherein R aN is C1-3 alkyl (e.g., methyl); c1 is 0 or 1; and R a2 is selected from the group consisting of halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 F. In some embodiments, L is selected from the group consisting of the moieties depicted in Table (L), wherein bb represents the point of attachment to Ring C: Table (L)

In some embodiments of Formula (I), L is selected from the group consisting of the moieties depicted in Table (L); and Ring , wherein aa represents the point of attachment to L, wherein the R a present on Ring A is methyl or CF3; R 1 is C(O)OH or C(O)OC 1-6 alkyl. In some embodiments of Formula (I), L is selected from the group consisting of the moieties depicted in Table (L); R aN is C1-3 alkyl (e.g., methyl); c1 is 0 or 1; and R a2 is selected from the group consisting of halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 F. In some embodiments of Formula (I), L is selected from the group consisting of the moieties depicted in Table (L); Ring , wherein aa represents the point of attachment to L, wherein the R a present on Ring A is methyl or CF 3 ; R 1 is C(O)OH or C(O)OC1-6 alkyl; and Ring , wherein R aN is C1-3 alkyl (e.g., methyl); c1 is 0 or 1; and R a2 is selected from the group consisting of halo (e.g., -F) and C1-3 alkyl optionally substituted with 1-3 F. In some embodiments, L is (L-1); and the compounds of Formula (I) are compounds of Formula (I-A): Formula (I-A) or pharmaceutically acceptable salts thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5 (e.g., from 2 to 5); L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; and L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . In some embodiments, L is (L-1); and the compounds of Formula (I) are compounds of Formula (I-B): Formula (I-B) or pharmaceutically acceptable salts thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5 (e.g., from 2 to 5); L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L ) 2 -; and L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . In some embodiments of Formula (I-A) or (I-B), a3 is 1. In some embodiments, L A3 is -O-. In some embodiments of Formula (I-A) or (I-B), a3 is 1; and L A3 is -O-. In some embodiments of Formula (I-A) or (I-B), a3 is 0. In some embodiments, L is (L-2); and the compounds of Formula (I) are compounds of Formula (I-C): Formula (I-C) or pharmaceutically acceptable salts thereof, wherein: m6 is 0 or 1; a3a is 0 or 1; L A3a and L A3b are independently selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 7; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . In some embodiments, L is (L-2); and the compounds of Formula (I) are compounds of Formula (I-D):

Formula (I-D) or pharmaceutically acceptable salts thereof, wherein: m6 is 0 or 1; a3a is 0 or 1; L A3a and L A3b are independently selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 7; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . In some embodiments of Formula (I-C) or (I-D), L A3b is -N(H)- or -N(C1-3 alkyl)-. In some embodiments of Formula (I-C) or (I-D), a3a is 1. In some embodiments, L A3a is -O-. In some embodiments of Formula (I-C) or (I-D), a3a is 0. In some embodiments, L is (L-3); and the compounds of Formula (I) are compounds of Formula (I-E):

Formula (I-E) or pharmaceutically acceptable salts thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and m8 is 0, 1, or 2. In some embodiments, L is (L-3a); and the compounds of Formula (I) are compounds of Formula (I-Ea): Formula (I-Ea) or pharmaceutically acceptable salts thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; n2 and n3 are independently 0, 1, or 2; and m8 is 0, 1, or 2. In some embodiments, L is (L-3b); and the compounds of Formula (I) are compounds of Formula (I-Eb): Formula (I-Eb) or pharmaceutically acceptable salts thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; n2 and n3 are independently 0, 1, or 2; and m8 is 0, 1, or 2. In some embodiments, L is (L-4); and the compounds of Formula (I) are compounds of Formula (I-F): Formula (I-F) or pharmaceutically acceptable salts thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and m8 is 0, 1, or 2. In some embodiments of Formula (I-E), (I-Ea), (I-Eb), or (I-F), m8 is 0. In some embodiments of Formula (I-E), (I-Ea), (I-Eb), or (I-F), m8 is 1. In some embodiments of Formula (I-E), (I-Ea), (I-Eb), or (I-F), m8 is 2. In some embodiments of Formula (I-Ea) or (I-Eb), n2 is 1; and n3 is 1. In some embodiments of Formula (I-Ea) or (I-Eb), n2 is 0; and n3 is 1. In some embodiments, L is (L-3) or (L-4); and the compounds of Formula (I) are compounds of Formula (I-E) or (I-F), or pharmaceutically acceptable salts thereof. In some embodiments, the compounds are compounds of Formula (I-E), or pharmaceutically acceptable salts thereof. In some embodiments, the compounds are compounds of Formula (I- F), or pharmaceutically acceptable salts thereof. In some embodiments, the compounds are compounds of Formula (I-Ea), or pharmaceutically acceptable salts thereof. In some embodiments, the compounds are compounds of Formula (I-Eb), or pharmaceutically acceptable salts thereof. In some embodiments, L is (L-3); and the compounds of Formula (I) are compounds of Formula (I-G): Formula (I-G) or pharmaceutically acceptable salts thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and m8 is 0, 1, or 2. In some embodiments, L is (L-3a); and the compounds of Formula (I) are compounds of Formula (I-Ga): Formula (I-Ga) or pharmaceutically acceptable salts thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; n2 and n3 are independently 0, 1, or 2; and m8 is 0, 1, or 2. In some embodiments, L is (L-4); and the compounds of Formula (I) are compounds of Formula (I-H): Formula (I-H) or pharmaceutically acceptable salts thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; and L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and m8 is 0, 1, or 2. In some embodiments, L is (L-3) or (L-4); and the compounds of Formula (I) are compounds of Formula (I-G) or (I-H), or pharmaceutically acceptable salts thereof. In some embodiments, the compounds are compounds of Formula (I-G), or pharmaceutically acceptable salts thereof. In some embodiments, the compounds are compounds of Formula (I- H), or pharmaceutically acceptable salts thereof. In some embodiments, the compounds are compounds of Formula (I-Ga), or pharmaceutically acceptable salts thereof. In some embodiments of Formula (I-E), (I-Ea), (I-Eb), (I-F), (I-G), (I-Ga), or (I-H), a3 is 1. In some embodiments, L A3 is -O-. In some embodiments of Formula (I-E), (I-Ea), (I- Eb), (I-F), (I-G), (I-Ga), or (I-H), a3 is 1; and L A3 is -O-. In some embodiments of Formula (I-E), (I-Ea), (I-Eb), (I-F), (I-G), (I-Ga), or (I-H), a3 is 0. In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb) (I-F), (I-G), (I-Ga), or (I-H), a1a + a1b is 3. In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb) (I-F), (I-G), (I-Ga), or (I-H), a1a + a1b is 4. In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb) (I-F), (I-G), (I-Ga), or (I-H), a1a + a1b is 2 or 5. For example, a1a + a1b can be 5. In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb) (I-F), (I-G), (I-Ga), or (I-H), each occurrence of L A1a and L A1b is -CH2-. In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb) (I-F), (I-G), (I-Ga), or (I-H), one occurrence of L A1a is -CHR L - or -C(R L ) 2 -; each remaining occurrence of L A1a is -CH2-; and each occurrence of L A1b is -CH2-. In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb) (I-F), (I-G), (I-Ga), or (I-H), one occurrence of L A1b is -CHR L - or -C(R L ) 2 -; each remaining occurrence of L A1b is -CH 2 -; and each occurrence of L A1a is -CH 2 -. In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb) (I-F), (I-G), (I-Ga), or (I-H), L A4 is 4-10 membered (e.g., monocyclic or bicyclic) heterocyclylene optionally substituted with 1-3 R a . For example, L A4 can be selected from the group consisting substituted with 1-3 R a at one or more ring carbon atoms, wherein cc represents the point of attachment to L A1b . In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb) (I-F), (I-G), (I-Ga), or (I-H), each R a is independently selected from the group consisting of: halo, - OH, C1-3 alkoxy, C1-3 haloalkoxy, and C1-3 alkyl optionally substituted with 1-3 R c . In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb) (I-F), (I-G), (I-Ga), or (I-H), each R a present on L A4 is independently a C1-3 alkyl optionally substituted with 1-3 F. In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb) (I-F), (I-G), (I-Ga), or (I-H), L A4 is C3-10 cycloalkylene optionally substituted with 1-3 R a . For example, L A4 can be 1,4-cyclohexylene optionally substituted with 1-3 R a . In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb) (I-F), (I-G), (I-Ga), or (I-H), L A4 is phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . For example, L A4 can be 1,4-phenylene optionally substituted with 1-3 R a . For example, L A4 can be 1,2-phenylene or 1,3-phenylene, each of which is optionally substituted with 1-3 R a . In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb) (I-F), (I-G), (I-Ga), or (I-H), m6 is 0. In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb) (I-F), (I-G), (I-Ga), or (I-H), the R a present on Ring A is C 1-3 alkyl optionally substituted with 1-3 -F. In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb) (I-F), (I-G), (I-Ga), or (I-H), R 1 is C(O)OH. In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb) (I-F), (I-G), (I-Ga), or (I-H), R 1 is C(O)OC 1-6 alkyl. In some embodiments, L is (L-7) wherein a3a is 0; and the compounds of Formula (I) are compounds of Formula (II-A): Formula (II-A) or pharmaceutically acceptable salts thereof, wherein: Ring A is phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; L T1 is a bond or CH 2 ; A * is H or C3-15 cycloalkyl optionally substituted with 1-3 R a ; L A3b is selected from the group consisting of: C(=O), -O-, N(H)-, and -N(C 1-3 alkyl)-; a3b is 1 or 2; and a1 is an integer from 1 to 15. In some embodiments of Formula (II-A), Ring A is 5-membered heteroarylene optionally substituted with 1-2 R a . In some embodiments of Formula (II-A), Ring A is selected from the group consisting of: , wherein aa represents the point of attachment to L T1 . In some embodiments of Formula (II-A), L T1 is CH2. In some embodiments of Formula (II-A), A* is H. In some embodiments of Formula (II-A), A* is C 3-15 cycloalkyl optionally substituted with 1-3 R a . For example, A* can be adamantyl optionally substituted with 1-3 R a . In some embodiments of Formula (II-A), a1 is an integer from 1 to 3. In some embodiments of Formula (II-A), a1 is an integer from 4 to 6. In some embodiments of Formula (II-A), a1 is an integer from 7 to 9. In some embodiments of Formula (II-A), a1 is an integer from 10 to 15. In some embodiments of Formula (II-A), –(L A3b )a3b- is , wherein bb represents the point of attachment to Ring C. In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb), (I- F), (I-G), (I-Ga), (I-H), or (II-A), m2 is 0. In some embodiments of Formula (I-A), (I-B), (I- C), (I-D), (I-E), (I-Ea), (I-Eb), (I-F), (I-G), (I-Ga), (I-H), or (II-A), m4 is 0. In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb), (I- F), (I-G), (I-Ga), (I-H), or (II-A), Ring . In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb), (I- F), (I-G), (I-Ga), (I-H), or (II-A), Ring . In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb), (I- F), (I-G), (I-Ga), (I-H), or (II-A), Ring . In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb), (I- F), (I-G), (I-Ga), (I-H), or (II-A), R aN is C1-3 alkyl. For example, R aN can be methyl. In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb), (I- F), (I-G), (I-Ga), (I-H), or (II-A), Ring methyl); c1 is 0 or 1; and R a2 is selected from the group consisting of: halo (e.g., -F) and C 1-3

. In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb), (I- F), (I-G), (I-Ga), (I-H), or (II-A), c1 is 0. In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb), (I- F), (I-G), (I-Ga), (I-H), or (II-A), L C is a bond. In some embodiments of Formula (I-A), (I- B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb), (I-F), (I-G), (I-Ga), (I-H), or (II-A), L C is -NH-. In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb), (I- F), (I-G), (I-Ga), (I-H), or (II-A), X is CH. In some embodiments of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-Ea), (I-Eb), (I- . In some embodiments, the compounds of Formula (I-A) are compounds of Formula (I- Formula (I-A-1) or pharmaceutically acceptable salts thereof, wherein: R 1 is C(O)OH or C(O)OC 1-6 alkyl; R a1 is C1-3 alkyl optionally substituted with 1-3 F; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5 (e.g., from 2 to 5); L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . In some embodiments, the compounds of Formula (I-A) are compounds of Formula (I- A-2):

Formula (I-A-2) or pharmaceutically acceptable salts thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5 (e.g., from 2 to 5); L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C1-3 alkyl optionally substituted with 1-3 F; and R aN is C1-3 alkyl. In some embodiments, the compounds of Formula (I-A) are compounds of Formula (I- A-3): Formula (I-A-3) or pharmaceutically acceptable salts thereof, wherein: R 1 is C(O)OH or C(O)OC1-6 alkyl; R a1 is C1-3 alkyl optionally substituted with 1-3 F; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5 (e.g., from 2 to 5); L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and Ring , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 F; and R aN is C1-3 alkyl. In some embodiments of Formula (I-A-1), (I-A-2), or (I-A-3): a3 is 1; L A3 is -O-; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, - CH(Me)-, and -CF 2 -; and L A4 is 4-10 membered (e.g., nitrogen containing) heterocyclylene optionally substituted with 1-3 R a . In some embodiments of Formula (I-A-1), (I-A-2), or (I-A-3): a3 is 1; L A3 is -O-; a1b is 1; L A1b is -CH2-; each L A1a is independently selected from the group consisting of: -CH2- and -CH(Me)- , provided that 0-1 L A1a is -CH(Me)-; and L A4 is 4-10 membered nitrogen-containing heterocyclylene optionally substituted with 1-2 substituents independently selected from the group consisting of: -F and C1-3 alkyl optionally substituted with 1-3 -F. In some embodiments of Formula (I-A-1), (I-A-2), or (I-A-3): a3 is 1; L A3 is -O-; a1b is 1; L A1b is -CH2-; a1a is 1, 2, or 3; each L A1a is independently selected from the group consisting of: -CH 2 - and -CH(Me)- , provided that 0-1 L A1a is -CH(Me)-; and , , , , , , wherein cc represents the point of attachment to L A1b . In some embodiments of Formula (I-A), (I-A-1), (I-A-2), or (I-A-3), the moiety is selected from the groups depicted in Table (L-I- A): Table (L-I-A)

wherein bb represents the point of attachment to Ring C. In some embodiments, the compounds of Formula (I-B) are compounds of Formula (I- Formula (I-B-1) or pharmaceutically acceptable salts thereof, wherein: R 1 is C(O)OH or C(O)OC1-6 alkyl; R a1 is C1-3 alkyl optionally substituted with 1-3 F; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5 (e.g., from 2 to 5); L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . In some embodiments, the compounds of Formula (I-B) are compounds of Formula (I- Formula (I-B-2) or pharmaceutically acceptable salts thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5 (e.g., from 2 to 5); L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and Ring from the group consisting of: halo (e.g., -F) and C1-3 alkyl optionally substituted with 1-3 F; and R aN is C 1-3 alkyl. In some embodiments, the compounds of Formula (I-B) are compounds of Formula (I- B-3): Formula (I-B-3) or pharmaceutically acceptable salts thereof, wherein: R 1 is C(O)OH or C(O)OC1-6 alkyl; R a1 is C1-3 alkyl optionally substituted with 1-3 F; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5 (e.g., from 2 to 5); L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and Ring , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 F; and R aN is C1-3 alkyl. In some embodiments of Formula (I-B), (I-B-1), (I-B-2), or (I-B-3): a3 is 1; L A3 is -O-; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, - CH(Me)-, and -CF 2 -; and L A4 is 4-10 membered (e.g., nitrogen containing) heterocyclylene optionally substituted with 1-3 R a . In some embodiments of Formula (I-B-1), (I-B-2), or (I-B-3): a3 is 1; L A3 is -O-; a1b is 1; L A1b is -CH2-; each L A1a is independently selected from the group consisting of: -CH 2 - and -CH(Me)- , provided that 0-1 L A1a is -CH(Me)-; and L A4 is 4-10 membered nitrogen-containing heterocyclylene optionally substituted with 1-2 substituents independently selected from the group consisting of: -F and C1-3 alkyl optionally substituted with 1-3 -F. In some embodiments of Formula (I-B-1), (I-B-2), or (I-B-3): a3 is 1; L A3 is -O-; a1b is 1; L A1b is -CH 2 -; a1a is 1, 2, or 3; each L A1a is independently selected from the group consisting of: -CH 2 - and -CH(Me)- , provided that 0-1 L A1a is -CH(Me)-; and L A4 is selected from the group consisting of: , , , wherein cc represents the point of attachment to L A1b . In some embodiments of Formula (I-B-1), (I-B-2), or (I-B-3), the moiety is selected from the groups depicted in Table (L-I- B):

wherein bb represents the point of attachment to Ring C. In some embodiments, the compounds of Formula (I-E) are compounds of Formula (I- E-1): Formula (I-E-1) or pharmaceutically acceptable salts thereof, wherein: R 1 is C(O)OH or C(O)OC1-6 alkyl; R a1 is C 1-3 alkyl optionally substituted with 1-3 F; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; and L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and m8 is 0, 1, or 2. In some embodiments, the compounds of Formula (I-E) are compounds of Formula (I- E-2): Formula (I-E-2) or pharmaceutically acceptable salts thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; m8 is 0, 1, or 2; and Ring from the group consisting of: halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 F; and R aN is C1-3 alkyl. In some embodiments, the compounds of Formula (I-E) are compounds of Formula (I- E-3): Formula (I-E-3) or pharmaceutically acceptable salts thereof, wherein: R 1 is C(O)OH or C(O)OC1-6 alkyl; R a1 is C1-3 alkyl optionally substituted with 1-3 F; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; m8 is 0, 1, or 2; and Ring , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 F; and R aN is C1-3 alkyl. In some embodiments of Formula (I-E-1), (I-E-2), or (I-E-3): a3 is 1; L A3 is -O-; a1a is 0; and L A4 is selected from the group consisting of: C3-10 cycloalkylene optionally substituted with 1-3 R a ; and phenylene optionally substituted with 1-3 R a . In some embodiments of Formula (I-E-1), (I-E-2), or (I-E-3): a3 is 1; L A3 is -O-; a1a is 0; a1b is 3 or 4; each L A1b is independently selected from the group consisting of: -CH 2 - and -CH(Me)- , provided that 0-1 of L A1b is -CH(Me)-; and L A4 is selected from the group consisting of: .g., ) and . In some embodiments of Formula (I-E-1), (I-E-2), or (I-E-3): a3 is 1; L A3 is -O-; a1a is 2, 3, or 4; and L A4 is 4-10 membered heterocyclylene optionally substituted with 1-3 R a . In some embodiments of Formula (I-E-1), (I-E-2), or (I-E-3): a3 is 1; L A3 is -O-; a1a is 2, 3, or 4; each L A1a is CH2; a1b is 0, 1, or 2; each L A1b is CH 2 ; and L A4 is 4-8 membered nitrogen-containing heterocyclylene optionally substituted with 1-3 R a . In some embodiments of Formula (I-E-1), (I-E-2), or (I-E-3): a3 is 1; L A3 is -O-; a1a is 2, 3, or 4; each L A1a is CH2; a1b is 0, 1, or 2; each L A1b is CH 2 ; and L A4 is selected from the group consisting of: , , , wherein cc represents the point of attachment to L A1b . In some embodiments of Formula (I-E), (I-E-1), (I-E-2), or (I-E-3), the moiety is selected from the groups depicted in Table (L-I-E): Table (L-I-E)

wherein bb represents the point of attachment to Ring C. In some embodiments, the compounds of Formula (I-Ea) are compounds of Formula (I-Ea-1): Formula (I-Ea-1) or pharmaceutically acceptable salts thereof, wherein: R 1 is C(O)OH or C(O)OC 1-6 alkyl; R a1 is C1-3 alkyl optionally substituted with 1-3 F; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L ) 2 -; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; n2 and n3 are independently 0, 1, or 2; and m8 is 0, 1, or 2. In some embodiments, the compounds of Formula (I-Ea) are compounds of Formula (I-Ea-2): Formula (I-Ea-2) or pharmaceutically acceptable salts thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; n2 and n3 are independently 0, 1, or 2; m8 is 0, 1, or 2; and Ring , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 F; and R aN is C1-3 alkyl. In some embodiments, the compounds of Formula (I-Ea) are compounds of Formula (I-Ea-3): Formula (I-Ea-3) or pharmaceutically acceptable salts thereof, wherein: R 1 is C(O)OH or C(O)OC1-6 alkyl; R a1 is C1-3 alkyl optionally substituted with 1-3 F; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; m8 is 0, 1, or 2; n2 and n3 are independently 0, 1, or 2; and Ring , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 F; and R aN is C1-3 alkyl. In some embodiments of Formula (I-Ea-1), (I-Ea-2), or (I-Ea-3): a3 is 1; L A3 is -O-; a1a is 0; and L A4 is selected from the group consisting of: C3-10 cycloalkylene optionally substituted with 1-3 R a ; and phenylene optionally substituted with 1-3 R a . In some embodiments of Formula (I-Ea-1), (I-Ea-2), or (I-Ea-3): a3 is 1; L A3 is -O-; a1a is 0; a1b is 3 or 4; each L A1b is independently selected from the group consisting of: -CH 2 - and -CH(Me)- , provided that 0-1 of L A1b is -CH(Me)-; and L A4 is selected from the group consisting of: . In some embodiments of Formula (I-Ea-1), (I-Ea-2), or (I-Ea-3): a3 is 1; L A3 is -O-; a1a is 2, 3, or 4; and L A4 is 4-10 membered heterocyclylene optionally substituted with 1-3 R a . In some embodiments of Formula (I-Ea-1), (I-Ea-2), or (I-Ea-3): a3 is 1; L A3 is -O-; a1a is 2, 3, or 4; each L A1a is CH2; a1b is 0, 1, or 2; each L A1b is CH 2 ; and L A4 is 4-8 membered nitrogen-containing heterocyclylene optionally substituted with 1-3 R a . In some embodiments of Formula (I-Ea-1), (I-Ea-2), or (I-Ea-3): a3 is 1; L A3 is -O-; a1a is 2, 3, or 4; each L A1a is CH2; a1b is 0, 1, or 2; each L A1b is CH 2 ; and L A4 is selected from the group consisting of: , , , wherein cc represents the point of attachment to L A1b . In some embodiments of Formula (I-Ea-1), (I-Ea-2), or (I-Ea-3), n2 is 1; and n3 is 1. In some embodiments of Formula (I-Ea-1), (I-Ea-2), or (I-Ea-3), the moiety is selected from the groups depicted in Table (L-I-Ea): Table (L-I-Ea) wherein bb represents the point of attachment to Ring C. In some embodiments, the compounds of Formula (I-Eb) are compounds of Formula (I-Eb-1):

Formula (I-Eb-1) or pharmaceutically acceptable salts thereof, wherein: R 1 is C(O)OH or C(O)OC 1-6 alkyl; R a1 is C 1-3 alkyl optionally substituted with 1-3 F; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; and L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; n2 and n3 are independently 0, 1, or 2; and m8 is 0, 1, or 2. In some embodiments, the compounds of Formula (I-Eb) are compounds of Formula (I-Eb-2):

Formula (I-Eb-2) or pharmaceutically acceptable salts thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; m8 is 0, 1, or 2; n2 and n3 are independently 0, 1, or 2; and , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 F; and R aN is C1-3 alkyl. In some embodiments, the compounds of Formula (I-Eb) are compounds of Formula (I-Eb-3):

Formula (I-Eb-3) or pharmaceutically acceptable salts thereof, wherein: R 1 is C(O)OH or C(O)OC 1-6 alkyl; R a1 is C 1-3 alkyl optionally substituted with 1-3 F; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; m8 is 0, 1, or 2; n2 and n3 are independently 0, 1, or 2; and Ring , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C1-3 alkyl optionally substituted with 1-3 F; and R aN is C 1-3 alkyl. In some embodiments of Formula (I-Eb-1), (I-Eb-2), or (I-Eb-3): a3 is 1; L A3 is -O-; a1a is 0; and L A4 is selected from the group consisting of: C3-10 cycloalkylene optionally substituted with 1-3 R a ; and phenylene optionally substituted with 1-3 R a . In some embodiments of Formula (I-Eb-1), (I-Eb-2), or (I-Eb-3): a3 is 1; L A3 is -O-; a1a is 0; a1b is 3 or 4; each L A1b is independently selected from the group consisting of: -CH2- and -CH(Me)- , provided that 0-1 of L A1b is -CH(Me)-; and L A4 is selected from the group consisting of: and . In some embodiments of Formula (I-Eb-1), (I-Eb-2), or (I-Eb-3): a3 is 1; L A3 is -O-; a1a is 2, 3, or 4; and L A4 is 4-10 membered heterocyclylene optionally substituted with 1-3 R a . In some embodiments of Formula (I-Eb-1), (I-Eb-2), or (I-Eb-3): a3 is 1; L A3 is -O-; a1a is 2, 3, or 4; each L A1a is CH2; a1b is 0, 1, or 2; each L A1b is CH 2 ; and L A4 is 4-8 membered nitrogen-containing heterocyclylene optionally substituted with 1-3 R a . In some embodiments of Formula (I-Eb-1), (I-Eb-2), or (I-Eb-3): a3 is 1; L A3 is -O-; a1a is 2, 3, or 4; each L A1a is CH2; a1b is 0, 1, or 2; each L A1b is CH2; and wherein cc represents the point of attachment to L A1b . In some embodiments of Formula (I-Eb-1), (I-Eb-2), or (I-Eb-3), n2 is 1; and n3 is 1. In some embodiments of Formula (I-Eb-1), (I-Eb-2), or (I-Eb-3), the

wherein bb represents the point of attachment to Ring C. In some embodiments, the compounds of Formula (I-F) are compounds of Formula (I- F-1): Formula (I-F-1) or pharmaceutically acceptable salts thereof, wherein: R 1 is C(O)OH or C(O)OC 1-6 alkyl; R a1 is C 1-3 alkyl optionally substituted with 1-3 F; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; and L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and m8 is 0, 1, or 2. In some embodiments, the compounds of Formula (I-F) are compounds of Formula (I- F-2): Formula (I-F-2) or pharmaceutically acceptable salts thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L ) 2 -; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; m8 is 0, 1, or 2; and Ring , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 F; and R aN is C1-3 alkyl. In some embodiments, the compounds of Formula (I-F) are compounds of Formula (I- F-3): Formula (I-F-3) or pharmaceutically acceptable salts thereof, wherein: R 1 is C(O)OH or C(O)OC 1-6 alkyl; R a1 is C1-3 alkyl optionally substituted with 1-3 F; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; m8 is 0, 1, or 2; and Ring , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 F; and R aN is C1-3 alkyl. In some embodiments of Formula (I-F-1), (I-F-2), or (I-F-3): a3 is 1; L A3 is -O-; a1a is 0; and L A4 is selected from the group consisting of: C3-10 cycloalkylene optionally substituted with 1-3 R a ; and phenylene optionally substituted with 1-3 R a . In some embodiments of Formula (I-F-1), (I-F-2), or (I-F-3): a3 is 1; L A3 is -O-; a1a is 0; a1b is 3 or 4; each L A1b is independently selected from the group consisting of: -CH 2 - and -CH(Me)- , provided that 0-1 of L A1b is -CH(Me)-; and L A4 is selected from the group consisting of: . In some embodiments of Formula (I-F-1), (I-F-2), or (I-F-3): a3 is 1; L A3 is -O-; a1a is 2, 3, or 4; and L A4 is 4-10 membered heterocyclylene optionally substituted with 1-3 R a . In some embodiments of Formula (I-F-1), (I-F-2), or (I-F-3): a3 is 1; L A3 is -O-; a1a is 2, 3, or 4; each L A1a is CH2; a1b is 0, 1, or 2; each L A1b is CH 2 ; and L A4 is 4-8 membered nitrogen-containing heterocyclylene optionally substituted with 1-3 R a . In some embodiments of Formula (I-F-1), (I-F-2), or (I-F-3), the moiety is selected from the groups depicted in Table (L-I-F): Table (L-I-F)

wherein bb represents the point of attachment to Ring C. In some embodiments of Formula (I-G): a3 is 1; L A3 is -O-; a1a is 0; and L A4 is selected from the group consisting of: C3-10 cycloalkylene optionally substituted with 1-3 R a ; and phenylene optionally substituted with 1-3 R a . In some embodiments of Formula (I-G): a3 is 1; L A3 is -O-; a1a is 0; a1b is 3 or 4; each L A1b is independently selected from the group consisting of: -CH 2 - and -CH(Me)- , provided that 0-1 of L A1b is -CH(Me)-; and L A4 is selected from the group consisting of: ) and . In some embodiments of Formula (I-G): a3 is 1; L A3 is -O-; a1a is 2, 3, or 4; and L A4 is 4-10 membered heterocyclylene optionally substituted with 1-3 R a . In some embodiments of Formula (I-G-1): a3 is 1; L A3 is -O-; a1a is 2, 3, or 4; each L A1a is CH 2 ; a1b is 0, 1, or 2; each L A1b is CH2; and L A4 is 4-8 membered nitrogen-containing heterocyclylene optionally substituted with 1-3 R a . In some embodiments of Formula (I-G): a3 is 1; L A3 is -O-; a1a is 2, 3, or 4; each L A1a is CH 2 ; a1b is 0, 1, or 2; each L A1b is CH2; and L A4 is selected from the group consisting of: , , , wherein cc represents the point of attachment to L A1b . In some embodiments of Formula (I-G), the moiety is selected from the groups depicted in Table (L-I-G): wherein bb represents the point of attachment to Ring C. In some embodiments, the compounds of Formula (I-H) are compounds of Formula (I- Formula (I-H-1) or pharmaceutically acceptable salts thereof, wherein: R 1 is C(O)OH or C(O)OC1-6 alkyl; R a1 is C1-3 alkyl optionally substituted with 1-3 F; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and m8 is 0, 1, or 2. In some embodiments, L is (L-4); and the compounds of Formula (I) are compounds of Formula (I-H-2): Formula (I-H-2) or pharmaceutically acceptable salts thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; m8 is 0, 1, or 2; and Ring , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C1-3 alkyl optionally substituted with 1-3 F; and R aN is C 1-3 alkyl. In some embodiments, L is (L-4); and the compounds of Formula (I) are compounds of Formula (I-H-3): Formula (I-H-3) or pharmaceutically acceptable salts thereof, wherein: R 1 is C(O)OH or C(O)OC1-6 alkyl; R a1 is C 1-3 alkyl optionally substituted with 1-3 F; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; m8 is 0, 1, or 2; and Ring , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 F; and R aN is C 1-3 alkyl. In some embodiments of Formula (I-H-1), (I-H-2), or (I-H-3): a3 is 1; L A3 is -O-; a1a is 0; and L A4 is selected from the group consisting of: C3-10 cycloalkylene optionally substituted with 1-3 R a ; and phenylene optionally substituted with 1-3 R a . In some embodiments of Formula (I-H-1), (I-H-2), or (I-H-3): a3 is 1; L A3 is -O-; a1a is 0; a1b is 3 or 4; each L A1b is independently selected from the group consisting of: -CH2- and -CH(Me)- , provided that 0-1 of L A1b is -CH(Me)-; and L A4 is selected from the group consisting of: . In some embodiments of Formula (I-H-1), (I-H-2), or (I-H-3): a3 is 1; L A3 is -O-; a1a is 2, 3, or 4; and L A4 is 4-10 membered heterocyclylene optionally substituted with 1-3 R a . In some embodiments of Formula (I-H-1), (I-H-2), or (I-H-3): a3 is 1; L A3 is -O-; a1a is 2, 3, or 4; each L A1a is CH 2 ; a1b is 0, 1, or 2; each L A1b is CH2; and L A4 is 4-8 membered nitrogen-containing heterocyclylene optionally substituted with 1-3 R a . In some embodiments of Formula (I-H-1), (I-H-2), or (I-H-3), the moiety is selected from the groups depicted in Table (L-I-H): wherein bb represents the point of attachment to Ring C. In some embodiments, the compounds are selected from the group consisting of the compounds in Table C1, or pharmaceutically acceptable salts thereof. Table C1 209a 210 210a 210b 211 211a Note: for compounds in Table C1, when a stereogenic center is denoted with “or1” in a structural formula, the stereogenic center has been resolved but the configuration at the stereogenic center has not been determined. For example, the structure represents (R)-2-methyl-3-(1-methylpiperidin-4-yl)propan-1-ol or (S)-2-methyl-3-(1- methylpiperidin-4-yl)propan-1-ol. For example, the structure represents (R)-2-methyl-3-(1-methylpiperidin-4-yl)propan-1-ol or (S)-2-methyl-3-(1-methylpiperidin-4- yl)propan-1-ol. Exemplary compounds of Formula (I) or (II) also include those depicted in Table C1 of U.S. Provisional Application Serial No.63/457,997, filed April 7, 2023; Table C1 of U.S. Provisional Application Serial No. 63/454,545, filed March 24, 2023; Table C1 of U.S. Provisional Application Serial No. 63/449,756, filed March 3, 2023; Table C1 of U.S. Provisional Application Serial No. 63/429,862, filed December 2, 2022; Table C1 of U.S. Provisional Application Serial No.63/398,783, filed August 17, 2022; and Table C1 of U.S. Provisional Application Serial No. 63/339,253, filed May 6, 2022; or pharmaceutically acceptable salts thereof, wherein each Table C1 is incorporated herein by reference in its entirety. Exemplary compounds of Formula (I-A) (e.g., Formula (I-A-1), (I-A-2), or (I-A-3)) include compounds 101, 102, 103, 104, 105, 106, 107, 116, 116a, 117, 117a, 120, 121, 122, 124, 124a, 125, 125a, 126, 127, 128, 129, 130, 131, 131a, 132, 134, 156, 158, 159, 159a, 160, 160a, 160b, 162, 162a, 162b, 164, 164a, 164b, 166, 167, 167a, 167b, 169, 169a, 169b, 170, 173, 173a, 173b, 177, 177a, 180, 180a, 180b, 181, 186, 186a, 186b, 187, 187a, 187b, 193, 193a, 193b, 196, 197, 197a, 197b, 202, 202a, 202b, 203, 207, 207a, 207b, 209, 209a, 210, 210a, 210b, 213, 216, 216a, 216b, 218, 218a, 218b, 219, 219a, 219b, 226, 229, 231, 231a, 231b, 240, 240a, 240b, 243, 245, 245a, 245b, 246, 246a, 246b, 262, 262a, 262b, 263, 263a, 263b, 267, 268, 268a, 268b, 272, 272a, 272b, 277, 280, 286, 289, 289a, 289b, 292, 295, 302, 309, 313, 317, 343, 343a, 343b, 343c, 343d, and 357, as depicted in Table C1, or pharmaceutically acceptable salts thereof. Exemplary compounds of Formula (I-B) (e.g., Formula (I-B-1), (I-B-2), or (I-B-3)) include compounds 119, 139, 184, 236, 242, 264, 265, 266, 266a, 266b, 271, 271a, 276, 276a, 276b, 294, 344, 344a, 344b, 344c, 344d, 355, and 356, as depicted in Table C1, or pharmaceutically acceptable salts thereof. Exemplary compounds of Formula (I-C) include compounds 123, 133, 136, 165, and 172 as depicted in Table C1, or pharmaceutically acceptable salts thereof. Exemplary compounds of Formula (I-E) (e.g., Formula (I-E-1), (I-E-2), or (I-E-3)) include compounds 147, 147a, 147b, 148, 148a, 148b, 149, 150, 161, 161a, 163, 168, 168a, 175, 188, 195, 200, 201, 204, 204a, 208, 211, 211a, 212, 212a, 215, 215a, 220, 225, 230, 232, 233, 234, 235, 241, 273, 273a, 273b, 274, 274a, 274b, 275, 275a, 275b, 278, 278a, 278b, 281, 281a, 281b, 282, 282a, 282b, 283, 283a, 283b, 284, 284a, 284b, 293, 293a, 296, 296a, 297, 297a, 297b, 300, 300a, 300b, 301, 301a, 301b, 307, 307a, 307b, 308, 308a, 308b, 310, 310a, 316, 316a, 318, 318a, 319, 319a, 322, 322a, 323, 323a, 324, 324a, 325, 325a, 328, 328a, 333, 333a, 334, 334a, 335, 335a, 336, 336a, 339, 339a, 339b, 340, 340a, 341, 341a, 342, 342a, 345, 345a, 346, 346a, 346b, 347, 347a, 347b, 349, 349a, 349b, 350, 350a, 354, 354a, and 354b as depicted in Table C1, or pharmaceutically acceptable salts thereof. Exemplary compounds of Formula (I-Ea) (e.g., Formula (I-Ea-1), (I-Ea-2), or (I-Ea- 3)) include compounds 189, 192, 198, 221, 223, 228, 247, 257, 258, 261, 279, 285, 287, 288, 306, 311, 311a, 311b, 312, 353, 353a, and 353b as depicted in Table C1, or pharmaceutically acceptable salts thereof. Exemplary compounds of Formula (I-Eb) (e.g., Formula (I-Eb-1), (I-Eb-2), or (I-Eb- 3)) include compounds 248, 249, 255, 256, 269, 269a, 270, 270a, 298, 298a, 299, 299a, 320, 320a, 320b, 321, 321a, 321b, 331, 331a, 331b, 332, 332a, 332b, 351, 351a, 351b, 352, 352a, and 352b as depicted in Table C1, or pharmaceutically acceptable salts thereof. Exemplary compounds of Formula (I-F) (e.g., Formula (I-F-1), (I-F-2), or (I-F-3)) include compounds 138, 138a, 138b, 140, 141, 142, 143, 144, 145, 146, 146a, 146b, 151, 171, 171a, 176, 176a, 178, 179, 179a, 182, 182a, 185, 185a, 191, 214, 214a, 227, 227a, 239, 239a, and 244 as depicted in Table C1, or pharmaceutically acceptable salts thereof. Exemplary compounds of Formula (I-G) include compounds 190, 199, 250, 250a, 254, and 254a as depicted in Table C1, or pharmaceutically acceptable salts thereof. Exemplary compounds of Formula (I-H) (e.g., Formula (I-H-1), (I-H-2), or (I-H-3)) include compounds 152, 153, 154, 154a, 183, 183a, 251, 251a, 252, and 252a as depicted in Table C1, or pharmaceutically acceptable salts thereof. In some embodiments, the compounds of Formula (I) or (II) are selected from the group consisting of the compounds in Table C2, or pharmaceutically acceptable salts thereof. Table C2

Certain examples of Formula (I) or (II) compounds were synthesized using methods involving resolution of stereoisomeric mixture(s) (e.g., SFC separation of stereoisomers). In Table C1, the resolved stereogenic centers in these compounds are labelled with the “or1” or “or2” enhanced stereochemical notations. In some instances, the stereoisomeric resolutions were performed during the last step of the synthesis, thereby providing the individual stereoisomers of the Formula (I) or (II) compounds. Alternatively, in some other instances, the resolutions were performed on an intermediate or starting material, wherein each of the constituent stereoisomers of the intermediate or starting material could be separately subjected to the subsequent steps of the synthesis to provide the respective Formula (I) or (II) compounds as separate stereoisomers. Methods of resolution and correlation between resolved intermediates and Formula (I) or (II) compounds are disclosed in the examples herein and in Table P1. A person of ordinary skill in the art would understand that, under either approach for stereoisomeric resolution, stereoisomers having both (R)- and (S)-configurations at a resolved stereogenic center are provided. See Table C3, wherein Table C1 compounds whose stereoisomers contain the or1 notations are provided in non-stereogenic form, followed by the respective stereoisomers having the (R)- and (S)-configurations. Table C3 In some embodiments, the compounds of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A- 2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E- 2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H- 2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or pharmaceutically acceptable salts thereof, reduce cell viability in a cell line expressing a BCL-XL protein with an EC50 of less than 1 µM (e.g., less than 750 nM, less than 500 nM, or less than 200 nM). In some embodiments, the compounds reduce cell viability in a cell line expressing the BCL-XL protein with an EC50 of less than 200 nM (e.g., less than 150 nM, less than 200 nM, less than 100 nM, less than 10 nM, less than 1 nM). For example, the compounds can reduce cell viability in a cell line expressing the BCL-X L protein with an EC 50 of between 0.1 nM to 100 nM, between 0.1 nM to 50 nM, between 1 nM to 50 nM, between 1 nM to 20 nM, or between 0.1 nM to 1 nM. In some embodiments, the compounds of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A- 2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E- 2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H- 2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or pharmaceutically acceptable salts thereof, induce degradation of a BCL-X L protein in a cell line expressing the BCL-X L protein with a DC50 of less than 1 µM (e.g., less than 750 nM, less than 500 nM, or less than 200 nM). In some embodiments, the compounds of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A- 3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E- 3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H- 3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, induce degradation of a BCL-XL protein in a cell line expressing the BCL-XL protein with a DC50 of less than 200 nM (e.g., less than 150 nM, less than 200 nM, less than 100 nM, less than 10 nM, less than 1 nM). For example, the compounds can induce degradation of a BCL-X L protein in a cell line expressing the BCL-X L protein with a DC 50 of between 0.1 nM to 100 nM, between 0.1 nM to 50 nM, between 1 nM to 50 nM, between 1 nM to 20 nM, or between 0.1 nM to 1 nM. In some embodiments, the compounds of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A- 2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E- 2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H- 2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or pharmaceutically acceptable salts thereof, induce degradation of a BCL-XL protein in a cell line expressing the BCL-XL protein with a Y min of less than 70% (e.g., less than 50%, less than 30%, less than 20%, or less than 10%). In some embodiments, the compounds of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A- 3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E- 3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H- 3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, induce degradation of a BCL-X L protein in a cell line expressing the BCL-X L protein with a Y min of less than 50% (e.g., less than 40%, less than 30%, less than 20%, less than 10%, or less than 5%). In some embodiments, the compounds of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, induce degradation of a BCL-X L protein in a cell line expressing the BCL-X L protein with a Y min of less than 30% (e.g., less than 25%, less than 20%, less than 15%, less than 10%, or less than 5%). For example, the compounds can induce degradation of a BCL-XL protein in a cell line expressing the BCL-X L protein with a Y min of about 1% to about 70% (e.g., about 5% to about 50% or about 10% to about 30%). Also provided herein is a BCL-XL protein non-covalently bound with a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof. Also provided herein is a ternary complex comprising a BCL-X L protein, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I- F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, and a CRBN protein, or a portion thereof. Chemical definitions The term “halo” refers to fluoro (F), chloro (Cl), bromo (Br), or iodo (I). The term “oxo” refers to a divalent doubly bonded oxygen atom (i.e., “=O”). As used herein, oxo groups are attached to carbon atoms to form carbonyls. The term “alkyl” refers to a saturated acyclic hydrocarbon radical that may be a straight chain or branched chain, containing the indicated number of carbon atoms. For example, C 1-10 indicates that the group may have from 1 to 10 (inclusive) carbon atoms in it. Alkyl groups can either be unsubstituted or substituted with one or more substituents. Non-limiting examples include methyl, ethyl, iso-propyl, tert-butyl, n-hexyl. The term “saturated” as used in this context means only single bonds present between constituent carbon atoms and other available valences occupied by hydrogen and/or other substituents as defined herein. The term “haloalkyl” refers to an alkyl, in which one or more hydrogen atoms is/are replaced with an independently selected halo (e.g., -CF 3 , -CHF 2 , or -CH 2 F). The term “alkoxy” refers to an -O-alkyl radical (e.g., -OCH3). The term “alkylene” refers to a divalent alkyl (e.g., -CH2-). Similarly, terms such as “cycloalkylene” and “heterocyclylene” refer to divalent cycloalkyl and heterocyclyl respectively. For avoidance of doubt, in “cycloalkylene” and “heterocyclylene”, the two radicals can be on the same ring carbon atom (e.g., a geminal diradical such different ring atoms (e.g., ring carbon and/or nitrogen atoms (e.g., vicinal ring carbon and/or nitrogen atoms)) The term “alkenyl” refers to an acyclic hydrocarbon chain that may be a straight chain or branched chain having one or more carbon-carbon double bonds. The alkenyl moiety contains the indicated number of carbon atoms. For example, C2-6 indicates that the group may have from 2 to 6 (inclusive) carbon atoms in it. Alkenyl groups can either be unsubstituted or substituted with one or more substituents. The term “alkynyl” refers to an acyclic hydrocarbon chain that may be a straight chain or branched chain having one or more carbon-carbon triple bonds. The alkynyl moiety contains the indicated number of carbon atoms. For example, C 2-6 indicates that the group may have from 2 to 6 (inclusive) carbon atoms in it. Alkynyl groups can either be unsubstituted or substituted with one or more substituents. The term “aryl” refers to a 6-20 carbon mono-, bi-, tri- or polycyclic group wherein at least one ring in the system is aromatic (e.g., 6-carbon monocyclic, 10-carbon bicyclic, or 14- carbon tricyclic aromatic ring system); and wherein 0, 1, 2, 3, or 4 atoms of each ring may be substituted by a substituent. Examples of aryl groups include phenyl, naphthyl, tetrahydronaphthyl, and the like. The term “cycloalkyl” as used herein refers to mono-, bi-, tri-, or polycyclic (e.g., fused, bridged, or spirocyclic bi-, tri-, or polycyclic) saturated or partially unsaturated hydrocarbon groups having, e.g., 3 to 20 ring carbons, preferably 3 to 15 ring carbons, and more preferably 3 to 12 ring carbons or 3-10 ring carbons or 3-6 ring carbons, wherein the cycloalkyl group may be optionally substituted. The term “saturated” as used in this context means only single bonds present between constituent carbon atoms. Examples of saturated cycloalkyl groups include, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Partially unsaturated cycloalkyl may have any degree of unsaturation provided that one or more double bonds is present in the cycloalkyl, none of the rings in the ring system are aromatic, and the partially unsaturated cycloalkyl group is not fully saturated overall. Examples of partially unsaturated cycloalkyl include, without limitation, cyclopentenyl, cyclohexenyl, cycloheptenyl, and cyclooctenyl. Non-limiting examples of fused/bridged cycloalkyl includes: bicyclo[1.1.0]butyl, bicyclo[2.1.0]pentyl, bicyclo[1.1.1]pentyl, bicyclo[3.1.0]hexyl, bicyclo[2.1.1]hexyl, bicyclo[3.2.0]heptyl, bicyclo[4.1.0]heptyl, bicyclo[2.2.1]heptyl, bicyclo[3.1.1]heptyl, bicyclo[4.2.0]octyl, bicyclo[3.2.1]octyl, bicyclo[2.2.2]octyl, and the like. Non-limiting examples of spirocyclic cycloalkyls include spiro[2.2]pentyl, spiro[2.5]octyl, spiro[3.5]nonyl, spiro[3.5]nonyl, spiro[3.5]nonyl, spiro[4.4]nonyl, spiro[2.6]nonyl, spiro[4.5]decyl, spiro[3.6]decyl, spiro[5.5]undecyl, and the like. The term “heteroaryl”, as used herein, means a mono-, bi-, tri- or polycyclic group having 5 to 20 ring atoms, alternatively 5, 6, 9, 10, or 15 ring atoms; wherein at least one ring in the system contains one or more heteroatoms independently selected from the group consisting of N, O, S (inclusive of oxidized forms such as: and P (inclusive of oxidized forms such as: (e.g., N, O, and S (inclusive of oxidized forms such as: and at least one ring in the system is aromatic (but does not have to be a ring which contains a heteroatom, e.g. tetrahydroisoquinolinyl, e.g., tetrahydroquinolinyl). In some embodiments, heteroaryl groups contain 1-4 (e.g., 1, 2, or 3) ring heteroatoms each independently selected from the group consisting of N, O, and S (inclusive of oxidized forms such as: Heteroaryl groups can either be unsubstituted or substituted with one or more substituents. Examples of heteroaryl include thienyl, pyridinyl, furyl, oxazolyl, oxadiazolyl, pyrrolyl, imidazolyl, triazolyl, thiodiazolyl, pyrazolyl, isoxazolyl, thiadiazolyl, pyranyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, thiazolyl benzothienyl, benzoxadiazolyl, benzofuranyl, benzimidazolyl, benzotriazolyl, cinnolinyl, indazolyl, indolyl, isoquinolinyl, isothiazolyl, naphthyridinyl, purinyl, thienopyridinyl, pyrido[2,3-d]pyrimidinyl, pyrrolo[2,3-b]pyridinyl, quinazolinyl, quinolinyl, thieno[2,3-c]pyridinyl, pyrazolo[3,4- b]pyridinyl, pyrazolo[3,4-c]pyridinyl, pyrazolo[4,3-c]pyridinyl, pyrazolo[4,3-b]pyridinyl, tetrazolyl, chromanyl, 2,3-dihydrobenzo[b][1,4]dioxinyl, benzo[d][1,3]dioxolyl, 2,3- dihydrobenzofuranyl, tetrahydroquinolinyl, 2,3-dihydrobenzo[b][1,4]oxathiinyl, isoindolinyl, and others. In some embodiments, the heteroaryl is selected from thienyl, pyridinyl, furyl, pyrazolyl, imidazolyl, isoindolinyl, pyranyl, pyrazinyl, and pyrimidinyl. For purposes of clarification, heteroaryl also includes aromatic lactams, aromatic cyclic ureas, or vinylogous analogs thereof, in which each ring nitrogen adjacent to a carbonyl is tertiary (i.e., all three valences are occupied by non-hydrogen substituents), such as one or more of pyridone (e.g., imidazolone (e.g., ), wherein each ring nitrogen adjacent to a carbonyl is tertiary (i.e., the oxo group (i.e., “=O”) herein is a constituent part of the heteroaryl ring). The term “heterocyclyl” refers to a mono-, bi-, tri-, or polycyclic (e.g., fused, bridged, or spirocyclic bi-, tri-, or polycyclic) saturated or partially unsaturated ring system with 3-15 ring atoms (e.g., 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-15 membered tricyclic ring system) having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1- 9 heteroatoms if tricyclic or polycyclic, said heteroatoms selected from O, N, S (inclusive of oxidized forms such as: and P (inclusive of oxidized forms such as: ) (e.g., O, N, and S (inclusive of oxidized forms such as: )) (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, S, or P if monocyclic, bicyclic, or tricyclic, respectively), wherein 0, 1, 2 or 3 atoms of each ring may be substituted by a substituent. In some embodiments, heterocyclyl groups contain 1-4 (e.g., 1, 2, or 3) ring heteroatoms each independently selected from the group consisting of N, O, and S (inclusive of oxidized forms such as: The term “saturated” as used in this context means only single bonds present between constituent ring atoms and other available valences occupied by hydrogen and/or other substituents as defined herein. Examples of saturated heterocyclyl groups include piperazinyl, pyrrolidinyl, dioxanyl, morpholinyl, tetrahydrofuranyl, and the like. Partially unsaturated heterocyclyl groups may have any degree of unsaturation provided that one or more double bonds is present in the heterocyclyl, none of the rings in the ring system are aromatic, and the partially unsaturated heterocyclyl group is not fully saturated overall. Examples of partially unsaturated heterocyclyl groups include, without limitation, tetrahydropyridyl, dihydropyrazinyl, dihydropyridyl, dihydropyrrolyl, dihydrofuranyl, dihydrothiophenyl. Non-limiting examples of fused/bridged heteorocyclyl includes: 2- azabicyclo[1.1.0]butyl, 2-azabicyclo[2.1.0]pentyl, 2-azabicyclo[1.1.1]pentyl, 3- azabicyclo[3.1.0]hexyl, 5-azabicyclo[2.1.1]hexyl, 3-azabicyclo[3.2.0]heptyl, octahydrocyclopenta[c]pyrrole, 3-azabicyclo[4.1.0]heptyl, 7-azabicyclo[2.2.1]heptyl, 6- azabicyclo[3.1.1]heptyl, 7-azabicyclo[4.2.0]octyl, 2-azabicyclo[2.2.2]octyl, 3- azabicyclo[3.2.1]octyl, 2-oxabicyclo[1.1.0]butyl, 2-oxabicyclo[2.1.0]pentyl, 2- oxabicyclo[1.1.1]pentyl, 3-oxabicyclo[3.1.0]hexyl, 5-oxabicyclo[2.1.1]hexyl, 3- oxabicyclo[3.2.0]heptyl, 3-oxabicyclo[4.1.0]heptyl, 7-oxabicyclo[2.2.1]heptyl, 6- oxabicyclo[3.1.1]heptyl, 7-oxabicyclo[4.2.0]octyl, 2-oxabicyclo[2.2.2]octyl, oxabicyclo[3.2.1]octyl, and the like. Non-limiting examples of spirocyclic heterocyclyls include 2-azaspiro[2.2]pentyl, 4-azaspiro[2.5]octyl, 1-azaspiro[3.5]nonyl, 2- azaspiro[3.5]nonyl, 7-azaspiro[3.5]nonyl, 2-azaspiro[4.4]nonyl, 6-azaspiro[2.6]nonyl, 1,7- diazaspiro[4.5]decyl, 7-azaspiro[4.5]decyl 2,5-diazaspiro[3.6]decyl, 3-azaspiro[5.5]undecyl, 2-oxaspiro[2.2]pentyl, 4-oxaspiro[2.5]octyl, 1-oxaspiro[3.5]nonyl, 2-oxaspiro[3.5]nonyl, 7- oxaspiro[3.5]nonyl, 2-oxaspiro[4.4]nonyl, 6-oxaspiro[2.6]nonyl, 1,7-dioxaspiro[4.5]decyl, 2,5-dioxaspiro[3.6]decyl, 1-oxaspiro[5.5]undecyl, 3-oxaspiro[5.5]undecyl, 3-oxa-9- azaspiro[5.5]undecyl and the like. A nitrogen-containing heterocyclyl as used herein refers to a heterocyclyl having 1-2 ring nitrogen atoms and 0-2 additional ring heteroatoms selected from the group consisting of O and S (inclusive of oxidized such as: or ). The nitrogen-containing heterocyclyl can be monocyclic, bicyclic, or polycyclic as defined elsewhere herein. Examples of monocyclic nitrogen-containing heterocyclyl include azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, and the like. Examples of bicyclic nitrogen-containing heterocyclyl include 7-azaspiro[3.5]nonyl, 1,7-diazaspiro[4.5]decyl, 3-oxa-7,9-diazabicyclo[3.3.1]nonanyl, 2,6-diazaspiro[3.3]heptanyl, and the like. As used herein, when a ring is described as being “partially unsaturated”, it means said ring has one or more additional degrees of unsaturation (in addition to the degree of unsaturation attributed to the ring itself; e.g., one or more double or triple bonds between constituent ring atoms), provided that the ring is not aromatic. Examples of such rings include: cyclopentene, cyclohexene, cycloheptene, dihydropyridine, tetrahydropyridine, dihydropyrrole, dihydrofuran, dihydrothiophene, and the like. For the avoidance of doubt, and unless otherwise specified, for rings and cyclic groups (e.g., aryl, heteroaryl, heterocyclyl, heterocycloalkenyl, cycloalkenyl, cycloalkyl, and the like described herein) containing a sufficient number of ring atoms to form bicyclic or higher order ring systems (e.g., tricyclic, polycyclic ring systems), it is understood that such rings and cyclic groups encompass those having fused rings, including those in which the points of fusion are located (i) on adjacent ring atoms (e.g., [x.x.0] ring systems, in which 0 represents a zero atom bridge (e.g., )); (ii) a single ring atom (spiro-fused ring systems) , (iii) a contiguous array of ring atoms (bridged ring systems In addition, atoms making up the compounds of the present embodiments are intended to include all isotopic forms of such atoms. Isotopes, as used herein, include those atoms having the same atomic number but different mass numbers. By way of general example and without limitation, isotopes of hydrogen include tritium and deuterium, and isotopes of carbon include 13 C and 14 C. In addition, the compounds generically or specifically disclosed herein are intended to include all tautomeric forms. Thus, by way of example, a compound containing the moiety: encompasses the tautomeric form containing the moiety: . Similarly, a pyridinyl or pyrimidinyl moiety that is described to be optionally substituted with hydroxyl encompasses pyridone or pyrimidone tautomeric forms. The compounds provided herein may encompass various stereochemical forms. The compounds also encompass diastereomers as well as optical isomers, e.g., mixtures of enantiomers including racemic mixtures, as well as individual enantiomers and diastereomers, which arise as a consequence of structural asymmetry in certain compounds. Unless otherwise indicated, when a disclosed compound is named or depicted by a structure without specifying the stereochemistry and has one or more chiral centers, it is understood to represent all possible stereoisomers of the compound. Methods of Treatment Indications Provided herein are methods for inducing degradation of a BCL-X L protein. For example, provided herein are compounds capable of inducing degradation of a BCL-XL protein useful for treating or preventing cancers. See, e.g., Guo, et al. Aging (Albany NY) 13.15 (2021): 19750; Park, et al. Proceedings of the National Academy of Sciences 112.40 (2015): 12492- 12497; Zhang, et al. Molecular Cancer 14.1 (2015): 1-9; Beroukhim, et al. Nature 463.7283 (2010): 899-905. Additional methods of evaluating the binding of a compound to BCL-XL protein or the inhibition of a BCL-X L protein are described in, for example, U.S. Patent Publication Nos.2007/027135; 2010/305122, and 2013/096120. It will be understood that the effect of protein degradation typically increases over time, though the appearance of degradation (e.g., as expressed by the percentage degradation compared to a control, or the parameters Ymin, DC50, and/or Dmax) is affected by the resynthesis rate of the protein. It is common in the art to examine degradation after a specified period of time, such as 6 hours, 12 hours, 18 hours, 1 day, 2 days, 3 days, or more. For example, degradation can be expressed as the percent degradation after 24 hours. Exemplary assays for validating the degradation-inducing mechanism of a compound as provided herein are known in the art and are described, for example, in International Publication No. WO 2019/144117 and Wu, et al. Nature Structural & Molecular Biology 27.7 (2020): 605-614. Degradation assays can be used to quantify both on- and off-target degradation- inducing effects of compounds, such as those provided herein. Exemplary assays include, quantitative immunoblotting, other immunoassays (e.g., MesoScale Discovery (MSD) immunoassays), homogenous time resolved florescence (HTRF), and HiBiT. In some embodiments, cells can be contacted with a compound of Formula (I) (e.g., (I-A) (e.g., (I-A- 1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E- 1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I- Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, incubated, and then the lysate can be prepared for gel electrophoresis (e.g., SDS-PAGE), followed by immunoblotting and quantification compared to a control (e.g., a DMSO-treated control). As another example, a cell line can be engineered to express a HiBiT-tagged BCL-X L protein, and the amount of fluorescence observed when the complementary LgBiT peptide is added can be compared between cells treated with a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb- 1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, and a control (e.g., a DMSO-treated control). See, for instance, Example B1 and Example B2. See also, e.g., International Publication Nos. WO 2020/163823 and WO 2019/144117. In some embodiments, off-target degradation inducing effects can be assessed for the proteins Eukaryotic peptide chain release factor GTP-binding subunit ERF3A (GSPT1), Ikaros (IKZF1), Helios (IKZF2), Aiolos (IKZF3), and/or casein kinase I isoform alpha (CK1α). See also the assays described in International Publication Nos. WO 2023/044046; WO 2022/169780; WO 2021/222114; WO 2021/146536; WO 2021/078301; WO 2021/007307; WO 2020/163823; WO 2019/144117; WO 2017/184995, and Khan, et al. Nature Medicine 25.12 (2019): 1938-1947; Balachander, et al. Clinical Cancer Research 26.24 (2020): 6535- 6549. Binding affinity of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I- A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I- E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb- 3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I- H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, as provided herein to BCL-XL can be determined by, for example, a binding IC50 or Ki value (e.g., using a competition assay), or by a K D value (e.g., using a biophysical assay). A compound with a lower binding IC50 value, as determined under substantially similar conditions, is a more potent binder relative to a compound with a higher binding IC50 value. A compound with a lower binding K i value, as determined under substantially similar conditions, is a more potent binder relative to a compound with a higher binding K i value. Similarly, a compound with a lower KD value, as determined under substantially similar conditions, is a more potent binder relative to a compound with a higher K D value. For example, a binding IC 50 value can be determined in a fluorescence polarization assay using a fluorescently labeled BH3-only peptide (e.g., BAD or BAX) as the competitor. As another example, a binding Ki value can be determined using a time resolved-fluorescence resonance energy transfer (TR-FRET) assay using a fluorescently labeled BH3-only peptide (e.g., BAK) and a fluorescently labeled antibody that binds to BCL- XL, where the fluorophores are a FRET pair, and using a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, as provided herein as a competitor for the BH3-only peptide. See, e.g., U.S. Patent Publication Nos.2007/0027135, 2010/305122, and 2013/096120. The ability of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A- 3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E- 3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H- 3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, as provided herein to inhibit BCL-X L can be determined using an IC 50 value. A compound with a lower IC50 value, as determined under substantially similar conditions, is a more potent inhibitor relative to a compound with a higher IC50 value. One way that inhibition of BCL-XL can be measured is measuring the interruption of the formation of a complex of BCL-X L with a BH3- only peptide (e.g., BIM). For example, an electrochemiluminescence-based sandwich ELISA assay (e.g., a Meso Scale Discovery (MSD)-ELISA assay) can be used. See, for example, Phillips, D. C., et al. Blood Cancer Journal 5.11 (2015): e368-e368 and Xiao, Yu, et al. Molecular Cancer Therapeutics 14.8 (2015): 1837-1847. In such assays, cells expressing BCL- XL can be incubated with a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I- A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I- E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb- 3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I- H-3))) or Formula (II) (e.g., (II-a)) for a period of time, lysed, and then evaluated in the assay. A tagged anti-BCL-X L antibody (e.g., a biotin-tagged anti-BCL-X L antibody) can be immobilized on an assay plate (e.g., a streptavidin assay plate), and then the lysate can be applied to pull down BCL-XL. An anti-BIM antibody (e.g., a rabbit anti-BIM antibody) can be introduced, followed by addition of a detection antibody (e.g., a sulfo-tagged goat anti-rabbit antibody) and measurement of the detection antibody. As another example, interruption of the formation of a complex of BCL-XL with a BH3-only peptide (e.g., BIM) can be measured using a mammalian two-hybrid assay. In such assays, a plasmid encoding the ‘bait’ and ‘prey’ fusion proteins (e.g., the DNA binding domain of GAL4 fused to BCL-X L and the transcriptional activation domain of VP16 fused to BIM) can be introduced to cells (e.g., HeLa cells) stably expressing a GAL4-luciferase reporter. A compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb- 2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I- H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)) can be added to the cells in culture, incubated, and the luciferase activity can be measured. See, for example, Souers, Andrew J., et al. Nature Medicine 19.2 (2013): 202-208. Potency of degradation by a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A- 2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E- 2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H- 2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, as provided herein can be determined by DC50 value. As used herein, DC50 refers to the concentration of the compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I- F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)) that results in a 50% decrease in the concentration of a protein (e.g., BCL-X L protein) in a cell compared to the concentration of the protein before the cell is contacted with the compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I- B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I- Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or compared to the concentration of the protein in a cell not contacted with the compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I- F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)). A compound with a lower DC 50 value, as determined under substantially similar conditions, is a more efficient inducer of degradation relative to a compound with a higher DC50 value. In some embodiments, a DC50 value can be determined (e.g., using HiBiT detection) in vitro or in vivo (e.g., in tumor cells (e.g., cell lines such as MOLT4, RS4;11, NCI- H146, EJM, HEK293T, HT1080, and/or H929) expressing a BCL-XL protein). Potency of degradation by a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A- 2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E- 2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H- 2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, as provided herein can be determined by EC 50 value. As used herein, EC 50 refers to the concentration of the compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I- F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)) that results in a 50% decrease in the concentration of a protein (e.g., BCL-X L protein) relative to the trough concentration of the protein in a cell, when compared to the concentration of the protein before the cell is contacted with the compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or compared to the concentration of the protein in a cell not contacted with the compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)). A compound with a lower EC50 value, as determined under substantially similar conditions, is a more potent compound relative to a compound with a higher EC50 value. In some embodiments, an EC 50 value can be determined (e.g., using HiBiT detection) in vitro or in vivo (e.g., in tumor cells (e.g., cell lines such as MOLT4, RS4;11, NCI-H146, EJM, HEK293T, HT1080, and/or H929) expressing a BCL-XL protein). Potency of degradation by a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A- 2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E- 2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H- 2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, as provided herein can be determined by Ymin value. As used herein, Ymin refers to the ratio of trough concentration of a protein (e.g., BCL-XL protein) in a cell compared to the concentration of the protein before the cell is contacted with the compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb- 1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or compared to the concentration of the protein in a cell not contacted with the compound of Formula (I) (e.g., (I-A) (e.g., (I-A- 1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E- 1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I- Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), expressed as a percentage. As used herein, D max is 1-Y min . Y min can be measured by a HiBiT assay as described in Example B1. A compound with a lower Ymin value, as determined under substantially similar conditions, is a more potent inducer of degradation relative to a compound with a higher Ymin value. A compound with a lower Y min value, as determined under substantially similar conditions, is a more potent compound relative to a compound with a higher Ymin value. In some embodiments, a Ymin value can be determined (e.g., using HiBiT detection) in vitro or in vivo (e.g., in tumor cells (e.g., cell lines such as MOLT4, RS4;11, NCI-H146, EJM, HEK293T, HT1080, and/or H929) expressing a BCL-X L protein). An exemplary assay for determining the potency of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, includes measuring the effect of the compound of Formula (I) (e.g., (I- A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I- E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, on cell proliferation and/or viability. Cell proliferation assays can be performed in a number of formats, including 2D and 3D. Similarly, a cell proliferation assay can be performed with any appropriate cell line, including, for example, MOLT4, RS4;11, NCI-H146, EJM, HEK293T, HT1080, and/or H929. As an illustrative example, a 3D cell proliferation assay can include growing cells in a 3D medium, contacting the cells with a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea- 1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I- F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, measuring the cellular proliferation using an appropriate reagent (e.g., CELLTITERGLO® 3D), and then comparing the signal from an experiment with a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I- B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I- Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, to the signal from a control experiment (e.g., lacking the compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A- 2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E- 2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H- 2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof). As another illustrative example, a 2D cell proliferation assay can include plating cells onto a growth surface, optionally letting the cells grow for a period of time, contacting the cells with a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea- 1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I- F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, measuring the cellular proliferation using an appropriate reagent (e.g., CELLTITERGLO®), and then comparing the signal from an experiment with compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I- B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I- Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, to the signal from a control experiment (e.g., lacking a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A- 2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E- 2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H- 2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof). Additional cell viability assays include MTT assays, which are colorimetric assays based on the reduction of the tetrazolium dye MTT (3-(4,5-dimethylthiazol-2-yl)-2,5- diphenyltetrazolium bromide) to the insoluble purple formazan, and other similar assays based on related tetrazolium salts. See, for instance, Example B3 and Example B4. A cell viability assay can be used to measure the effect of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I- Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I- Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, on cell death. For example, cells expressing BCL-X L protein (e.g., MOLT-4 cells) can be incubated with various concentrations of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, then exposed to a detection reagent (e.g., a CELLTITER-GLO® Cell Viability Assay kit) to determine cell viability. An exemplary assay for evaluating the affinity of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I- Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, includes using a competition assay with recombinant BCL-X L protein. For example, purified recombinant affinity-tagged (e.g., His-tagged) BCL-X L protein can be incubated with various concentrations of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I- F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, and a fixed concentration of affinity-tagged (e.g., biotin-tagged) BAD protein. After a period of incubation, FRET acceptor beads with a complementary affinity tag (e.g., His-acceptor beads) and FRET donor beads (e.g., streptavidin-tagged donor beads) can be added to the mixture, and a FRET reaction can be used to determine an inhibition constant of the compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or pharmaceutically acceptable salt thereof. For example, an AlphaLISA competitive assay can be performed. See, e.g., International Publication No. WO 2019/144117. An exemplary assay for determining the mechanism of cell death using of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I- F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, includes measuring the effect of the compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, on one or more markers of a mechanism of cell death (e.g., apoptosis). Exemplary markers of apoptosis include caspase induction (e.g., caspase 3/7 induction) and annexin V staining. Such assays may also be used as determinants of cell viability. For example, cells expressing BCL-X L protein (e.g., MOLT-4 cells) can be incubated with various concentrations of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, and relative caspase activity can be assessed using a luciferase substrate that is activated by caspase 3/7 (e.g., using the CASPASE-GLO® 3/7 assay). As another example, cells expressing BCL- XL protein (e.g., MOLT-4 cells) can be incubated with various concentrations of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I- F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, and relative caspase activity can be assessed using a dye that is coupled to an activated caspase motif (e.g., INCUCYTE® Caspase 3/7 Green Apoptosis Assay Reagent), followed by analysis using a platform for live cell imaging (e.g., an INCUCYTE® SX5 Live-Cell Analysis Instrument). As another example, cells expressing BCL-X L protein (e.g., MOLT-4 cells) can be incubated with various concentrations of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea- 1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I- F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, and annexin V positivity can be assessed using phosphatidylserine dye (e.g., an INCUCYTE® Annexin V dye), followed by analysis using a platform for live cell imaging (e.g., an INCUCYTE® SX5 Live-Cell Analysis Instrument). See, for instance, Example B5. As another example, the potency and/or efficacy of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, can be evaluated in an animal model, for example, a xenograft model (e.g., using an established cancer cell line such as MOLT4, HEL, TF1, F36P, OCI-M1, OCI- M2, SET-2, CMK, M07E, or UKE-1, or a patient-derived xenograft (PDX) model). For example, a PDX model can be run in immunodeficient mice (e.g., athymic nude, outbred homozygous (e.g., Crl:NU(NCr)-Foxn1 nu ) or Fox Chase SCID (CB17/Icr-Prkdc scid /IcrIcoCrl), mice). The mice can be female, 6-12 weeks old at tumor implantation and have access to food and water ad libitum. Approximately 70 mg of a tumor can be implanted subcutaneously in the right flank of each mouse. Following implantation, tumors can be measured weekly and once the tumor volumes reach 150-300 mm 3 , the mice can be randomized into treatment and control groups. In some embodiments, one or more experimental arms can be added to evaluate pharmacokinetics and/or pharmacodynamics. The mice can be treated with a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, (e.g., via IP or PO (oral) administration) and optionally an additional therapy or therapeutic agent (e.g., any of the additional therapies or therapeutic agents described herein). Throughout the study, health condition, body weight and tumor volumes of the mice can be recorded on a weekly basis. The mice can be sacrificed at 28 days or when the tumor reaches 1 cm 3 , and the tumors can be evaluated (e.g., by tumor weight, by tumor volume). At the end of each study, the Best Response can be calculated for each treatment arm. Best Response is defined as the minimum value of ∆Volumet for t ≥ 10 days. Best Responses between the control arm(s) and the treatment arm(s) can be compared to determine if the treatment(s) work better than the control(s). In some embodiments, tumor samples can also be collected at the end of each study and relevant proteins (e.g., BCL-XL, BCL-2, MCL-1, BIM, BAX, and/or BAK) can be measured to determine if the treatment might have a better protein modulation profile compared to a control. In some embodiments, tumor samples can also be collected at the end of each study and analyzed for signaling pathway activity (e.g., via phosphoERK levels). For pharmacokinetic and pharmacodynamic studies, tumor and/or blood samples from the mice can be obtained at the same or different time points than efficacy studies. For example, for pharmacokinetic and pharmacodynamic studies, tumor and/or blood samples from the mice can be obtained at Day 5, 6 hours post dosing, and relevant proteins can be measured in the tumor samples and pharmacokinetic studies can be performed on the blood samples or a portion thereof (e.g., plasma). In some embodiments, the PDX is a model of a myeloproliferative neoplasm (MPN) (e.g., CEL, CML, CNL, essential thrombocythemia (e.g., JAK2 mutant (e.g., JAK2 V617F mutant) essential thrombocythemia or JAK2 wild type essential thrombocythemia), polycythemia vera (e.g., JAK2 mutant (e.g., JAK2 V617F mutant) polycythemia vera or JAK2 wild type polycythemia vera), or myelofibrosis (e.g., primary myelofibrosis (e.g., JAK2 mutant (e.g., JAK2 V617F mutant) primary myelofibrosis or JAK2 wild type primary myelofibrosis), post-essential thrombocythemia myelofibrosis (e.g., JAK2 mutant (e.g., JAK2 V617F mutant) post-essential thrombocythemia myelofibrosis or JAK2 wild type post-essential thrombocythemia myelofibrosis), or post-polycythemia vera myelofibrosis (e.g., JAK2 mutant (e.g., JAK2 V617F mutant) post-polycythemia vera myelofibrosis or JAK2 wild type post- polycythemia vera myelofibrosis)))), a CRC (e.g., BRaf mutant CRC (e.g., Braf V600E CRC) or KRas mutant CRC (e.g., KRas G12C mutant CRC or KRas G12D CRC)), SCLC (e.g., ASCL1 subtype SCLC or NEUROD1 subtype SCLC), a NSCLC (e.g., BRaf mutant NSCLC (e.g., Braf V600E NSCLC), an EGFR mutant NSCLC (e.g., EGFR L858R NSCLC or EGFR exon 19 deletion NSCLC), MET mutant NSCLC (e.g., MET exon 14 deletion NSCLC, MET amplified NSCLC), a KRas mutant NSCLC (e.g., KRas G12C NSCLC)), a lung squamous cell carcinoma, a malignant pleural mesothelioma (e.g., a BAP1 mutant malignant pleural mesothelioma), a melanoma (e.g., Braf mutant melanoma (e.g., Braf V600E melanoma)), a breast cancer (e.g., HER2+ breast cancer (e.g., HER2+ breast cancer with ER expression, HER2+ breast cancer without ER expression), HER2 negative breast cancer (e.g., HER2 negative breast cancer with ER expression, HER2 negative breast cancer without ER expression), triple negative breast cancer, or HER2 low breast cancer), a lymphoma (e.g., a T cell lymphoma (e.g., anaplastic large T cell lymphoma, cutaneous T cell lymphoma, or peripheral T cell lymphoma), or a non-Hodgkin lymphoma (e.g., DLBCL, anaplastic large T cell lymphoma, cutaneous T cell lymphoma, or peripheral T cell lymphoma), a leukemia (e.g., T cell leukemia (e.g., a T-ALL (e.g., relapsed/refractory T-ALL)), post-MPN leukemia, M6- AML, M7-AML), a head and neck cancer, a pancreatic cancer, a bladder cancer, an ovarian cancer (e.g., BRCA1 mutant ovarian cancer or BRCA2 mutant ovarian cancer, HGSOC (e.g., BRCA1 mutant HGSOC or BRCA2 mutant HGSOC)), a cervical cancer, an intrahepatic cholangiocarcinoma, or a mesenchymal cancer (e.g., mesenchymal breast cancer or mesenchymal kidney cancer). See, e.g., Khan, et al. Nature Medicine 25.12 (2019): 1938-1947; Balachander, et al. Clinical Cancer Research 26.24 (2020): 6535-6549. The pharmacokinetic parameters of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A- 1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E- 1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I- Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, can be evaluated in an animal model, for instance, a mouse model, a rat model, a dog model, or a nonhuman primate (e.g., cynomolgus monkey) model. An exemplary assay includes the following. For example, pharmacokinetics (PK) studies can be conducted on animals (e.g., male or female CD-1 mice, Sprague Dawley rats, beagle dogs, or cynomolgus monkeys) by two delivery routes: intravenous (IV) injection and oral gavage (PO). Animals in both the IV and PO groups (e.g., n = 3) are allowed free access to food and water. A compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I- C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea- 3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I- G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, can be formulated in solution for the IV route and solution or suspension for the PO route. On the day of the experiment, the compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, can be administered via vein injection (e.g., at 1 mg/kg) for IV route or via oral gavage (e.g., at 5 to 10 mg/kg) for PO route. In some cases, the animals can be orally pre-dosed with a cytochrome P450 inhibitor (e.g., 1-aminobenzotriazole) prior to (e.g., 16 hours prior to) dosing the compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb- 2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I- H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof. Blood samples can be collected via serial bleeding (e.g., at 8 timepoints from 0.83 to 24 hours post dose). At each timepoint, blood can be collected (e.g., approximately 30 µL of blood/timepoint) in a K 2 EDTA tube via a vein (e.g., the saphenous vein). Blood samples can be put on wet ice and centrifuged (e.g., at 4600 RPM for 4 minutes) to obtain plasma samples. Plasma samples can be diluted (e.g., with an equal volume of pH 3.0 phosphate buffer) and submitted to LC-MS/MS for sample analysis. Pharmacokinetic parameters, including clearance (IV or PO, depending on the mode of dosing), area under the curve (AUC), and oral bioavailability (%F) can be calculated using a non-compartmental model. In some embodiments, the %F for a compound of Formula (I) (e.g., (I-A) (e.g., (I-A- 1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E- 1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I- Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, is at least 4%. In some embodiments, the %F for a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb- 1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, is at least 10%. In some embodiments, the %F for a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, is at least 20%. In some embodiments, the %F for a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, is at least 30%. In some embodiments, the %F for a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea- 1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I- F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, is at least 40%. In some embodiments, the %F for a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, is about 4% to about 80% (e.g., about 4% to about 60%, about 4% to about 40%, about 4% to about 20%, about 4% to about 10%, about 20% to about 40%, or about 20% to about 30%). In some embodiments, the %F for a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B- 1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I- Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F- 1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, is about 4% to about 20%. In some embodiments, the %F for a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I- A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I- E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb- 3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I- H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, is about 20% to about 40%. In some embodiments, the %F for a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb- 1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, is about 40% to about 60%. In some embodiments, the %F for a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, is about 60% to about 80%. In some embodiments, the clearance for a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb- 1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, dosed PO in rats at 10 mg/kg, is less than 10 mL/min/kg (e.g., less than 5 mL/min/kg, less than 3 mL/min/kg, or less than 1 mL/min/kg). In some embodiments, clearance for a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, dosed PO in rats at 10 mg/kg, is about 0.05 mL/min/kg to about 5 mL/min/kg (e.g., about 0.05 mL/min/kg to about 3 mL/min/kg, about 0.05 mL/min/kg to about 1 mL/min/kg, or about 0.05 mL/min/kg to about 0.5 mL/min/kg). In some embodiments, the AUC for a compound of Formula (I) (e.g., (I-A) (e.g., (I-A- 1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E- 1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I- Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, dosed PO in rats at 10 mg/kg, is about 10 µM•h to about 150 µM•h (e.g., about 10 µM•h to about 100 µM•h, about 10 µM•h to about 50 µM•h, or about 30 µM•h to about 80 µM•h). Heterobifunctional degraders can, in some cases, induce the degradation of off-target proteins. For heterobifunctional degraders that utilize CRBN, common off-target proteins that can be degraded include GSPT1, IKZF1, IKZF2, IKZF3, and/or CK1α. This degradation is generally believed to be due to the E3 binding moiety of the heterobifunctional degrader facilitating ternary complex formation between the off-target protein and CRBN. GSPT1 is a translation termination factor, and CK1α is a kinase that is involved in many key cellular processes including cell cycle progression and chromosome segregation; these are both commonly essential genes, so undesired degradation of either or both may lead to nonspecific cytotoxicity. The IKZF proteins are zinc finger transcription factors that are involved with cell fate during hematopoiesis, and degradation of these proteins has been associated with hematotoxicity. See, e.g., Moreau, Kevin, et al. British Journal of Pharmacology 177.8 (2020): 1709-1718. In some embodiments, the compounds of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A- 2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E- 2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H- 2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or pharmaceutically acceptable salts thereof, can exhibit potent and selective induction of degradation of a BCL-X L protein. In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt thereof, can selectively target a BCL-X L protein for degradation over an off- target protein, such as another BCL-2 family member (e.g., BCL-2 and/or MCL-1) or non- BCL-2 family member target (e.g., GSPT1, IKZF1, IKZF2, IKZF3, and/or CK1α). As used herein, “selective” or “selectively”, when referring to a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I- C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea- 3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I- G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof in a protein degradation assay, indicates at least a 5- fold (e.g., at least a 10-fold, at least a 25-fold, at least a 50-fold, or at least a 100-fold) superior performance in the protein degradation assay for a specified protein with reference to a comparator protein in the assay. In some embodiments, the specified protein is BCL-X L protein and the comparator is BCL-2 protein. For example, if a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, “selectively” induces degradation of BCL-X L protein over BCL-2 protein as determined by a degradation assay, then the compound has at least a 5-fold (e.g., at least a 10-fold, at least a 25-fold, at least a 50-fold, or at least a 100-fold) smaller DC50 value for BCL-X L protein than for the BCL-2 protein when measured by the degradation assay. In some embodiments, the compounds provided herein can exhibit potency (e.g., nanomolar potency) against a BCL-XL protein with minimal activity (e.g., micromolar potency) against BCL-2 family members (e.g., BCL-2 or MCL-1 proteins). In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, can exhibit potent degradation of a BCL-XL protein and have minimal potency in degrading (e.g., as measured by Ymin, DC50, and/or D max values) an off-target protein (e.g., a BCL-2 family member (e.g., BCL-2 and/or MCL-1), GSPT1, IKZF1, IKZF2, IKZF3 and/or CK1α). In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I- F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, can exhibit greater induction of degradation of a BCL-X L protein relative to induction of degradation (e.g., as measured by Y min , DC 50 , and/or Dmax values) of an off-target protein (e.g., a BCL-2 family member (e.g., BCL-2 and/or MCL- 1), GSPT1, IKZF1, IKZF2, IKZF3, and/or CK1α). In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, can exhibit at least 2-fold, 3-fold, 5-fold, 10-fold, 25- fold, 50-fold, or 100-fold greater induction of degradation of a BCL-XL protein relative to induction of degradation of an off-target protein. In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, can exhibit up to 1000-fold greater induction of degradation of a BCL-XL protein relative to induction of degradation of an off-target protein. In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A- 3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E- 3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H- 3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, can exhibit from about 2-fold to about 10-fold greater induction of degradation of a BCL-XL protein relative to induction of degradation of an off-target protein (e.g., GSPT1, IKZF1, IKZF2, and/or IKZF3 and/or CK1α) (e.g., as measured by Y min , DC 50 , and/or D max values). In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, can exhibit from about 10-fold to about 100-fold greater induction of degradation of a BCL-X L protein relative to induction of degradation of an off-target protein. In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, can exhibit from about 100-fold to about 1000-fold greater induction of degradation of a BCL-X L protein relative to induction of degradation of an off-target protein. In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A- 3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E- 3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H- 3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, can exhibit from about 1000-fold to about 10000-fold greater induction of degradation of a BCL-XL protein relative to induction of degradation of an off-target protein. Certain agents that inhibit or induce degradation of BCL-XL have demonstrated platelet toxicity, which resulted in dose-limiting toxicity (e.g., thrombocytopenia) in the clinic. See, e.g., Adams and Cory, Cell Death & Differentiation 25.1 (2018): 27-36; Campbell and Tait. Open Biology 8.5 (2018): 180002; Pullarkat et al., Cancer Discovery (2021) 10.1158/2159- 8290.CD-20-1465; Negi and Voisin-Chiret. ChemBioChem (2022) (doi: 10.1002/cbic.202100689). The viability of platelets may be monitored with any appropriate assay, such as those described herein. See, for instance, Example B6. In some embodiments, a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I- C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea- 3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I- G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, shows at least about 30% platelet viability (e.g., at least about 50% platelet viability, or at least about 80% platelet viability) when administered to a subject. In some embodiments, a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, shows about 30% to about 100% platelet viability (e.g., about 50% to about 100% platelet viability, or about 80% to about 100% platelet viability) when administered to a subject. In some embodiments, a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I- C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea- 3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I- G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, shows at least about 30% platelet viability (e.g., at least about 50% platelet viability, or at least about 80% platelet viability) when administered to a subject and has a Y min value of about 50% to about 70%. In some embodiments, a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A- 2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E- 2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H- 2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, shows at least about 30% platelet viability (e.g., at least about 50% platelet viability, or at least about 80% platelet viability) when administered to a subject and has a Ymin value of less than about 50%. In some embodiments, a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, shows at least about 30% platelet viability (e.g., at least about 50% platelet viability, or at least about 80% platelet viability) when administered to a subject and has a Y min value of about 0% to about 50%. In some embodiments, a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I- C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea- 3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I- G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, shows between about 30% and 100% platelet viability (e.g., about 50% to about 100% platelet viability, or about 80% to about 100% platelet viability) when administered to a subject and has a Y min value of about 50% to about 70%. In some embodiments, a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, shows between about 30% and 100% platelet viability (e.g., about 50% to about 100% platelet viability, or about 80% to about 100% platelet viability) when administered to a subject and has a Ymin value of less than about 50%. In some embodiments, a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I- A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I- E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb- 2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I- H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, shows between about 30% and 100% platelet viability (e.g., about 50% to about 100% platelet viability, or about 80% to about 100% platelet viability) when administered to a subject and has a Y min value of about 0% to about 50%. In some embodiments, therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I- Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I- Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, shows at least about 50% platelet viability (e.g., at least about 80% platelet viability) when administered to a subject and has a Ymin value of about 50% to about 70%. In some embodiments, a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I- F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, shows at least about 50% platelet viability (e.g., at least about 80% platelet viability) when administered to a subject and has a Ymin value of less than about 50%. In some embodiments, a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, shows at least about 50% platelet viability (e.g., at least about 80% platelet viability) when administered to a subject and has a Ymin value of about 0% to about 50%. In some embodiments, a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I- C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea- 3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I- G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, shows between about 50% and 100% platelet viability (e.g., about 80% to about 100% platelet viability) when administered to a subject and has a Y min value of about 50% to about 70%. In some embodiments, a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, shows between about 50% and 100% platelet viability (e.g., about 80% to about 100% platelet viability) when administered to a subject and has a Ymin value of less than about 50%. In some embodiments, a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A- 2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E- 2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H- 2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, shows between about 50% and 100% platelet viability (e.g., about 80% to about 100% platelet viability) when administered to a subject and has a Ymin value of about 0% to about 50%. In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, when tested at a concentration of about 0.25 µM to about 3 µM in the assay in Example B6, shows at least about 30% platelet viability (e.g., at least about 50% platelet viability, or at least about 80% platelet viability). In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I- A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I- E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H- 1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, when tested at a concentration of about 0.25 µM to about 3 µM in the assay in Example B6, shows about 30% to about 100% platelet viability (e.g., about 50% to about 100% platelet viability, or about 80% to about 100% platelet viability). In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, when tested at a concentration of about 0.25 µM to about 3 µM in the assay in Example B6, shows at least about 30% platelet viability (e.g., at least about 50% platelet viability, or at least about 80% platelet viability) and has a Ymin value of about 50% to about 70% in the assay described in Example B1. In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb- 2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I- H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, when tested at a concentration of about 0.25 µM to about 3 µM in the assay in Example B6, shows at least about 30% platelet viability (e.g., at least about 50% platelet viability, or at least about 80% platelet viability) and has a Y min value of less than about 50% in the assay described in Example B1. In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb- 2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I- H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, when tested at a concentration of about 0.25 µM to about 3 µM in the assay in Example B6, shows at least about 30% platelet viability (e.g., at least about 50% platelet viability, or at least about 80% platelet viability) and has a Y min value of about 0% to about 50% in the assay described in Example B1. In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, when tested at a concentration of about 0.25 µM to about 3 µM in the assay in Example B6, shows between about 30% and 100% platelet viability (e.g., about 50% to about 100% platelet viability, or about 80% to about 100% platelet viability) and has a Y min value of about 50% to about 70% in the assay described in Example B1. In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, when tested at a concentration of about 0.25 µM to about 3 µM in the assay in Example B6, shows between about 30% and 100% platelet viability (e.g., about 50% to about 100% platelet viability, or about 80% to about 100% platelet viability) and has a Ymin value of less than about 50% in the assay described in Example B1. In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, when tested at a concentration of about 0.25 µM to about 3 µM in the assay in Example B6, shows between about 30% and 100% platelet viability (e.g., about 50% to about 100% platelet viability, or about 80% to about 100% platelet viability) and has a Y min value of about 0% to about 50% in the assay described in Example B1. In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, when tested at a concentration of about 0.25 µM to about 3 µM in the assay in Example B6, shows at least about 50% platelet viability (e.g., at least about 80% platelet viability) and has a Ymin value of about 50% to about 70% in the assay described in Example B1. In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea- 1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I- F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, when tested at a concentration of about 0.25 µM to about 3 µM in the assay in Example B6, shows at least about 50% platelet viability (e.g., at least about 80% platelet viability) and has a Y min value of less than about 50% in the assay described in Example B1. In some embodiments, a compound of Formula (I) (e.g., (I- A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I- E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, when tested at a concentration of about 0.25 µM to about 3 µM in the assay in Example B6, shows at least about 50% platelet viability (e.g., at least about 80% platelet viability) and has a Ymin value of about 0% to about 50% in the assay described in Example B1. In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, when tested at a concentration of about 0.25 µM to about 3 µM in the assay in Example B6, shows between about 50% and 100% platelet viability (e.g., about 80% to about 100% platelet viability) and has a Y min value of about 50% to about 70% in the assay described in Example B1. In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I- Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, when tested at a concentration of about 0.25 µM to about 3 µM in the assay in Example B6, shows between about 50% and 100% platelet viability (e.g., about 80% to about 100% platelet viability) and has a Ymin value of less than about 50% in the assay described in Example B1. In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A- 3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E- 3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H- 3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, when tested at a concentration of about 0.25 µM to about 3 µM in the assay in Example B6, shows between about 50% and 100% platelet viability (e.g., about 80% to about 100% platelet viability) and has a Y min value of about 0% to about 50% in the assay described in Example B1. Provided herein is a method of treating a cancer in a subject in need of such treatment, the method comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea- 1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I- F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof. In some embodiments, the subject is treatment naïve with respect to the cancer. In some embodiments, the subject has received one or more lines of previous therapy for the cancer. Also provided herein is a method of treating a cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea- 1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I- F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, as a monotherapy. In some embodiments, the subject is treatment naïve with respect to the cancer. In some embodiments, the subject has received one or more lines of previous therapy for the cancer. Provided herein is a method of treating a cancer in a subject in need of such treatment, the method comprising: (a) detecting a biomarker (e.g., a mutation, an amplification, a copy number increase, and/or expression (optionally including level of expression) of the biomarker) associated with the cancer; and (b) administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I- B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I- F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, as a monotherapy or in combination with an additional therapy or therapeutic agent. Also provided herein is a method of treating a cancer in a subject, wherein the subject has been determined (e.g., prior to administration of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a))) to have a biomarker (e.g., a mutation, an amplification, a copy number increase, and/or expression (optionally including level of expression) of the biomarker) associated with the cancer, the method comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I- Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I- Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, as a monotherapy or in combination with an additional therapy or therapeutic agent. Provided herein is a method of treating a cancer in a subject in need of such treatment, the method comprising: (a) detecting a biomarker (e.g., a mutation, an amplification, a copy number increase, and/or expression (optionally including level of expression) of a marker of susceptibility to particular agents (e.g., HER2 expression, ER expression, PR expression, folate receptor expression)) associated with the cancer; and (b) administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I- B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I- F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, as a monotherapy or in combination with an additional therapy or therapeutic agent. Also provided herein is a method of treating a cancer in a subject, wherein the subject has been determined (e.g., prior to administration of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a))) to have a biomarker (e.g., a mutation, an amplification, a copy number increase, and/or expression (optionally including level of expression) of a marker of susceptibility to particular agents (e.g., HER2 expression, ER expression, PR expression, folate receptor expression)) associated with the cancer, the method comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea- 1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I- F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, as a monotherapy or in combination with an additional therapy or therapeutic agent. Provided herein is use of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for the treatment of cancer, for example, any of the cancers provided herein. Provided herein is use of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, as a medicament for the treatment of cancer, for example, any of the cancers provided herein. Provided herein is use of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of cancer, for example, any of the cancers provided herein. Provided herein is a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I- A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I- E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb- 3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I- H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use as a medicament. Also provided herein is a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea- 1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I- F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use as a medicament for the treatment of cancer, for example, any of the cancers provided herein. Provided herein is a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I- A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I- E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb- 3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I- H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use in treating a cancer, for example, any of the cancers provided herein. As used herein, “monotherapy”, when referring to a compound of Formula (I) (e.g., (I- A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I- E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, means that the compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I- A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I- E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb- 2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I- H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, is the only therapeutic agent or therapy (e.g., anticancer agent or therapy) administered to the subject during the treatment cycle (e.g., no additional targeted therapeutics, anticancer agents, chemotherapeutics, or checkpoint inhibitors are administered to the subject during the treatment cycle). As a person of ordinary skill in the art would understand, monotherapy does not exclude the co-administration of medicaments for the treatment of side effects or general symptoms associated with the cancer or treatment, such as pain, rash, edema, photosensitivity, pruritis, skin discoloration, hair brittleness, hair loss, brittle nails, cracked nails, discolored nails, swollen cuticles, fatigue, weight loss, general malaise, shortness of breath, infection, anemia, or gastrointestinal symptoms, including nausea, diarrhea, and lack of appetite. As used herein, “the subject has previously received one or more therapeutic agents or therapies for the cancer” means that the subject has been previously administered one or more therapeutic agents or therapies (e.g., anticancer agent or therapy) for the cancer other than a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea- 1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I- F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, during a prior treatment cycle. In some embodiments, the subject cannot tolerate the one or more therapeutic agents or therapies previously administered for the cancer. In some embodiments, the subject did not respond to the one or more therapeutic agents or therapies previously administered for the cancer. In some embodiments, the subject did not adequately respond to one or more therapeutic agents or therapies previously administered for the cancer. In some embodiments, the subject has stopped responding to the one or more therapeutic agents or therapies previously administered for the cancer. In some embodiments, a lack of response, an inadequate response, or a discontinued response can be determined by objective criteria (e.g., tumor volume, or by criteria such as RECIST 1.1). In some embodiments, a lack of response, an inadequate response, or a discontinued response can be determined by the subject’s physician. As used herein, “the subject is treatment naïve with respect to the cancer” means that the subject has not been previously administered one or more therapeutic agents or therapies for the cancer. For any of the solid tumors described herein, the solid tumor can be primary tumors or metastatic (or secondary) tumors. As used herein, “primary” tumors are those located at the site where the tumor began to grow (i.e., where it originated). As used herein, “metastatic” (or “secondary”) tumors are those that have spread to other parts of body from the original tumor site. In some embodiments, the metastatic or secondary tumors are the same type of cancer as the primary tumor. In some embodiments, the metastatic or secondary tumors are not genetically identical to the primary tumor. In some embodiments of any of the methods or uses described herein, the cancer is breast cancer (e.g., breast invasive carcinoma, breast invasive ductal carcinoma), central or peripheral nervous system tissue cancer (e.g., brain cancer (e.g., astrocytoma, glioblastoma, glioma, oligoastrocytoma)), endocrine or neuroendocrine cancer (e.g., adrenal cancer (e.g., adrenocortical carcinoma, neuroblastoma, pheochromocytoma, paraganglioma), multiple neuroendocrine type I and type II tumors, parathyroid cancer, pituitary tumors, thyroid cancer (e.g., papillary thyroid cancer)), eye cancer (e.g., uveal cancer (e.g., uveal melanoma)), gastrointestinal cancer (e.g., anal cancer, bile duct cancer (e.g., cholangiocarcinoma (e.g., intrahepatic cholangiocarcinoma)), colorectal cancer (e.g., colon adenocarcinoma, rectal adenocarcinoma, mucinous adenocarcinoma, mucinous carcinoma), esophageal cancer (e.g., esophageal adenocarcinoma), gallbladder cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor (GIST), liver cancer (e.g., hepatocellular carcinoma, intrahepatic bile duct cancer), pancreatic cancer (e.g., pancreatic adenocarcinoma, pancreatic islet cell cancer), small intestine cancer, or stomach cancer (e.g., stomach adenocarcinoma, signet ring cell carcinoma of the stomach)), genitourinary cancer (e.g., bladder cancer (e.g., bladder urothelial carcinoma), kidney cancer (e.g., renal clear cell carcinoma, renal papillary cell carcinoma, kidney chromophobe), prostate cancer (e.g., prostate adenocarcinoma), testicular cancer (e.g., testicular germ cell tumors), or ureter cancer), gynecologic cancer (e.g., cervical cancer (e.g., cervical squamous cell carcinoma, endocervical adenocarcinoma, mucinous carcinoma), ovarian cancer (e.g., serous ovarian cancer, ovarian serous cystadenocarcinoma), uterine cancer (e.g., uterine carcinosarcoma, uterine endometrioid carcinoma, uterine serous carcinoma, uterine papillary serous carcinoma, uterine corpus endometrial carcinoma), or vulvar cancer), head and neck cancer (e.g., ear cancer (e.g., middle ear cancer), head and neck squamous cell carcinoma, nasal cavity cancer, oral cancer, pharynx cancer (e.g., hypopharynx cancer, nasopharynx cancer, oropharyngeal cancer), hematological cancer (e.g., leukemia (e.g., chronic eosinophilic leukemia (CEL), chronic lymphocytic leukemia (CLL), chronic neutrophilic leukemia (CNL), acute lymphocytic leukemia (ALL) (e.g., Philadelphia chromosome positive ALL or T cell ALL (T-ALL)), acute myeloid leukemia (AML) (e.g., acute promyelocytic leukemia (APL), post-MPN AML, post-myelodysplastic syndrome (post- MDS) AML, M6-AML (also known as pure erythroid leukemia (PEL)), or M7-AML (also known as acute megakaryoblastic leukemia (AKML))), chronic myeloid leukemia (CML)), lymphoma (e.g., Hodgkin lymphoma (e.g., nodular lymphocyte predominant Hodgkin lymphoma (NLPHL)), or non-Hodgkin lymphoma (e.g., Burkitt lymphoma (BL), diffuse large B cell lymphoma (DLBCL), diffuse histiocytic lymphoma (DHL), follicular lymphoma (FL), intravascular large B cell lymphoma (IVLBCL), mantle cell lymphoma (MCL), small lymphocytic lymphoma (SLL), a T cell lymphoma (e.g., anaplastic large T cell lymphoma, cutaneous T cell lymphoma, or peripheral T cell lymphoma))), essential thrombocythemia, polycythemia vera, myelofibrosis (e.g., primary myelofibrosis, post-essential thrombocythemia myelofibrosis, or post-polycythemia vera myelofibrosis), a myelodysplastic syndrome (MDS) (e.g., M6 MDS or M7 MDS), or myeloma (e.g., multiple myeloma)), Li- Fraumeni tumors, mesentery cancer (e.g., omentum cancer, peritoneal cancer), pleural cancer, respiratory cancer (e.g., larynx cancer, lung cancer (e.g., lung squamous cell carcinoma, lung adenocarcinoma, malignant pleural mesothelioma, non-small cell lung cancer (NSCLC), small cell lung cancer), tracheal cancer), sarcoma (e.g., bone cancer (e.g., osteosarcoma, chondrosarcoma) or soft tissue sarcoma (Ewing sarcoma, leiomyosarcoma, myxofibrosarcoma, rhabdomyosarcoma)), skin cancer (e.g., melanoma or Merkel cell carcinoma), thymus cancer (e.g., thymoma), or a combination thereof. In some embodiments, the cancer is breast cancer, gastrointestinal cancer (e.g., bile duct cancer (e.g., cholangiocarcinoma (e.g., intrahepatic cholangiocarcinoma)), colorectal cancer (CRC), gastrointestinal stromal tumor, or pancreatic cancer), genitourinary cancer (e.g., bladder cancer (e.g., bladder urothelial carcinoma) or kidney cancer), gynecologic cancer (e.g., cervical cancer, ovarian cancer (e.g., high grade serous ovarian cancer (HGSOC), low grade serous ovarian cancer (LGSOC)), or uterine cancer), head and neck cancer (e.g., head and neck squamous cell carcinoma), hematological cancer (e.g., leukemia (e.g., acute lymphocytic leukemia (ALL) (e.g., T-ALL), acute myeloid leukemia (AML) (e.g., APL, post-MPN AML, post-MDS AML, M6-AML, or M7-AML), chronic lymphocytic leukemia (CLL)), lymphoma (e.g., follicular lymphoma (FL), small lymphocytic lymphoma (SLL), a T cell lymphoma (e.g., anaplastic large T cell lymphoma, cutaneous T cell lymphoma, or peripheral T cell lymphoma), or diffuse large B cell lymphoma (DLBCL)), essential thrombocythemia, polycythemia vera, myelofibrosis (e.g., primary myelofibrosis, post-essential thrombocythemia myelofibrosis, or post-polycythemia vera myelofibrosis), or a myelodysplastic syndrome (MDS) (e.g., M6 MDS or M7 MDS)), lung cancer (e.g., non small cell lung cancer (NSCLC), small cell lung cancer (SCLC), lung squamous cell carcinoma, or malignant pleural mesothelioma (MPM)), neuroblastoma, sarcoma (e.g., bone cancer (e.g., osteosarcoma, chondrosarcoma) or soft tissue sarcoma (Ewing sarcoma, leiomyosarcoma, myxofibrosarcoma, rhabdomyosarcoma)), or skin cancer (e.g., melanoma or Merkel cell carcinoma). In some embodiments, the cancer is a myeloproliferative neoplasm (MPN). In some embodiments, the myeloproliferative neoplasm is CEL, CML, CNL, essential thrombocythemia, polycythemia vera, or myelofibrosis (e.g., primary myelofibrosis, post- essential thrombocythemia myelofibrosis, or post-polycythemia vera myelofibrosis). In some embodiments, the MPN has a JAK2 mutation (e.g., a JAK2 V617F mutation). In some embodiments, the MPN does not have a JAK2 mutation. In some embodiments, the cancer is low-risk myelofibrosis. In some embodiments, the cancer is intermediate (e.g., intermediate-1 and/or intermediate-2) or high-risk myelofibrosis (e.g., primary myelofibrosis, post-essential thrombocythemia myelofibrosis, or post-polycythemia vera myelofibrosis). In some embodiments, the cancer is intermediate or high-risk myelofibrosis (e.g., primary myelofibrosis, post-essential thrombocythemia myelofibrosis, or post-polycythemia vera myelofibrosis) with a JAK2 mutation (e.g., a JAK2 V617F mutation). In some embodiments, the cancer is intermediate or high-risk myelofibrosis (e.g., primary myelofibrosis, post- essential thrombocythemia myelofibrosis, or post-polycythemia vera myelofibrosis) without a JAK2 V617F mutation. In some embodiments, the subject has a platelet count below 50 x 10 9 /L. In some embodiments, the cancer is polycythemia vera. In some embodiments, the cancer is polycythemia vera, and the subject has had an inadequate response to or is intolerant of hydroxyurea. In some embodiments, the cancer is an MDS. In some embodiments, the cancer is M6 MDS. In some embodiments, the cancer is M7 MDS. In some embodiments, the cancer is T-ALL. In some embodiments, the cancer is relapsed/refractory T-ALL. In some embodiments, the cancer is CRC (e.g., Braf mutant CRC (e.g., Braf V600E CRC) or KRas mutant CRC (e.g., KRas G12C CRC or KRas G12D CRC)). In some embodiments, the cancer is SCLC (e.g., ASCL1 subtype SCLC or NEUROD1 subtype SCLC). In some embodiments, the cancer is NSCLC (e.g., Braf mutant NSCLC (e.g., Braf V600E NSCLC), EGFR mutant NSCLC (e.g., EGFR L858R NSCLC or EGFR exon 19 deletion NSCLC), MET mutant NSCLC (e.g., MET exon 14 deletion NSCLC, MET amplified NSCLC), KRas mutant NSCLC (e.g., KRas G12C NSCLC)). In some embodiments, the cancer is lung squamous cell carcinoma. In some embodiments, the cancer is malignant pleural mesothelioma (e.g., BAP1 mutant malignant pleural mesothelioma). In some embodiments, the cancer is malignant pleural mesothelioma, and a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I- A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I- E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb- 3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I- H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, is administered as a monotherapy. In some embodiments, the cancer is melanoma (e.g., Braf mutant melanoma (e.g., Braf V600E melanoma)). In some embodiments, the cancer is breast cancer (e.g., HER2+ breast cancer (e.g., HER2+ breast cancer with ER expression, HER2+ breast cancer without ER expression), HER2 negative breast cancer (e.g., HER2 negative breast cancer with ER expression, HER2 negative breast cancer without ER expression), HER2 low breast cancer, triple negative breast cancer, hormone receptor positive breast cancer (ER+ and/or PR+, with or without HER2 positivity)). In some embodiments, the cancer is lymphoma. In some embodiments, the lymphoma is a T cell lymphoma (e.g., anaplastic large T cell lymphoma, cutaneous T cell lymphoma, or peripheral T cell lymphoma). In some embodiments, the lymphoma is a non-Hodgkin lymphoma (e.g., DLBCL, anaplastic large T cell lymphoma, cutaneous T cell lymphoma, or peripheral T cell lymphoma). In some embodiments, the lymphoma is peripheral T cell lymphoma. In some embodiments, the cancer is leukemia. In some embodiments, the leukemia is a T cell leukemia (e.g., T cell ALL). In some embodiments, the cancer is post-MPN leukemia. In some embodiments, the cancer is a T cell leukemia (e.g., T cell ALL), and a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, is administered as a monotherapy. In some embodiments, the cancer is M6-AML. In some embodiments, the M6-AML is a post-MPN AML. In some embodiments, the M6-AML is a post-myelodysplastic syndrome (MDS) AML. In some embodiments, the cancer is M6-AML, and the compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I- C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea- 3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I- G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or pharmaceutically acceptable salt thereof, is administered as a monotherapy. In some embodiments, the cancer is M7-AML. In some embodiments, the M7-AML is a post-MPN AML. In some embodiments, the M7-AML is a post-MDS AML. In some embodiments, the cancer is M7-AML, and the compound of Formula (I) (e.g., (I-A) (e.g., (I- A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I- E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H- 1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or pharmaceutically acceptable salt thereof, is administered as a monotherapy. In some embodiments, the cancer is head and neck cancer. In some embodiments, the cancer is essential thrombocythemia. In some embodiments, the cancer is polycythemia vera. In some embodiments, the cancer is myelofibrosis. In some embodiments, the cancer is primary myelofibrosis. In some embodiments, the cancer is post-essential thrombocythemia myelofibrosis. In some embodiments, the cancer is post-polycythemia vera myelofibrosis. In some embodiments, the cancer is an MDS. In some embodiments, the MDS is M6 MDS. IN some embodiments, the MDS is M7 MDS. In some embodiments, the cancer is an MDS, and a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, is administered as a monotherapy. In some embodiments, the cancer is pancreatic cancer. In some embodiments, the cancer is bladder cancer (e.g., bladder urothelial carcinoma). In some embodiments, the cancer is ovarian cancer (e.g., BRCA1 mutant ovarian cancer or BRCA2 mutant ovarian cancer). In some embodiments, the cancer is HGSOC (e.g., BRCA1 mutant HGSOC or BRCA2 mutant HGSOC). In some embodiments, the cancer is cervical cancer. In some embodiments, the cancer is colorectal cancer. In some embodiments, the cancer is skin cancer. In some embodiments, the skin cancer is melanoma. In some embodiments, the cancer is Merkel cell carcinoma. In some embodiments, the cancer is Merkel cell carcinoma, and a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, is administered as a monotherapy. In some embodiments, the cancer is neuroblastoma. In some embodiments, the cancer is intrahepatic cholangiocarcinoma. In some embodiments, the cancer is a mesenchymal cancer. In some embodiments, the mesenchymal cancer is mesenchymal breast cancer or mesenchymal kidney cancer. In some embodiments, a BCL-XL copy number gain or a BCL-XL amplification can be detected in a sample from the subject (e.g., detecting three or more copies of a BCL2L1 gene in the sample from the subject). In some embodiments, the subject was determined (e.g., prior to administration of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I- F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a))) to have a cancer having a BCL-XL copy number gain or a BCL- X L amplification. In some embodiments, the cancer has a BCL-X L copy number gain. In some embodiments, the cancer has a BCL-XL amplification. Non-limiting examples of BCL-X L involvement in cancers can be found in: Wilson, Wyndham H., et al. The Lancet Oncology 11.12 (2010): 1149-1159; Keitel, Ulrike, et al. Oncotarget 5.23 (2014): 11778; Chonghaile, Triona Ni, et al. Cancer Discovery 4.9 (2014): 1074-1087; Zaanan, Aziz, et al. Journal of Biological Chemistry 290.39 (2015): 23838-23849; Zhang, Haichao, et al. Molecular Cancer 14.1 (2015): 1-9; Soderquist, Ryan S., et al. Nature Communications 9.1 (2018): 1-13; Stover, Elizabeth H., et al. Molecular Cancer Research 17.11 (2019): 2281-2293; Concoran, R.B., et al. Annals of Oncology (2019) 30 (suppl_5): v159-v193; Lakhani, Nehal J., et al. Journal of Clinical Oncology (2020): 3509-3509 doi: 10.1200/JCO.2020.38; He, Yonghan, et al. Journal of Hematology & Oncology 13.1 (2020): 1-13; Grubb, Treg, et al. Clinical Cancer Research (2022) doi: 10.1158/1078-0432.CCR-22- 0669; Joly, Florence, et al. Gynecologic Oncology 165.1 (2022): 30-39; and Nanjo, Shigeki, et al. The Journal of Clinical Investigation (2022) doi: 10.1172/JCI145099. In some embodiments, the subject has previously been treated with another anticancer agent, a chemotherapeutic agent, radiation, surgery, a multi-kinase inhibitor, or a combination thereof. Provided herein is a method of treating an ocular disease or condition in a subject in need of such treatment, the method comprising administering (e.g., intravitreally or topically) to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof. In some embodiments, the ocular disease or condition is diabetic macular edema. In some embodiments, the ocular disease or condition is age-related macular degeneration. In some embodiments, the ocular disease or condition is diabetic retinopathy. See, e.g., Crespo-Garcia, Sergio, et al. Cell Metabolism 33.4 (2021): 818-832; Hassan, Jannah Waled, and Ashay D. Bhatwadekar, Frontiers in Pharmacology 13 (2022): 896907. Provided herein is use of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for the treatment of an ocular disease or condition, for example, any of the ocular diseases or conditions provided herein. Provided herein is use of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, as a medicament for the treatment of an ocular disease or condition, for example, any of the ocular diseases or conditions provided herein. Provided herein is use of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of an ocular disease or condition, for example, any of the ocular diseases or conditions provided herein. Also provided herein is a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use as a medicament for the treatment of an ocular disease or condition, for example, any of the ocular diseases or conditions provided herein. Provided herein is a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I- A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I- E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb- 3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I- H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use in treating an ocular disease or condition, for example, any of the ocular diseases or conditions provided herein. Also provided herein is a method of treating a fibrotic disease or condition and/or a disease or condition associated with senescent cells in a subject in need of such treatment, the method comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea- 1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I- F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof. Non-limiting examples of fibrotic diseases or conditions and/or diseases or conditions associated with senescent cells include pulmonary fibrosis (e.g., idiopathic pulmonary fibrosis, systemic fibrosis-associated lung disease, radiation-associated pulmonary fibrosis), radiation- associated skin fibrosis, liver fibrosis, primary sclerosing cholangitis, diabetic macular edema, age-related macular degeneration, diabetic retinopathy, geographic atrophy, ischemia and reperfusion injury, heart failure, recovery from acute myocardial infarction, pulmonary hypertension, inflammatory bowel disease, colitis, Crohn’s disease, diabetes, aging skin (including photoaging-related pigmentation), donor organ transplant survival and function, stem cell transplant survival and function, osteoarthritis, recovery from spinal cord injury, Alzheimer’s disease, tau-opathies, progressive supranuclear palsy, and age-related neurological decline (e.g., related to impaired neurovascular coupling). See, e.g., Zhu, Y. I., et al. Aging Cell 14.4 (2015): 644-658; Zhu, Y. I., et al. Aging Cell 15.3 (2016): 428-435; Chang, Jianhui, et al. Nature Medicine 22.1 (2016): 78-83; Zhu, Yi, et al. Aging (Albany NY) 9.3 (2017): 955; Lagares, David, et al. Science Translational Medicine 9.420 (2017): eaal3765; Pan, Jin, et al. International Journal of Radiation Oncology Biology Physics 99.2 (2017): 353- 361; Bussian, Tyler J., et al. Nature 562.7728 (2018): 578-582; Moncsek, Anja, et al. Hepatology 67.1 (2018): 247-259; van Willigenburg, Hester, Peter LJ de Keizer, and Ron WF de Bruin. Pharmacological Research 130 (2018): 322-330; Walaszczyk, Anna, et al. Aging Cell 18.3 (2019): e12945; Aguayo-Mazzucato, Cristina, et al. Cell Metabolism 30.1 (2019): 129-142; Sessions, Garrett A., et al. The FASEB Journal 33.11 (2019): 12364; Gerdes, Erin O. Wissler, et al. International Review of Neurobiology 155 (2020): 203-234; Sasaki, Motoko, Yasunori Sato, and Yasuni Nakanuma. Journal of Autoimmunity 107 (2020): 102377; Yabluchanskiy, Andriy, et al. Geroscience 42 (2020): 409-428; Dookun, Emily, et al. Aging Cell 19.10 (2020): e13249; Jia, Kangni, et al. Journal of Cardiovascular Pharmacology 76.4 (2020): 452-460; Sierra-Ramirez, Arantzazu, et al. Aging (Albany NY) 12.12 (2020): 11337; Yang, Hao, et al. Aging (Albany NY) 12.13 (2020): 12750; Lawrie, Allan, and Sheila E. Francis. The Journal of Clinical Investigation 131.11 (2021): e149721; Paramos-de-Carvalho, Diogo, et al. Cell Reports 36.1 (2021): 109334; Tarantini, Stefano, et al. GeroScience 43.5 (2021): 2427-2440; Park, Ji Hee, et al. The British Journal of Dermatology (2021); Fielder, Edward, et al. ELife 11 (2022): e75492; Suzuki, Keiji, et al. Mutation Research/Genetic Toxicology and Environmental Mutagenesis 876 (2022): 503448; He, An, et al. American Journal of Transplantation 22.11 (2022): 2529-2547; Johnson, Laura A., et al. Inflammatory Bowel Diseases 28.2 (2022): 161-175; Miura, Yugo, et al. Stem Cell Research & Therapy 13.1 (2022): 222; Cooley, Joseph C., et al. JCI Insight 8.3 (2023): e163762. Watanabe, Yusuke, et al. Hepatology Research 53 (2023): 460-472; and Takaya, Kento, et al., Rejuvenation Research 26.1 (2023): 9-20. Provided herein is use of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for the treatment of a fibrotic disease or condition and/or a disease or condition associated with senescent cells, for example, any of the fibrotic diseases or conditions and/or diseases or conditions associated with senescent cells provided herein. Provided herein is use of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, as a medicament for the treatment of a fibrotic disease or condition and/or a disease or condition associated with senescent cells, for example, any of the fibrotic diseases or conditions and/or diseases or conditions associated with senescent cells provided herein. Provided herein is use of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a fibrotic disease or condition and/or a disease or condition associated with senescent cells, for example, any of the fibrotic diseases or conditions and/or diseases or conditions associated with senescent cells provided herein. Also provided herein is a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use as a medicament for the treatment of a fibrotic disease or condition and/or a disease or condition associated with senescent cells, for example, any of the fibrotic diseases or conditions and/or diseases or conditions associated with senescent cells provided herein. Provided herein is a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I- A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I- E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb- 3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I- H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for use in treating a fibrotic disease or condition and/or a disease or condition associated with senescent cells, for example, any of the fibrotic diseases or conditions and/or diseases or conditions associated with senescent cells provided herein. Also provided is a method for modulating (e.g., decreasing) BCL-X L protein activity in a cell, comprising contacting the cell with an effective compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof. In some embodiments, the contacting is in vitro. In some embodiments, the contacting is in vivo. In some embodiments, the contacting is in vivo, wherein the method comprises administering an effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb- 1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, to a subject. In some embodiments, the cell is a cancer cell. In some embodiments, the cell is a mammalian cell. In some embodiments, the cell is a mammalian cancer cell. In some embodiments, the cancer cell is any cancer as described herein. As used herein, the term “contacting” refers to the bringing together of indicated moieties in an in vitro system or an in vivo system. For example, “contacting” a cell with a compound provided herein includes the administration of a compound provided herein to the cell, in vitro or in vivo, including, for example, introducing a compound provided herein into a sample containing cells (e.g., grown in culture or derived from a patient), an organoid, or an organism (e.g., an animal (e.g., an animal bearing a tumor), or a human). Also provided is a method of modulating (e.g., decreasing) the level of BCL-XL protein in a cell, comprising contacting the cell with a compound of Formula (I) (e.g., (I-A) (e.g., (I- A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I- E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H- 1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof. In some embodiments, the level of BCL-XL protein is decreased by at least 30% (e.g., at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 97%, or at least 99%) compared to a cell not contacted with the compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I- C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea- 3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I- G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof. In some embodiments, the contacting is in vitro. In some embodiments, the contacting is in vivo. In some embodiments, the contacting is in vivo, wherein the method comprises administering an effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I- C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea- 3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I- G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, to a subject having a cell having a BCL-XL protein. In some embodiments, the cell is a cancer cell. In some embodiments, the cell is a mammalian cell. In some embodiments, the cell is a mammalian cancer cell. In some embodiments, the cancer cell is any cancer as described herein. Also provided is a method of inducing ubiquitination of a BCL-X L protein in a cell, comprising contacting the cell with a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I- A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I- E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb- 2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I- H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof. In some embodiments, the contacting is in vitro. In some embodiments, the contacting is in vivo. In some embodiments, the contacting is in vivo, wherein the method comprises administering an effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I- A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I- E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb- 2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I- H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, to a subject having a cell having a BCL-XL protein. In some embodiments, the cell is a cancer cell. In some embodiments, the cell is a mammalian cell. In some embodiments, the cell is a mammalian cancer cell. In some embodiments, the cancer cell is any cancer as described herein. Also provided is a method of forming a ternary complex comprising a BCL-X L protein, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea- 1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I- F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, and a CRBN protein or fragment thereof in a cell, comprising contacting the cell with a compound of Formula (I) (e.g., (I-A) (e.g., (I- A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I- E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H- 1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof. In some embodiments, the contacting is in vitro. In some embodiments, the contacting is in vivo. In some embodiments, the contacting is in vivo, wherein the method comprises administering an effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I- A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I- E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb- 2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I- H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, to a subject having a cell having a BCL-XL protein. In some embodiments, the cell is a cancer cell. In some embodiments, the cell is a mammalian cell. In some embodiments, the cell is a mammalian cancer cell. In some embodiments, the cancer cell is any cancer as described herein. Also provided herein is a method of inhibiting cell proliferation, in vitro or in vivo, the method comprising contacting a cell with an effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I- Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I- Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof as defined herein. Further provided herein is a method of increasing cell death, in vitro or in vivo, the method comprising contacting a cell with an effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I- Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I- Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof as defined herein. Also provided herein is a method of increasing tumor cell death in a subject. The method comprises administering to the subject a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb- 2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I- H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, in an amount effective to increase tumor cell death. When employed as pharmaceuticals, the compounds of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb- 1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or pharmaceutically acceptable salts thereof, can be administered in the form of pharmaceutical compositions as described herein. Also provided herein is a method for inducing degradation of a BCL-X L protein in a mammalian cell, the method comprising contacting the mammalian cell with an effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof. Also provided herein is a method of treating a subject having a cancer, wherein the method comprises: administering a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, as a monotherapy or in conjunction with a first anticancer agent to the subject who has been administered one or more doses of the first anticancer agent to the subject for a period of time. Also provided herein is a method of treating a subject having a cancer, wherein the method comprises: (a) administering one or more doses of a first anticancer agent to the subject for a period of time; and (b) after (a), administering a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, as a monotherapy or in conjunction with the first anticancer agent to the subject. Also provided herein is a method of treating a subject having a cancer, wherein the method comprises: (a) administering one or more doses of a first anticancer agent to the subject for a period of time; and (b) after (a), administering a therapeutically effective amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, as a monotherapy or in conjunction with a second anticancer agent to the subject. Combinations In any of the indications described herein, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb- 1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, can be used as a monotherapy. In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I- Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I- Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, can be used prior to administration of an additional therapeutic agent or additional therapy. For example, a subject in need thereof can be administered one or more doses of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A- 2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E- 2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H- 2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, for a period of time and then undergo at least partial resection of the tumor. In some embodiments, the treatment with one or more doses of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb- 2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I- H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, reduces the size of the tumor (e.g., the tumor burden) prior to the at least partial resection of the tumor. In some embodiments, a subject in need thereof can be administered one or more doses of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B- 1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I- Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F- 1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, for a period of time and under one or more rounds of radiation therapy. In some embodiments, the treatment with one or more doses of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I- F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, reduces the size of the tumor (e.g., the tumor burden) prior to the one or more rounds of radiation therapy. In some embodiments of any the methods described herein, the compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I- C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea- 3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I- G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, is administered in combination with a therapeutically effective amount of at least one additional therapeutic agent selected from one or more additional therapies or therapeutic (e.g., chemotherapeutic) agents. Non-limiting examples of additional therapies and therapeutic agents include: RAS pathway targeted therapeutic agents (e.g., Ras/RAF/MEK/PI3K pathway inhibitors, (e.g., Ras inhibitors (e.g., KRas inhibitors), KRas-targeted therapeutic agents, SOS1 inhibitors, SOS1/Ras protein-protein interaction inhibitors, SHP2 inhibitors, PI3K-AKT-mTOR pathway inhibitors)), kinase-targeted therapeutics (e.g., MEK inhibitors, ERK inhibitors, Raf inhibitors (e.g., BRaf inhibitors), PI3K inhibitors, Abl inhibitors (e.g., BCR-Abl inhibitors), ALK inhibitors, AKT inhibitors, AURKA inhibitors, mTOR inhibitors, CDK2 inhibitors, CDK4/5 inhibitors, CDK4/6 inhibitors, CDK7 inhibitors, CDK9 inhibitors, MET (also known as cMET) inhibitors, FAK inhibitors, FGFR1 inhibitors, FGFR2 inhibitors, FGFR3 inhibitors, FGFR4 inhibitors, ErbB family inhibitors (e.g., EGFR inhibitors, anti-EGFR antibodies or anti-EGFR antibody-drug conjugates, HER2 inhibitors, anti-HER2 antibodies or anti-HER2 antibody- utilizing biologics), JAK inhibitors (e.g., JAK inhibitors having activity against JAK1, JAK2, and/or JAK1/2), Src inhibitors, VEGFR inhibitors), LSD1 inhibitors, EZH2 inhibitors, BET inhibitors, STING agonists, telomerase inhibitors, mTORC1 inhibitors, YAP inhibitors, proteasome inhibitors, farnesyl transferase inhibitors, Hif2α inhibitors, HSP90 inhibitors, PTEN inhibitors, PARP inhibitors, signal transduction pathway inhibitors, checkpoint inhibitors, modulators of the apoptosis pathway (e.g., BCL-2 inhibitors), chemotherapeutics, angiogenesis-targeted therapies, immune-targeted agents, including immunomodulatory imide drugs (sometimes called “IMiDs” or “CELMoDs”) and immunotherapy (e.g., anti-PD1 therapy or anti-PD-L1 therapy), and radiotherapy. As used herein, a biosimilar antibody refers to an antibody or antigen-binding fragment that has the same primary amino acid sequence as compared to a reference antibody and optionally, may have detectable differences in post-translation modifications (e.g., glycosylation and/or phosphorylation) as compared to the reference antibody (e.g., a different glycoform). In some embodiments, the additional therapy or therapeutic agent is an Abl inhibitor (e.g., a BCR-Abl inhibitor), an ALK inhibitor, an AURKA inhibitor, a BCL-2 inhibitor, a Braf inhibitor, a CDK2 inhibitor, a CDK4/6 inhibitor, a CDK7 inhibitor, a CDK9 inhibitor, an EGFR inhibitor, an anti-EGFR antibody or anti-EGFR antibody-drug conjugate, an ERK inhibitor, an EZH2 inhibitor, a FGFR1 inhibitor, a FGFR2 inhibitor, a FGFR3 inhibitor, a FGFR4 inhibitor, a HER2 inhibitor, an anti-HER2 antibody or anti-HER2 antibody-drug conjugate, a JAK inhibitor, a KRas inhibitor, a MEK inhibitor, a MET inhibitor, a Hif2α inhibitor, a PARP inhibitor, a VEGFR inhibitor, an LSD1 inhibitor, a BET inhibitor, a STING agonist, a telomerase inhibitor, a TORC1/2 inhibitor, an immunomodulatory imide drug, immunotherapy (e.g., a PD-1 inhibitor (e.g., anti-PD1 therapy), a PD-L1 inhibitor (e.g., anti-PD-L1 therapy)), chemotherapy, radiotherapy, or a combination thereof. In some embodiments, the additional therapy or therapeutic agent is an Abl degrader (e.g., a BCR-Abl degrader), an ALK degrader, an AURKA degrader, a BCL-2 degrader, a BRaf degrader, a CDK2 degrader, a CDK4/6 degrader, a CDK7 degrader, a CDK9 degrader, an EGFR degrader, an ERK degrader, an EZH2 degrader, a FGFR1 degrader, a FGFR2 degrader, a FGFR3 degrader, a FGFR4 degrader, a HER2 degrader, a JAK2 degrader, a KRas degrader, a MEK degrader, a MET degrader, a Hif2α degrader, a PARP degrader, a VEGFR degrader, an LSD1 degrader, a BET degrader, a telomerase degrader, a TORC1/2 degrader, an immunomodulatory imide drug, immunotherapy (e.g., anti-PD1 therapy or anti-PD-L1 therapy), chemotherapy, radiotherapy, or a combination thereof. In some embodiments, the Abl inhibitor (e.g., BCR-Abl inhibitor) is asciminib (e.g., asciminib hydrochloride), bafetinib, bosutinib (e.g., bosutinib monohydrate), danusertib, dasatinib (e.g., dasatinib monohydrate), flumatinib (e.g., flumatinib mesylate), imatinib (e.g., imatinib mesylate), nilotinib (e.g., nilotinib monochloride monohydrate), olverembatinib (e.g., olverembatinib mesylate), ponatinib (e.g., ponatinib hydrochloride), radotinib (e.g., radotinib dihydrochloride), ruserontinib, vandetanib, AN-019, AT-9283, IkT-148009, NPB-001-056, or a combination thereof. In some embodiments, the ALK inhibitor is alectinib (e.g., alectinib hydrochloride), brigatinib, ceritinib, crizotinib, ensartinib (e.g., ensartinib hydrochloride), entrectinib, fidrisertib, lorlatinib, TQ-B-3101, TQ-B-3139, or a combination thereof. In some embodiments, the ALK inhibitor is alectinib (e.g., alectinib hydrochloride), brigatinib, ceritinib, crizotinib, ensartinib (e.g., ensartinib hydrochloride), fidrisertib, lorlatinib, TQ-B- 3101, TQ-B-3139, or a combination thereof. In some embodiments, the AURKA inhibitor is alisertib, danusertib, ilorasertib, tinengotinib, AT-9283, BI-811283, ENMD-2076, or a combination thereof. In some embodiments, the BCL-2 inhibitor is lisaftoclax, navitoclax, obatoclax, venetoclax, oblimersen (e.g., oblimersen sodium), beclanorsen, AZD-0466, BGB-11417, UBX-1325 (or a phosphate prodrug thereof), UBX-1967 (or a phosphate prodrug thereof), ZN- d5, or a combination thereof. In some embodiments, the cancer is a lung cancer (e.g., SCLC), and the additional therapy or therapeutic agent is a BCL-2 inhibitor (e.g., lisaftoclax, navitoclax, obatoclax, venetoclax, oblimersen (e.g., oblimersen sodium), beclanorsen, AZD-0466, BGB-11417, UBX-1325 (or a phosphate prodrug thereof), UBX-1967 (or a phosphate prodrug thereof), or ZN-d5). In some embodiments, the cancer is a lung cancer (e.g., SCLC), and the additional therapy or therapeutic agent is venetoclax. In some embodiments, the cancer is a non-Hodgkin lymphoma, and the additional therapy or therapeutic agent is a BCL-2 inhibitor (e.g., lisaftoclax, navitoclax, obatoclax, venetoclax, oblimersen (e.g., oblimersen sodium), beclanorsen, AZD-0466, BGB-11417, UBX-1325 (or a phosphate prodrug thereof), UBX-1967 (or a phosphate prodrug thereof), or ZN-d5). In some embodiments, the cancer is a non-Hodgkin lymphoma, and the additional therapy or therapeutic agent is venetoclax. In some embodiments, the BRaf inhibitor is avutometinib (RO5126766), dabrafenib (e.g., dabrafenib mesylate, GSK2118436), encorafenib (e.g., BRAFTOVI™, LGX818), naporafenib (LXH254), sorafenib (e.g., sorafenib tosylate), vemurafenib (e.g., ZELBORAF®, RO5185426), ARQ-736, AZ304, BMS-908662 (XL281), C17071479-F, CHIR-265 (RAF265), FORE-8394 (PLX-8394), GDC-0879, GDC-5573 (HM95573), HLX-208, PLX- 3603, PLX-4720, or a combination thereof. In some embodiments, a BRaf V600E mutation can be detected in a sample from the subject (e.g., detecting a BRAF gene having a mutation corresponding to a V600E mutation in BRaf protein and/or detecting a BRaf protein having a V600E mutation). In some embodiments, the subject was determined (e.g., prior to administration of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a))) to have a cancer having a BRaf V600E mutation. In some embodiments, the cancer is a BRaf mutant CRC (e.g., BRaf V600E mutant CRC), and the additional therapy or therapeutic agent is a BRaf inhibitor (e.g., avutometinib (RO5126766), dabrafenib (e.g., dabrafenib mesylate, GSK2118436), encorafenib (e.g., BRAFTOVI™, LGX818), naporafenib (LXH254), sorafenib (e.g., sorafenib tosylate), vemurafenib (e.g., ZELBORAF®, RO5185426), ARQ-736, AZ304, BMS-908662 (XL281), C17071479-F, CHIR-265 (RAF265), FORE-8394 (PLX-8394), GDC-0879, GDC-5573 (HM95573), HLX-208, PLX-3603, or PLX-4720). In some embodiments, the cancer is a BRaf mutant CRC (e.g., BRaf V600E mutant CRC), and the additional therapy or therapeutic agent is a BRaf inhibitor (e.g., avutometinib (RO5126766), dabrafenib (e.g., dabrafenib mesylate, GSK2118436), encorafenib (e.g., BRAFTOVI™, LGX818), naporafenib (LXH254), sorafenib (e.g., sorafenib tosylate), vemurafenib (e.g., ZELBORAF®, RO5185426), ARQ-736, AZ304, BMS-908662 (XL281), C17071479-F, CHIR-265 (RAF265), FORE-8394 (PLX-8394), GDC- 0879, GDC-5573 (HM95573), HLX-208, PLX-3603, or PLX-4720) and an anti-EGFR antibody or anti-EGFR antibody-drug conjugate (e.g., amivantamab (e.g., amivantamab-vmjw, or a biosimilar thereof), cetuximab (e.g., ERBITUX® (cetuximab), or a biosimilar thereof (e.g., CMAB-009, CPGJ-602, or KL-140)), cetuximab sarotalocan (AKALUX® (cetuximab sarotalocan), or a biosimilar thereof), depatuxizumab, duligotuzumab, futuximab, imgatuzumab, modotuximab, necitumumab (e.g., PORTRAZZA® (necitumumab), or a biosimilar thereof), nimotuzumab (e.g., BIOMAb EGFR® (nimotuzumab), or a biosimilar thereof), panitumumab (e.g., VECTIBIX® (panitumumab), or a biosimilar thereof), tomuzotuximab, zalutumumab, EMD-55900, EMD-82633, GC-1118 , HLX-07, ICR-62, SCT- 200, SI-B-001, TAS-0313, or biosimilars thereof). In some embodiments, the cancer is a BRaf mutant CRC (e.g., BRaf V600E mutant CRC), and the additional therapy or therapeutic agent is a BRaf inhibitor (e.g., avutometinib (RO5126766), dabrafenib (e.g., dabrafenib mesylate, GSK2118436), encorafenib (e.g., BRAFTOVI™, LGX818), naporafenib (LXH254), sorafenib (e.g., sorafenib tosylate), vemurafenib (e.g., ZELBORAF®, RO5185426), ARQ-736, AZ304, BMS-908662 (XL281), C17071479-F, CHIR-265 (RAF265), FORE-8394 (PLX-8394), GDC- 0879, GDC-5573 (HM95573), HLX-208, PLX-3603, or PLX-4720) and cetuximab (e.g., ERBITUX® (cetuximab), or a biosimilar thereof (e.g., CMAB-009, CPGJ-602, or KL-140)). In some embodiments, the cancer is a BRaf mutant CRC (e.g., BRaf V600E mutant CRC), and the additional therapy or therapeutic agent is a BRaf inhibitor (e.g., avutometinib (RO5126766), dabrafenib (e.g., dabrafenib mesylate, GSK2118436), encorafenib (e.g., BRAFTOVI™, LGX818), naporafenib (LXH254), sorafenib (e.g., sorafenib tosylate), vemurafenib (e.g., ZELBORAF®, RO5185426), ARQ-736, AZ304, BMS-908662 (XL281), C17071479-F, CHIR-265 (RAF265), FORE-8394 (PLX-8394), GDC-0879, GDC-5573 (HM95573), HLX-208, PLX-3603, or PLX-4720) and panitumumab (e.g., VECTIBIX® (panitumumab), or a biosimilar thereof). In some embodiments, the cancer is a BRaf mutant CRC (e.g., BRaf V600E mutant CRC), and the additional therapy or therapeutic agent is a dabrafenib (e.g., dabrafenib mesylate, GSK2118436), encorafenib (e.g., BRAFTOVI™, LGX818), or vemurafenib (e.g., ZELBORAF®, RO5185426) and cetuximab (e.g., ERBITUX® (cetuximab), or a biosimilar thereof (e.g., CMAB-009, CPGJ-602, or KL-140)). In some embodiments, the cancer is a BRaf mutant CRC (e.g., BRaf V600E mutant CRC), and the additional therapy or therapeutic agent is dabrafenib (e.g., dabrafenib mesylate, GSK2118436), encorafenib (e.g., BRAFTOVI™, LGX818), or vemurafenib (e.g., ZELBORAF®, RO5185426) and panitumumab (e.g., VECTIBIX® (panitumumab), or a biosimilar thereof). In some embodiments, the cancer is a BRaf mutant CRC (e.g., BRaf V600E mutant CRC), and the additional therapy or therapeutic agent is a dabrafenib (e.g., dabrafenib mesylate, GSK2118436) and cetuximab (e.g., ERBITUX® (cetuximab), or a biosimilar thereof (e.g., CMAB-009, CPGJ-602, or KL-140)). In some embodiments, the cancer is a BRaf mutant CRC (e.g., BRaf V600E mutant CRC), and the additional therapy or therapeutic agent is dabrafenib (e.g., dabrafenib mesylate, GSK2118436) and panitumumab (e.g., VECTIBIX® (panitumumab), or a biosimilar thereof). In some embodiments, the cancer is a BRaf mutant NSCLC (e.g., BRaf V600E mutant NSCLC), and the additional therapy or therapeutic agent is a BRaf inhibitor (e.g., avutometinib (RO5126766), dabrafenib (e.g., dabrafenib mesylate, GSK2118436), encorafenib (e.g., BRAFTOVI™, LGX818), naporafenib (LXH254), sorafenib (e.g., sorafenib tosylate), vemurafenib (e.g., ZELBORAF®, RO5185426), ARQ-736, AZ304, BMS-908662 (XL281), C17071479-F, CHIR-265 (RAF265), FORE-8394 (PLX-8394), GDC-0879, GDC-5573 (HM95573), HLX-208, PLX-3603, or PLX-4720) and a MEK inhibitor (e.g., avutometinib, binimetinib, cobimetinib (e.g., cobimetinib fumarate), mirdametinib, pimasertib, refametinib, selumetinib (e.g., selumetinib sulfate), trametinib (e.g., trametinib dimethyl sulfoxide), zapnometinib, FCN-159, GSK-1120212, NFX-179, or TAK-733). In some embodiments, the cancer is a BRaf mutant NSCLC (e.g., BRaf V600E mutant NSCLC), and the additional therapy or therapeutic agent is dabrafenib (e.g., dabrafenib mesylate, GSK2118436), encorafenib (e.g., BRAFTOVI™, LGX818), or vemurafenib (e.g., ZELBORAF®, RO5185426), and binimetinib, cobimetinib (e.g., cobimetinib fumarate), selumetinib (e.g., selumetinib sulfate), or trametinib (e.g., trametinib dimethyl sulfoxide). In some embodiments, the cancer is a BRaf mutant NSCLC (e.g., BRaf V600E mutant NSCLC), and the additional therapy or therapeutic agent is dabrafenib (e.g., dabrafenib mesylate, GSK2118436) and trametinib (e.g., trametinib dimethyl sulfoxide). In some embodiments, the cancer is a BRaf mutant NSCLC (e.g., BRaf V600E mutant NSCLC), and the additional therapy or therapeutic agent is vemurafenib (e.g., ZELBORAF®, RO5185426), and cobimetinib (e.g., cobimetinib fumarate). In some embodiments, the cancer is a BRaf mutant NSCLC (e.g., BRaf V600E mutant NSCLC), and the additional therapy or therapeutic agent is encorafenib (e.g., BRAFTOVI™, LGX818) and binimetinib. In some embodiments, the cancer is a BRaf mutant melanoma (e.g., BRaf V600E mutant melanoma), and the additional therapy or therapeutic agent is a BRaf inhibitor (e.g., avutometinib (RO5126766), dabrafenib (e.g., dabrafenib mesylate, GSK2118436), encorafenib (e.g., BRAFTOVI™, LGX818), naporafenib (LXH254), sorafenib (e.g., sorafenib tosylate), vemurafenib (e.g., ZELBORAF®, RO5185426), ARQ-736, AZ304, BMS-908662 (XL281), C17071479-F, CHIR-265 (RAF265), FORE-8394 (PLX-8394), GDC-0879, GDC-5573 (HM95573), HLX-208, PLX-3603, or PLX-4720). In some embodiments, the cancer is a BRaf mutant melanoma (e.g., BRaf V600E mutant melanoma), and the additional therapy or therapeutic agent is a BRaf inhibitor (e.g., avutometinib (RO5126766), dabrafenib (e.g., dabrafenib mesylate, GSK2118436), encorafenib (e.g., BRAFTOVI™, LGX818), naporafenib (LXH254), sorafenib (e.g., sorafenib tosylate), vemurafenib (e.g., ZELBORAF®, RO5185426), ARQ-736, AZ304, BMS-908662 (XL281), C17071479-F, CHIR-265 (RAF265), FORE-8394 (PLX-8394), GDC-0879, GDC-5573 (HM95573), HLX-208, PLX- 3603, or PLX-4720) and a MEK inhibitor (e.g., avutometinib, binimetinib, cobimetinib (e.g., cobimetinib fumarate), mirdametinib, pimasertib, refametinib, selumetinib (e.g., selumetinib sulfate), trametinib (e.g., trametinib dimethyl sulfoxide), zapnometinib, FCN-159, GSK- 1120212, NFX-179, or TAK-733). In some embodiments, the cancer is a BRaf mutant melanoma (e.g., BRaf V600E mutant melanoma), and the additional therapy or therapeutic agent is dabrafenib (e.g., dabrafenib mesylate, GSK2118436), encorafenib (e.g., BRAFTOVI™, LGX818), or vemurafenib (e.g., ZELBORAF®, RO5185426), and binimetinib, cobimetinib (e.g., cobimetinib fumarate), selumetinib (e.g., selumetinib sulfate), or trametinib (e.g., trametinib dimethyl sulfoxide). In some embodiments, the cancer is a BRaf mutant melanoma (e.g., BRaf V600E mutant melanoma), and the additional therapy or therapeutic agent is dabrafenib (e.g., dabrafenib mesylate, GSK2118436) and trametinib (e.g., trametinib dimethyl sulfoxide). In some embodiments, the cancer is a BRaf mutant melanoma (e.g., BRaf V600E mutant melanoma), and the additional therapy or therapeutic agent is vemurafenib (e.g., ZELBORAF®, RO5185426), and cobimetinib (e.g., cobimetinib fumarate). In some embodiments, the cancer is a BRaf mutant melanoma (e.g., BRaf V600E mutant melanoma), and the additional therapy or therapeutic agent is encorafenib (e.g., BRAFTOVI™, LGX818) and binimetinib. In some such embodiments, the subject has previously been treated with an immunotherapy. In some embodiments, the CDK2 inhibitor is ebvaciclib, fadraciclib, milciclib, pacritinib (e.g., pacritinib citrate), roniciclib, roscovitine, BLU-222, NUV-422, PF-07104091, TQB-3616, or a combination thereof. In some embodiments, the CDK4/6 inhibitor is abemaciclib, birociclib, dalpiciclib, lerociclib, milciclib, palbociclib, ribociclib (e.g., ribociclib succinate), riviciclib, roniciclib, trilaciclib (e.g., trilaciclib dihydrochloride), FCN-437, TQB-3616, or a combination thereof. In some embodiments, the CDK7 inhibitor is milciclib, roscovitine, samuraciclib, or a combination thereof. In some embodiments, the CDK9 inhibitor is fadraciclib, riviciclib, roniciclib, roscovitine, zotiraciclib, AZD-4573, KB-0742, or a combination thereof. In some embodiments, the EGFR inhibitor is abivertinib, afatinib (e.g., afatinib dimaleate), alflutinib (e.g., alflutinib mesylate), almonertinib (e.g., almonertinib mesylate), befotertinib, brigatinib, canertinib, dacomitinib (e.g., dacomitinib monohydrate), dovitinib, erlotinib (e.g., erlotinib hydrochloride), gefitinib, icotinib, lapatinib (e.g., lapatinib ditosylate monohydrate), larotinib, lazertinib, limertinib, mobocertinib (e.g., mobocertinib succinate), nazartinib, neratinib (e.g., neratinib maleate), olmutinib, osimertinib (e.g., osimertinib mesylate), pelitinib, poziotinib, pyrotinib (e.g., pyrotinib maleate), ruserontinib (SKLB-1028), sapitinib, sunvozertinib, tesevatinib, vandetanib, varlitinib, zorifertinib, BIBW-2948, BPI- 7711, HA-121-28, SH-1028, an anti-EGFR antibody or anti-EGFR antibody-drug conjugate, or a combination thereof. In some embodiments, the anti-EGFR antibody or anti-EGFR antibody-drug conjugate is amivantamab (e.g., amivantamab-vmjw, or a biosimilar thereof), cetuximab (e.g., ERBITUX® (cetuximab), or a biosimilar thereof (e.g., CMAB-009, CPGJ-602, or KL-140)), cetuximab sarotalocan (AKALUX® (cetuximab sarotalocan), or a biosimilar thereof), depatuxizumab, duligotuzumab, futuximab, imgatuzumab, modotuximab, necitumumab (e.g., PORTRAZZA® (necitumumab), or a biosimilar thereof), nimotuzumab (e.g., BIOMAb EGFR® (nimotuzumab), or a biosimilar thereof), panitumumab (e.g., VECTIBIX® (panitumumab), or a biosimilar thereof), tomuzotuximab, zalutumumab, EMD-55900, EMD- 82633, GC-1118 , HLX-07, ICR-62, SCT-200, SI-B-001, TAS-0313, biosimilars thereof, or a combination thereof. In some embodiments, the anti-EGFR antibody or anti-EGFR antibody- drug conjugates is amivantamab (e.g., amivantamab-vmjw, or a biosimilar thereof), cetuximab (e.g., ERBITUX® (cetuximab), or a biosimilar thereof (e.g., CMAB-009, CPGJ-602, or KL- 140)), cetuximab sarotalocan (AKALUX® (cetuximab sarotalocan), or a biosimilar thereof), depatuxizumab, duligotuzumab, futuximab, imgatuzumab, modotuximab, necitumumab (e.g., PORTRAZZA® (necitumumab), or a biosimilar thereof), nimotuzumab (e.g., BIOMAb EGFR® (nimotuzumab), or a biosimilar thereof), panitumumab (e.g., VECTIBIX® (panitumumab), or a biosimilar thereof), tomuzotuximab, zalutumumab, EMD-55900, EMD- 82633, GC-1118 , HLX-07 , ICR-62, SCT-200, SI-B-001, biosimilars thereof, or a combination thereof. In some embodiments, an EGFR mutation (e.g., an EGFR exon 19 deletion or an EGFR L858R mutation (with or without an EGFR T790M mutation)) can be detected in a sample from the subject (e.g., detecting an EGFR gene having a mutation (e.g., a mutation corresponding to an EGFR exon 19 deletion or an EGFR L858R mutation (with or without an EGFR T790M mutation) in an EGFR protein) and/or detecting an EGFR protein having a mutation (e.g., an EGFR exon 19 deletion or an EGFR L858R mutation (with or without an EGFR T790M mutation))). In some embodiments, the subject was determined (e.g., prior to administration of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I- B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I- Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a))) to have a cancer having an EGFR mutation (e.g., an EGFR exon 19 deletion or an EGFR L858R mutation (with or without an EGFR T790M mutation)). In some embodiments, the cancer is an EGFR mutant NSCLC (e.g., EGFR exon 19 deletion NSCLC or EGFR L858R (with or without T790M) mutant NCLC), and the additional therapy or therapeutic agent is an EGFR inhibitor (e.g., abivertinib, afatinib (e.g., afatinib dimaleate), alflutinib (e.g., alflutinib mesylate), almonertinib (e.g., almonertinib mesylate), befotertinib, brigatinib, canertinib, dacomitinib (e.g., dacomitinib monohydrate), dovitinib, erlotinib (e.g., erlotinib hydrochloride), gefitinib, icotinib, lapatinib (e.g., lapatinib ditosylate monohydrate), larotinib, lazertinib, limertinib, mobocertinib (e.g., mobocertinib succinate), nazartinib, neratinib (e.g., neratinib maleate), olmutinib, osimertinib (e.g., osimertinib mesylate), pelitinib, poziotinib, pyrotinib (e.g., pyrotinib maleate), ruserontinib (SKLB-1028), sapitinib, sunvozertinib, tesevatinib, vandetanib, varlitinib, zorifertinib, BIBW-2948, BPI- 7711, HA-121-28, SH-1028, or an anti-EGFR antibody or anti-EGFR antibody-drug conjugate). In some embodiments, the cancer is an EGFR mutant NSCLC (e.g., EGFR exon 19 deletion NSCLC or EGFR L858R (with or without T790M) mutant NCLC), and the additional therapy or therapeutic agent is osimertinib (e.g., osimertinib mesylate). In some embodiments, the ERK inhibitor is rineterkib, temuterkib, ulixertinib, ASN- 0007, ASTX-029, ATG-017, BPI-27336, HH-2710, JSI-1187, MK-8353, or a combination thereof. In some embodiments, the EZH2 inhibitor is lirametostat, tazemetostat (e.g., tazemetostat hydrobromide), valemetostat (e.g., valemetostat tosylate), tulmimetostat (CPI- 0209), EBI-2511, HH-2853, HM-97662, PF-6821497, SHR-2554, XNW-5004, or a combination thereof. In some embodiments, the EZH2 inhibitor also inhibits EZH1 (also referred to as an EZH1/2 inhibitor). In some embodiments, the cancer is peripheral T cell lymphoma, and the additional therapy or therapeutic agent is an EZH2 inhibitor (e.g., lirametostat, tazemetostat (e.g., tazemetostat hydrobromide), valemetostat (e.g., valemetostat tosylate), tulmimetostat (CPI- 0209), EBI-2511, HH-2853, HM-97662, PF-6821497, SHR-2554, or XNW-5004). In some embodiments, the FGFR1 inhibitor is danusertib, dovitinib, erdafitinib, futibatinib, infigratinib (e.g., infigratinib phosphate), lenvatinib (e.g., lenvatinib mesylate), lucitanib, nintedanib (e.g., nintedanib esylate), pemigatinib, surufatinib, tasurgratinib, tinengotinib, zoligratinib, FH-2001, HMPL-453, LY-2874455, or a combination thereof. In some embodiments, the FGFR2 inhibitor is erdafitinib, futibatinib, infigratinib (e.g., infigratinib phosphate), lucitanib, pemigatinib, tasurgratinib, zoligratinib, bemarituzumab (or biosimilars thereof), FH-2001, HMPL-453, LY-2874455, or a combination thereof. In some embodiments, the FGFR3 inhibitor is dovitinib, erdafitinib, futibatinib, infigratinib (e.g., infigratinib phosphate), lucitanib, masitinib, nintedanib, pemigatinib, tasurgratinib, zoligratinib, vofatamab (or biosimilars thereof), EXEL-0999, FH-2001, HMPL- 453, LY-2874455, or a combination thereof. In some embodiments, the FGFR4 inhibitor is axitinib, erdafitinib, futibatinib, infigratinib (e.g., infigratinib phosphate), irpagratinib, nintedanib, pemigatinib, FH-2001, H3B-6527, LY-2874455, or a combination thereof. In some embodiments, the HER2 inhibitor is afatinib (e.g., afatinib dimaleate), dacomitinib (e.g., dacomitinib monohydrate), lapatinib (e.g., lapatinib ditosylate monohydrate), mobocertinib (e.g., mobocertinib succinate), neratinib (e.g., neratinib maleate), poziotinib, pyrotinib (e.g., pyrotinib maleate), sunvozertinib, tesevatinib, tucatinib, varlitinib, an anti-HER2 antibody or anti-HER2 antibody-drug conjugate, or a combination thereof. In some embodiments, the anti-HER2 antibody or anti-HER2 antibody-drug conjugate is anbenitamab, cinrebafusp alfa, coprelotamab, disitamab vedotin, ertumaxomab, gancotamab, inetetamab, margetuximab (e.g., margetuximab-cmkb, or a biosimilar thereof), pertuzumab (e.g., PERJETA® (pertuzumab), or a biosimilar thereof (e.g., HLX-11)), trastuzumab (e.g., HERCEPTIN® (trastuzumab), or a biosimilar thereof (e.g., FACEPTOR® (trastuzumab), HERTICAD® (trastuzumab), TUZNUE® (trastuzumab), ZERCEPAC® (trastuzumab), trastuzumab-anns, trastuzumab-dkst, trastuzumab-dttb, trastuzumab-pkrb, trastuzumab-qyyp, EG-12014, or TX-05)), trastuzumab deruxtecan (e.g., fam-trastuzumab deruxtecan-nxki, or a biosimilar thereof), trastuzumab duocarmazine, trastuzumab emtansine (e.g., KADCYLA® (trastuzumab emtansine), or a biosimilar thereof (e.g., UJVIRA® (trastuzumab emtansine))), trastuzumab hyaluronidase (e.g., trastuzumab hyaluronidase-oysk, or a biosimilar thereof), zanidatamab, zenocutuzumab, AVX-901, IDM-1, TPIV-100, TAA-013, SHR-A1811, BAT- 8001, MDX-210, Alpha-Her2-pAF1-AS-269, MRG-002, DF-1001, AC-101, MM-111, biosimilars thereof, or a combination thereof. In some embodiments, the anti-HER2 antibody or anti-HER2 antibody-drug conjugate is anbenitamab, cinrebafusp alfa, coprelotamab, disitamab vedotin, ertumaxomab, gancotamab, inetetamab, margetuximab (e.g., margetuximab-cmkb, or a biosimilar thereof), pertuzumab (e.g., PERJETA® (pertuzumab), or a biosimilar thereof (e.g., HLX-11)), trastuzumab (e.g., HERCEPTIN® (trastuzumab), or a biosimilar thereof (e.g., FACEPTOR® (trastuzumab), HERTICAD® (trastuzumab), TUZNUE® (trastuzumab), ZERCEPAC® (trastuzumab), trastuzumab-anns, trastuzumab- dkst, trastuzumab-dttb, trastuzumab-pkrb, trastuzumab-qyyp, EG-12014, or TX-05)), trastuzumab deruxtecan (e.g., fam-trastuzumab deruxtecan-nxki, or a biosimilar thereof), trastuzumab duocarmazine, trastuzumab emtansine (e.g., KADCYLA® (trastuzumab emtansine), or a biosimilar thereof (e.g., UJVIRA® (trastuzumab emtansine))), trastuzumab hyaluronidase (e.g., trastuzumab hyaluronidase-oysk, or a biosimilar thereof), zanidatamab, zenocutuzumab, TAA-013, SHR-A1811, BAT-8001, MDX-210, Alpha-Her2-pAF1-AS-269, MRG-002, DF-1001, AC-101, MM-111, biosimilars thereof, or a combination thereof. In some embodiments, HER2+ status can be detected in a sample from the subject (e.g., via immunohistochemistry (IHC) and/or fluorescent in situ hybridization (FISH)). In some embodiments, the subject was determined (e.g., prior to administration of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a))) to have a cancer with HER2+ status. In some embodiments, HER2 low status can be detected in a sample from the subject (e.g., via IHC and/or FISH). In some embodiments, the subject was determined (e.g., prior to administration of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I- B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I- Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a))) to have a cancer with HER2 low status. In some embodiments, HER2- status can be detected in a sample from the subject (e.g., via IHC and/or FISH). In some embodiments, the subject was determined (e.g., prior to administration of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I- B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I- Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a))) to have a cancer with HER2- status. In some embodiments, ER expression status can be detected in a sample from the subject (e.g., via IHC and/or FISH). In some embodiments, the subject was determined (e.g., prior to administration of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I- A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I- E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb- 3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I- H-3))) or Formula (II) (e.g., (II-a))) to have a cancer with ER expression. In some embodiments, the subject was determined (e.g., prior to administration of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a))) to have a cancer without ER expression. In some embodiments, PR expression status can be detected in a sample from the subject (e.g., via IHC, and/or FISH). In some embodiments, the subject was determined (e.g., prior to administration of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I- A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I- E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb- 3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I- H-3))) or Formula (II) (e.g., (II-a))) to have a cancer with PR expression. In some embodiments, the subject was determined (e.g., prior to administration of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a))) to have a cancer without PR expression. In some embodiments, the cancer is a HER2+ breast cancer (e.g., HER2+ breast cancer with ER expression, HER2+ breast cancer without ER expression), and the additional therapy is a HER2 inhibitor (e.g., afatinib (e.g., afatinib dimaleate), dacomitinib (e.g., dacomitinib monohydrate), lapatinib (e.g., lapatinib ditosylate monohydrate), mobocertinib (e.g., mobocertinib succinate), neratinib (e.g., neratinib maleate), poziotinib, pyrotinib (e.g., pyrotinib maleate), sunvozertinib, tesevatinib, tucatinib, varlitinib, or an anti-HER2 antibody or anti-HER2 antibody-drug conjugate). In some embodiments, the cancer is a HER2+ breast cancer (e.g., HER2+ breast cancer with ER expression, HER2+ breast cancer without ER expression), and the additional therapy is tucatinib. In some embodiments, the cancer is a HER2+ breast cancer (e.g., HER2+ breast cancer with ER expression, HER2+ breast cancer without ER expression), and the additional therapy is an anti-HER2 antibody or anti-HER2 antibody-drug conjugate (e.g., anbenitamab, cinrebafusp alfa, coprelotamab, disitamab vedotin, ertumaxomab, gancotamab, inetetamab, margetuximab (e.g., margetuximab-cmkb, or a biosimilar thereof), pertuzumab (e.g., PERJETA® (pertuzumab), or a biosimilar thereof (e.g., HLX-11)), trastuzumab (e.g., HERCEPTIN® (trastuzumab), or a biosimilar thereof (e.g., FACEPTOR® (trastuzumab), HERTICAD® (trastuzumab), TUZNUE® (trastuzumab), ZERCEPAC® (trastuzumab), trastuzumab-anns, trastuzumab-dkst, trastuzumab-dttb, trastuzumab-pkrb, trastuzumab-qyyp, EG-12014, or TX-05)), trastuzumab deruxtecan (e.g., fam-trastuzumab deruxtecan-nxki, or a biosimilar thereof), trastuzumab duocarmazine, trastuzumab emtansine (e.g., KADCYLA® (trastuzumab emtansine), or a biosimilar thereof (e.g., UJVIRA® (trastuzumab emtansine))), trastuzumab hyaluronidase (e.g., trastuzumab hyaluronidase-oysk, or a biosimilar thereof), zanidatamab, zenocutuzumab, AVX-901, IDM- 1, TPIV-100, TAA-013, SHR-A1811, BAT-8001, MDX-210, Alpha-Her2-pAF1-AS-269, MRG-002, DF-1001, AC-101, MM-111, or biosimilars thereof). In some embodiments, the cancer is a HER2+ breast cancer (e.g., HER2+ breast cancer with ER expression, HER2+ breast cancer without ER expression), and the additional therapy is trastuzumab (e.g., HERCEPTIN® (trastuzumab), or a biosimilar thereof (e.g., FACEPTOR® (trastuzumab), HERTICAD® (trastuzumab), TUZNUE® (trastuzumab), ZERCEPAC® (trastuzumab), trastuzumab-anns, trastuzumab-dkst, trastuzumab-dttb, trastuzumab-pkrb, trastuzumab-qyyp, EG-12014, or TX- 05)). In some embodiments, the cancer is a HER2+ breast cancer (e.g., HER2+ breast cancer with ER expression, HER2+ breast cancer without ER expression), and the additional therapy is trastuzumab deruxtecan (e.g., fam-trastuzumab deruxtecan-nxki, or a biosimilar thereof). In some embodiments, the cancer is a HER2+ breast cancer (e.g., HER2+ breast cancer with ER expression, HER2+ breast cancer without ER expression), and the additional therapy is trastuzumab emtansine, or a biosimilar thereof. In some embodiments, the cancer is a HER2 low breast cancer, and the additional therapy is an anti-HER2 antibody or anti-HER2 antibody- drug conjugate. In some embodiments, the cancer is a HER2 low breast cancer, and the additional therapy is trastuzumab deruxtecan (e.g., fam-trastuzumab deruxtecan-nxki, or a biosimilar thereof). In some embodiments, the JAK inhibitor is adelatinib, baricitinib, brepocitinib, deuruxolitinib, fedratinib (e.g., fedratinib dihydrochloride monohydrate), filgotinib (e.g., filgotinib maleate), gandotinib, gusacitinib, ilginatinib, izencitinib, jaktinib, momelotinib (e.g., momelotinib dihydrochloride), nezulcitinib, pacritinib (e.g., pacritinib citrate), peficitinib (e.g., peficitinib hydrobromide), ropsacitinib, ruxolitinib (e.g., ruxolitinib phosphate), tasocitinib (e.g., tofacitinib citrate), AT-9283, TQ-05105, or a combination thereof. In some embodiments, the JAK inhibitor is fedratinib (e.g., fedratinib dihydrochloride monohydrate), momelotinib (e.g., momelotinib dihydrochloride), pacritinib (e.g., pacritinib citrate), ruxolitinib (e.g., ruxolitinib phosphate), or a combination thereof. In some embodiments, the JAK inhibitor is fedratinib (e.g., fedratinib dihydrochloride monohydrate). In some embodiments, the JAK inhibitor is momelotinib (e.g., momelotinib dihydrochloride). In some embodiments, the JAK inhibitor is pacritinib (e.g., pacritinib citrate). In some embodiments, the JAK inhibitor is ruxolitinib (e.g., ruxolitinib phosphate). In some embodiments, a JAK V617F mutation can be detected in a sample from the subject (e.g., detecting a JAK2 gene having a mutation corresponding to a V617F mutation in JAK2 protein and/or detecting a JAK2 protein having a V617F mutation). In some embodiments, the subject was determined (e.g., prior to administration of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a))) to have a cancer having a JAK2 V617F mutation. In some embodiments, the subject was determined (e.g., prior to administration of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A- 1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E- 1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I- Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a))) to have a cancer lacking a JAK2 V617F mutation. In some embodiments, the cancer is post-MPN AML, M6-AML, or M7-AML, and the additional therapy or therapeutic agent is a JAK inhibitor (e.g., ruxolitinib (e.g., ruxolitinib phosphate), fedratinib (e.g., fedratinib dihydrochloride monohydrate), momelotinib (e.g., momelotinib dihydrochloride), or pacritinib (e.g., pacritinib citrate)). In some embodiments, the patient has received a previous line of therapy including a JAK inhibitor. In some embodiments, the patient has not received a previous line of therapy including a JAK inhibitor. In some embodiments, the cancer is intermediate (e.g., intermediate-1 and/or intermediate-2) or high-risk myelofibrosis (e.g., primary myelofibrosis, post-essential thrombocythemia myelofibrosis, or post-polycythemia vera myelofibrosis), and the additional therapy or therapeutic agent is a JAK inhibitor (e.g., ruxolitinib (e.g., ruxolitinib phosphate), fedratinib (e.g., fedratinib dihydrochloride monohydrate), momelotinib (e.g., momelotinib dihydrochloride), or pacritinib (e.g., pacritinib citrate)). In some embodiments, the cancer is intermediate (e.g., intermediate-1 and/or intermediate-2) or high-risk myelofibrosis (e.g., primary myelofibrosis, post-essential thrombocythemia myelofibrosis, or post-polycythemia vera myelofibrosis), and the additional therapy or therapeutic agent is a JAK inhibitor (e.g., ruxolitinib (e.g., ruxolitinib phosphate), fedratinib (e.g., fedratinib dihydrochloride monohydrate), momelotinib (e.g., momelotinib dihydrochloride), or pacritinib (e.g., pacritinib citrate)) and a BET inhibitor (e.g., alobresib, apabetalone, mivebresib, pelabresib, trotabresib, ABBV-744, BI-2536, BMS-986158, INCB-057643, JAB-8263, ODM-207, PLX-2853, or ZEN-003694). In some embodiments, the cancer is intermediate (e.g., intermediate-1 and/or intermediate-2) or high-risk myelofibrosis (e.g., primary myelofibrosis, post-essential thrombocythemia myelofibrosis, or post-polycythemia vera myelofibrosis) with a JAK2 mutation (e.g., a JAK2 V617F mutation), and the additional therapy or therapeutic agent is a JAK inhibitor (e.g., ruxolitinib (e.g., ruxolitinib phosphate), fedratinib (e.g., fedratinib dihydrochloride monohydrate), momelotinib (e.g., momelotinib dihydrochloride), or pacritinib (e.g., pacritinib citrate)). In some embodiments, the cancer is intermediate (e.g., intermediate- 1 and/or intermediate-2) or high-risk myelofibrosis (e.g., primary myelofibrosis, post-essential thrombocythemia myelofibrosis, or post-polycythemia vera myelofibrosis) with a JAK2 mutation (e.g., a JAK2 V617F mutation), and the additional therapy or therapeutic agent is a JAK inhibitor (e.g., ruxolitinib (e.g., ruxolitinib phosphate), fedratinib (e.g., fedratinib dihydrochloride monohydrate), momelotinib (e.g., momelotinib dihydrochloride), or pacritinib (e.g., pacritinib citrate)) and a BET inhibitor (e.g., alobresib, apabetalone, mivebresib, pelabresib, trotabresib, ABBV-744, BI-2536, BMS-986158, INCB-057643, JAB-8263, ODM- 207, PLX-2853, or ZEN-003694). In some embodiments, the JAK inhibitor is fedratinib (e.g., fedratinib dihydrochloride monohydrate). In some embodiments, the JAK inhibitor is momelotinib (e.g., momelotinib dihydrochloride). In some embodiments, the JAK inhibitor is pacritinib (e.g., pacritinib citrate). In some embodiments, the JAK inhibitor is ruxolitinib (e.g., ruxolitinib phosphate). In some embodiments, the patient has received a previous line of therapy including a JAK inhibitor. In some embodiments, the patient has not received a previous line of therapy including a JAK inhibitor. In some embodiments, treatment effect can be measured by Spleen Volume Reduction (e.g., Spleen Volume Reduction of greater than or equal to 35% (SVR 35 ), for example, measured by MRI or CT), Total Symptom Score (e.g., Total Symptom Score reduction of greater than or equal to 50% (TSS50), for example, measured by the Myelofibrosis Symptom Assessment Form (MFSAF) version 4.0), or both, such as at 24 weeks after beginning of treatment; see, e.g., Harrison, Claire, et al., New England Journal of Medicine 366.9 (2012): 787-798; and Verstovsek, Srdan, et al. New England Journal of Medicine 366.9 (2012): 799-807. In some embodiments, treatment effect can be measured (e.g., in addition to or instead of SVR35 and/or TSS50) by anemia response (e.g., measured by current International Working Group-Myeloproliferative Neoplasms Research and European LeukemiaNet (IWG-MRT/ELN) criteria), bone marrow fibrosis (e.g., according to the European Consensus Grading System through bone marrow biopsy, such as at 24 or 96 weeks after beginning of treatment), variant allele fraction (e.g., JAK2 V617F variant allele fraction), transfusion independence, overall survival, leukemia-free survival, change in physical functioning (e.g., measured by the physical functioning domain of the European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ)-C30 or death), change in fatigue (e.g., assessed using the Patient-Reported Outcomes Measurement Information System (PROMIS) Fatigue SF 7a), or a combination thereof. In some embodiments, the KRas inhibitor is adagrasib, divarasib (GDC-6036), sotorasib, ARS-1620, ARS-3248, ARS-853, ASP-3082, ATG-012, BI-1701963, BI-1823911, BPI-421286, D-1553, ERAS-3490, GFH-925, JAB-21822, JDQ-443, LY-3537982, MRTX- 1133, MRTX-1257, RMC-6236, RMC-6291, RSC-1255, or a combination thereof. In some embodiments, a KRas mutation (e.g., a KRas G12C mutation or a KRas G12D mutation) can be detected in a sample from the subject (e.g., detecting a KRAS gene having a mutation corresponding to a G12C mutation or a G12D mutation in KRas protein and/or detecting a KRas protein having a G12C mutation or a G12D mutation). In some embodiments, the subject was determined (e.g., prior to administration of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a))) to have a cancer having a KRas G12C mutation. In some embodiments, the subject was determined (e.g., prior to administration of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I- B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I- Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a))) to have a cancer having a KRas G12D mutation. In some embodiments, the cancer is a KRas mutant lung cancer (e.g., a KRas mutant NSCLC), a KRas mutant CRC, or a KRas mutant pancreatic cancer, and the additional therapy or therapeutic agent is a KRas inhibitor (e.g., adagrasib, divarasib (GDC-6036), sotorasib, ARS-1620, ARS-3248, ARS-853, ASP-3082, ATG-012, BI-1701963, BI-1823911, BPI- 421286, D-1553, ERAS-3490, GFH-925, JAB-21822, JDQ-443, LY-3537982, MRTX-1133, MRTX-1257, RMC-6236, RMC-6291, or RSC-1255). In some embodiments, the cancer is a KRas mutant lung cancer (e.g., a KRas mutant NSCLC (e.g., a KRas G12C mutant NSCLC)), a KRas mutant CRC (e.g., a KRas G12C mutant CRC), or a KRas mutant pancreatic cancer (e.g., a KRas G12C mutant pancreatic cancer), and the additional therapy or therapeutic agent is adagrasib. In some embodiments, the cancer is a KRas mutant lung cancer (e.g., a KRas mutant NSCLC (e.g., a KRas G12C mutant NSCLC)), a KRas mutant CRC (e.g., a KRas G12C mutant CRC), or a KRas mutant pancreatic cancer (e.g., a KRas G12C mutant pancreatic cancer), and the additional therapy or therapeutic agent is divarasib. In some embodiments, the cancer is a KRas mutant lung cancer (e.g., a KRas mutant NSCLC (e.g., a KRas G12C mutant NSCLC)), a KRas mutant CRC (e.g., a KRas G12C mutant CRC), or a KRas mutant pancreatic cancer (e.g., a KRas G12C mutant pancreatic cancer), and the additional therapy or therapeutic agent is sotorasib. In some embodiments, the cancer is a KRas mutant lung cancer (e.g., a KRas mutant NSCLC (e.g., a KRas G12D mutant NSCLC)), a KRas mutant CRC (e.g., a KRas G12D mutant CRC), or a KRas mutant pancreatic cancer (e.g., a KRas G12D mutant pancreatic cancer), and the additional therapy or therapeutic agent is MRTX1133. In some embodiments, the MEK inhibitor is avutometinib, binimetinib, cobimetinib (e.g., cobimetinib fumarate), mirdametinib, pimasertib, refametinib, selumetinib (e.g., selumetinib sulfate), trametinib (e.g., trametinib dimethyl sulfoxide), zapnometinib, FCN-159, GSK-1120212, NFX-179, TAK-733, or a combination thereof. In some embodiments, a BRCA1 mutation can be detected in a sample from the subject (e.g., detecting a BRCA1 gene having a mutation and/or detecting a BRCA1 protein having a mutation). In some embodiments, the subject was determined (e.g., prior to administration of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea- 1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I- F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a))) to have a cancer having a BRCA1 mutation. In some embodiments, a BRCA2 mutation can be detected in a sample from the subject (e.g., detecting a BRCA2 gene having a mutation and/or detecting a BRCA2 protein having a mutation). In some embodiments, the subject was determined (e.g., prior to administration of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea- 1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I- F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a))) to have a cancer having a BRCA2 mutation. In some embodiments, the cancer is ovarian cancer (e.g., BRCA1 mutant ovarian cancer or BRCA2 mutant ovarian cancer, HGSOC (e.g., BRCA1 mutant HGSOC or BRCA2 mutant HGSOC), or LGSOC), and the additional therapy or therapeutic agent is a MEK inhibitor (e.g., binimetinib, cobimetinib, selumetinib, or trametinib). In some embodiments, the cancer is BRCA1 mutant ovarian cancer, and the additional therapy or therapeutic agent is a MEK inhibitor (e.g., binimetinib, cobimetinib, selumetinib, or trametinib). In some embodiments, the cancer is BRCA2 mutant ovarian cancer, and the additional therapy or therapeutic agent is a MEK inhibitor (e.g., binimetinib, cobimetinib, selumetinib, or trametinib). In some embodiments, the cancer is HGSOC (e.g., BRCA1 mutant HGSOC or BRCA2 mutant HGSOC), and the additional therapy or therapeutic agent is a MEK inhibitor (e.g., binimetinib, cobimetinib, selumetinib, or trametinib). In some embodiments, the cancer is LGSOC (e.g., BRCA1 mutant HGSOC or BRCA2 mutant HGSOC), and the additional therapy or therapeutic agent is a MEK inhibitor (e.g., binimetinib, cobimetinib, selumetinib, or trametinib). In some embodiments, the cancer is a KRas mutant CRC (e.g., a KRas G12C mutant NSCLC), and the additional therapy or therapeutic agent is a MEK inhibitor (e.g., binimetinib, cobimetinib, selumetinib, or trametinib). In some embodiments, the MET inhibitor is cabozantinib (e.g., cabozantinib S-malate), capmatinib (e.g., capmatinib hydrochloride), crizotinib, foritinib, glesatinib, gumarontinib, merestinib, pamufetinib, savolitinib, sitravatinib, tepotinib (e.g., tepotinib hydrochloride hydrate), vebreltinib, zanzalintinib (XL-092), amivantamab (e.g., amivantamab-vmjw, or a biosimilar thereof), emibetuzumab (or biosimilars thereof), RC-108, telisotuzumab vedotin (or biosimilars thereof), ABBV-400, ABN-401, AL-2846, AMG-337, SAR-125844, TQ-B-3139, or a combination thereof. In some embodiments, a MET alteration can be detected in a sample from the subject (e.g., detecting a MET gene having an alteration (e.g., gene amplification or exon14 skipping) and/or detecting a MET protein having a mutation (e.g., exon14 skipping)). In some embodiments, the subject was determined (e.g., prior to administration of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I- B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F- 3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a))) to have a cancer having a MET alteration. In some embodiments, the cancer is a MET-altered NCLC (e.g., MET amplified NSCLC or MET exon14 skipping NSCLC), and the additional therapy or therapeutic agent is a MET inhibitor (e.g., cabozantinib (e.g., cabozantinib S-malate), capmatinib (e.g., capmatinib hydrochloride), crizotinib, foritinib, glesatinib, gumarontinib, merestinib, pamufetinib, savolitinib, sitravatinib, tepotinib (e.g., tepotinib hydrochloride hydrate), vebreltinib, zanzalintinib (XL-092), amivantamab (e.g., amivantamab-vmjw, or a biosimilar thereof), emibetuzumab (or biosimilars thereof), RC-108, telisotuzumab vedotin (or biosimilars thereof), ABBV-400, ABN-401, AL-2846, AMG-337, SAR-125844, or TQ-B-3139). In some embodiments, the cancer is a MET-altered NCLC (e.g., MET amplified NSCLC or MET exon14 skipping NSCLC), and the additional therapy or therapeutic agent is capmatinib (e.g., capmatinib hydrochloride) or tepotinib (e.g., tepotinib hydrochloride hydrate). In some embodiments, the cancer is a MET-altered NCLC (e.g., MET amplified NSCLC or MET exon14 skipping NSCLC), and the additional therapy or therapeutic agent is telisotuzumab vedotin (or biosimilars thereof). In some embodiments, the Hif2α inhibitor is belzutifan, AB-521, DFF-332, NKT-2152, PT-2399, or a combination thereof. In some embodiments, the PARP inhibitor is fuzuloparib (fluzoparib), niraparib (e.g., niraparib tosylate monohydrate), olaparib, pamiparib, rucaparib (e.g., rucaparib camsylate), saruparib (AZD5305), senaparib, stenoparib, talazoparib (e.g., talazoparib tosylate), veliparib, CEP-9722, JPI-289, NMS-03305293, or a combination thereof. In some embodiments, the PARP inhibitor is a PARP1 inhibitor. In some embodiments, the PARP1 inhibitor is saruparib (AZD5305), NMS-03305293, or a combination thereof. In some embodiments, the cancer is BRCA1 mutant breast cancer or BRCA2 mutant breast cancer, and the additional therapy or therapeutic agent is a PARP inhibitor (e.g., fuzuloparib (fluzoparib), niraparib (e.g., niraparib tosylate monohydrate), olaparib, pamiparib, rucaparib (e.g., rucaparib camsylate), saruparib (AZD5305), senaparib, stenoparib, talazoparib (e.g., talazoparib tosylate), veliparib, CEP-9722, JPI-289, or NMS-03305293). In some embodiments, the cancer is BRCA1 mutant breast cancer or BRCA2 mutant breast cancer, and the additional therapy or therapeutic agent is saruparib. In some embodiments, the cancer is triple negative breast cancer, and the additional therapy or therapeutic agent is a PARP inhibitor (e.g., fuzuloparib (fluzoparib), niraparib (e.g., niraparib tosylate monohydrate), olaparib, pamiparib, rucaparib (e.g., rucaparib camsylate), saruparib (AZD5305), senaparib, stenoparib, talazoparib (e.g., talazoparib tosylate), veliparib, CEP-9722, JPI-289, or NMS-03305293). In some embodiments, the cancer is triple negative breast cancer, and the additional therapy or therapeutic agent is saruparib. In some embodiments, the cancer is HGSOC (e.g., BRCA1 mutant HGSOC or BRCA2 mutant HGSOC), and the additional therapy or therapeutic agent is a PARP inhibitor (e.g., fuzuloparib (fluzoparib), niraparib (e.g., niraparib tosylate monohydrate), olaparib, pamiparib, rucaparib (e.g., rucaparib camsylate), saruparib (AZD5305), senaparib, stenoparib, talazoparib (e.g., talazoparib tosylate), veliparib, CEP-9722, JPI-289, or NMS-03305293). In some embodiments, the cancer is HGSOC (e.g., BRCA1 mutant HGSOC or BRCA2 mutant HGSOC), and the additional therapy or therapeutic agent is saruparib. In some embodiments, the LSD1 inhibitor is bomedemstat, iadademstat, pulrodemstat, seclidemstat (HCI-2577), vafidemstat, GSK-2879552, INCB-059872, JBI-802, or a combination thereof. In some embodiments, the BET inhibitor is alobresib, apabetalone, mivebresib, pelabresib, trotabresib, ABBV-744, BI-2536, BMS-986158, INCB-057643, JAB-8263, ODM- 207, PLX-2853, ZEN-003694, or a combination thereof. In some embodiments, the cancer is myelofibrosis (e.g., primary myelofibrosis, post- essential thrombocythemia myelofibrosis, or post-polycythemia vera myelofibrosis), and the additional therapy or therapeutic agent is a BET inhibitor (e.g., alobresib, apabetalone, mivebresib, pelabresib, trotabresib, ABBV-744, BI-2536, BMS-986158, INCB-057643, JAB- 8263, ODM-207, PLX-2853, or ZEN-003694). In some embodiments, the STING agonist is ulevostinag, ADU-S100, or a combination thereof. In some embodiments, the telomerase inhibitor is imetelstat (e.g., imetelstat sodium). In some embodiments, the TORC1/2 inhibitor is apitolisib, bimiralisib, dactolisib, deforolimus, everolimus, fosciclopirox (e.g., fosciclopirox sodium), gedatolisib, onatasertib, paxalisib, sapanisertib, sirolimus, sodium 2-hydroxylinoleate, temsirolimus, umirolimus, vistusertib, zandelisib, zotarolimus, BI-860585, CC-115, CLL-442, PF-04691502, or a combination thereof. In some embodiments, the VEGFR inhibitor is apatinib, axitinib, cabozantinib (e.g., cabozantinib S-malate), catequentinib (alontinib), cediranib, dovitinib, famitinib, fruquintinib, glesatinib , ibcasertib, ilorasertib, lenvatinib (e.g., lenvatinib mesylate), lucitanib, nintedanib (e.g., nintedanib esylate), pamufetinib, pazopanib (e.g., pazopanib hydrochloride), regorafenib (e.g., regorafenib monohydrate), sitravatinib, sorafenib (e.g., sorafenib tosylate), sunitinib (e.g., sunitinib malate), surufatinib (sulfatinib), telatinib, tinengotinib, tivozanib (e.g., tivozanib hydrochloride monohydrate), vandetanib, vorolanib, zanzalintinib, olinvacimab (or biosimilars thereof), ramucirumab (or biosimilars thereof), CEP-11981, ENMD-2076, ODM-203, or a combination thereof. In some embodiments, the chemotherapy is a platinum complex, a microtubule inhibitor (e.g., a microtubule destabilizer or a microtubule stabilizer), a topoisomerase inhibitor, a hypomethylating agent, or an antibody-drug conjugate including any thereof. In some embodiments, the platinum complex is carboplatin, cisplatin, lobaplatin, miriplatin, oxaliplatin, or a combination thereof. In some embodiments, the microtubule inhibitor is cabazitaxel, colchicine, desoxyepothilone B, docetaxel, eribulin, ixabepilone, nab-paclitaxel, paclitaxel, plinabulin, sabizabulin, tirbanibulin, vinblastine, vinflunine, vinorelbine, or a combination thereof. In some embodiments, the microtubule inhibitor is cabazitaxel, docetaxel, nab-paclitaxel, paclitaxel, or a combination thereof. In some embodiments, the topoisomerase inhibitor is aclarubicin, amsacrine, belotecan, camptothecin, daunorubicin, dexrazoxane, elliptinium, epirubicin, etoposide, gepotidacin, idarubicin, mitoxantrone, nemonoxacin, pirarubicin, pixantrone, razoxane, rubitecan, sobuzoxane, temozolomide, teniposide, topotecan, SN-38, or a combination thereof. In some embodiments, the hypomethylating agent is azacitidine, decitabine, or a combination thereof. In some embodiments, the chemotherapy is a platinum complex and a topoisomerase inhibitor (e.g., cisplatin and etoposide). In some embodiments, the antibody-drug conjugate including the microtubule inhibitor is belantamab mafodotin, brentuximab vedotin, cofetuzumab pelidotin, disitamab vedotin, enfortumab vedotin (e.g., enfortumab vedotin-ejfv, or a biosimilar thereof), mirvetuximab soravtansine (e.g., mirvetuximab soravtansine-gynx, or a biosimilar thereof), polatuzumab vedotin, telisotuzumab vedotin, tisotumab vedotin, trastuzumab emtansine (e.g., ado-trastuzumab emtansine, or a biosimilar thereof), tusamitamab ravtansine, upifitamab rilsodotin, zilovertamab vedotin, Alpha-Her2-pAF1-AS-269, BAT-8001, TAA-013, biosimilars thereof, or a combination thereof. In some embodiments, the antibody-drug conjugate including the microtubule inhibitor is enfortumab vedotin (e.g., enfortumab vedotin-ejfv, or a biosimilar thereof). In some embodiments, the antibody-drug conjugate including the microtubule inhibitor is mirvetuximab soravtansine (e.g., mirvetuximab soravtansine-gynx, or a biosimilar thereof). In some embodiments, the antibody-drug conjugate including the microtubule inhibitor is trastuzumab emtansine (e.g., ado-trastuzumab emtansine, or a biosimilar thereof). In some embodiments, the antibody-drug conjugate including the topoisomerase inhibitor is datopotamab deruxtecan, patritumab deruxtecan, sacituzumab govitecan (e.g., sacituzumab govitecan-hziy, or a biosimilar thereof), trastuzumab deruxtecan (fam-trastuzumab deruxtecan- nxki, or a biosimilar thereof), or a combination thereof. In some embodiments, the antibody- drug conjugate including the topoisomerase inhibitor is sacituzumab govitecan (e.g., sacituzumab govitecan-hziy, or a biosimilar thereof). In some embodiments, the antibody-drug conjugate including the topoisomerase inhibitor is trastuzumab deruxtecan (e.g., fam- trastuzumab deruxtecan-nxki, or a biosimilar thereof). In some embodiments, the cancer is a lung cancer (e.g., NSCLC (e.g., squamous cell carcinoma)), and the additional therapy or therapeutic agent is a microtubule inhibitor (e.g., cabazitaxel, docetaxel, nab-paclitaxel, or paclitaxel). In some embodiments, the cancer is a lung cancer (e.g., NSCLC (e.g., squamous cell carcinoma)), and the additional therapy or therapeutic agent is docetaxel. In some embodiments, the cancer is NSCLC (e.g., NSCLC with a MET amplification), and the additional therapy or therapeutic agent is telisotuzumab vedotin. In some embodiments, the cancer is a lung cancer (e.g., NSCLC), and the additional therapy or therapeutic agent is a platinum complex (e.g., carboplatin, cisplatin, lobaplatin, miriplatin, or oxaliplatin) and anti-PD1 therapy. In some embodiments, the cancer is a lung cancer (e.g., NSCLC), and the additional therapy or therapeutic agent is a platinum complex (e.g., carboplatin, cisplatin, lobaplatin, miriplatin, or oxaliplatin) and anti-PD-L1 therapy. In some embodiments, the cancer is a lung cancer (e.g., NSCLC), and the additional therapy or therapeutic agent is a pemetrexed and anti-PD1 therapy. In some embodiments, the cancer is a lung cancer (e.g., NSCLC), and the additional therapy or therapeutic agent is a pemetrexed and anti-PD-L1 therapy. In some embodiments, the cancer is a lung cancer (e.g., SCLC), and the additional therapy or therapeutic agent is a platinum complex (e.g., carboplatin, cisplatin, lobaplatin, miriplatin, or oxaliplatin) and a topoisomerase inhibitor (e.g., aclarubicin, amsacrine, belotecan, camptothecin, daunorubicin, dexrazoxane, elliptinium, epirubicin, etoposide, gepotidacin, idarubicin, mitoxantrone, nemonoxacin, pirarubicin, pixantrone, razoxane, rubitecan, sobuzoxane, temozolomide, teniposide, topotecan, or SN-38). In some embodiments, the cancer is a lung cancer (e.g., SCLC), and the additional therapy or therapeutic agent is carboplatin and etoposide. In some embodiments, the cancer is a lung cancer (e.g., SCLC), and the additional therapy or therapeutic agent is cisplatin and etoposide. In some embodiments, the cancer is breast cancer (e.g., HER2+ breast cancer (e.g., HER2+ breast cancer with ER expression, HER2+ breast cancer without ER expression), HER2 negative breast cancer (e.g., HER2 negative breast cancer with ER expression, HER2 negative breast cancer without ER expression), triple negative breast cancer, or HER2 low breast cancer), and the additional therapy or therapeutic agent is a microtubule inhibitor (e.g., cabazitaxel, docetaxel, nab-paclitaxel, or paclitaxel). In some embodiments, the cancer is triple negative breast cancer, and the additional therapy or therapeutic agent is a microtubule inhibitor (e.g., cabazitaxel, docetaxel, nab-paclitaxel, or paclitaxel). In some embodiments, the cancer is triple negative breast cancer, and the additional therapy or therapeutic agent is nab-paclitaxel or paclitaxel. In some embodiments, the cancer is breast cancer (e.g., HER2+ breast cancer (e.g., HER2+ breast cancer with ER expression, HER2+ breast cancer without ER expression), HER2 negative breast cancer (e.g., HER2 negative breast cancer with ER expression, HER2 negative breast cancer without ER expression), triple negative breast cancer, or HER2 low breast cancer), and the additional therapy or therapeutic agent is a microtubule inhibitor (e.g., cabazitaxel, docetaxel, nab-paclitaxel, or paclitaxel) and anti-PD1 therapy. In some embodiments, the cancer is breast cancer (e.g., HER2+ breast cancer (e.g., HER2+ breast cancer with ER expression, HER2+ breast cancer without ER expression), HER2 negative breast cancer (e.g., HER2 negative breast cancer with ER expression, HER2 negative breast cancer without ER expression), triple negative breast cancer, or HER2 low breast cancer), and the additional therapy or therapeutic agent is a microtubule inhibitor (e.g., cabazitaxel, docetaxel, nab-paclitaxel, or paclitaxel) and anti-PD-L1 therapy. In some embodiments, the cancer is breast cancer (e.g., HER2+ breast cancer (e.g., HER2+ breast cancer with ER expression, HER2+ breast cancer without ER expression), HER2 negative breast cancer (e.g., HER2 negative breast cancer with ER expression, HER2 negative breast cancer without ER expression), triple negative breast cancer, or HER2 low breast cancer), and the additional therapy or therapeutic agent is capecitabine. In some embodiments, the cancer is breast cancer (e.g., HER2+ breast cancer (e.g., HER2+ breast cancer with ER expression, HER2+ breast cancer without ER expression), HER2 negative breast cancer (e.g., HER2 negative breast cancer with ER expression, HER2 negative breast cancer without ER expression), triple negative breast cancer, or HER2 low breast cancer), and the additional therapy or therapeutic agent is sacituzumab govitecan (e.g., sacituzumab govitecan-hziy, or a biosimilar thereof). In some embodiments, the cancer is breast cancer (e.g., HER2+ breast cancer (e.g., HER2+ breast cancer with ER expression, HER2+ breast cancer without ER expression), HER2 negative breast cancer (e.g., HER2 negative breast cancer with ER expression, HER2 negative breast cancer without ER expression), triple negative breast cancer, or HER2 low breast cancer), and the additional therapy or therapeutic agent is trastuzumab deruxtecan (e.g., fam-trastuzumab deruxtecan-nxki, or a biosimilar thereof). In some embodiments, the cancer is breast cancer (e.g., HER2+ breast cancer (e.g., HER2+ breast cancer with ER expression, HER2+ breast cancer without ER expression), HER2 negative breast cancer (e.g., HER2 negative breast cancer with ER expression, HER2 negative breast cancer without ER expression), triple negative breast cancer, or HER2 low breast cancer), and the additional therapy or therapeutic agent is trastuzumab (e.g., HERCEPTIN® (trastuzumab), or a biosimilar thereof (e.g., FACEPTOR® (trastuzumab), HERTICAD® (trastuzumab), TUZNUE® (trastuzumab), ZERCEPAC® (trastuzumab), trastuzumab-anns, trastuzumab-dkst, trastuzumab-dttb, trastuzumab-pkrb, trastuzumab-qyyp, EG-12014, or TX-05)). In some embodiments, the cancer is hormone receptor positive breast cancer, and the additional therapy or therapeutic agent is hormone therapy (e.g., tamoxifen, toremifene, or a combination thereof). In some embodiments, the cancer is hormone receptor positive breast cancer, and the additional therapy or therapeutic agent is a selective estrogen receptor degrader (SERD) (e.g., fulvestrant, elacestrant, or a combination thereof). In some embodiments, the cancer is a HER2+ breast cancer (e.g., HER2+ breast cancer with ER expression, HER2+ breast cancer without ER expression), and the additional therapy or therapeutic agent is trastuzumab deruxtecan (e.g., fam-trastuzumab deruxtecan-nxki, or a biosimilar thereof) or trastuzumab emtansine (e.g., KADCYLA® (trastuzumab emtansine), or a biosimilar thereof (e.g., UJVIRA® (trastuzumab emtansine))). In some embodiments, the cancer is a HER2 low breast cancer, and the additional therapy or therapeutic agent is trastuzumab deruxtecan (e.g., fam-trastuzumab deruxtecan- nxki, or a biosimilar thereof). In some embodiments, the cancer is a head and neck cancer, and the additional therapy or therapeutic agent is a microtubule inhibitor (e.g., cabazitaxel, docetaxel, nab-paclitaxel, or paclitaxel). In some embodiments, the cancer is a head and neck cancer, and the additional therapy or therapeutic agent is docetaxel. In some embodiments, the cancer is a cervical cancer, and the additional therapy or therapeutic agent is a microtubule inhibitor (e.g., cabazitaxel, docetaxel, nab-paclitaxel, or paclitaxel). In some embodiments, the cancer is an endometrial cancer, and the additional therapy or therapeutic agent is a microtubule inhibitor (e.g., cabazitaxel, docetaxel, nab-paclitaxel, or paclitaxel). In some embodiments, the cancer is a prostate cancer, and the additional therapy or therapeutic agent is a microtubule inhibitor (e.g., cabazitaxel, docetaxel, nab-paclitaxel, or paclitaxel). In some embodiments, the cancer is an ovarian cancer (e.g., HGSOC) and the additional therapy or therapeutic agent is a microtubule inhibitor (e.g., cabazitaxel, docetaxel, nab- paclitaxel, or paclitaxel). In some embodiments, the cancer is an ovarian cancer (e.g., HGSOC) and the additional therapy or therapeutic agent is a microtubule inhibitor (e.g., cabazitaxel, docetaxel, nab-paclitaxel, or paclitaxel) and a platinum complex (e.g., carboplatin, cisplatin, lobaplatin, miriplatin, or oxaliplatin). In some embodiments, the cancer is an ovarian cancer (e.g., HGSOC) and the additional therapy or therapeutic agent is nab-paclitaxel or paclitaxel and carboplatin. In some embodiments, the cancer is a pancreatic cancer, and the additional therapy or therapeutic agent is a microtubule inhibitor (e.g., cabazitaxel, docetaxel, nab-paclitaxel, or paclitaxel). In some embodiments, the cancer is a pancreatic cancer, and the additional therapy or therapeutic agent is a microtubule inhibitor (e.g., cabazitaxel, docetaxel, nab-paclitaxel, or paclitaxel) and gemcitabine. In some embodiments, the cancer is a bladder cancer (e.g., bladder urothelial carcinoma), and the additional therapy or therapeutic agent is enfortumab vedotin (e.g., enfortumab vedotin-ejfv, or a biosimilar thereof). In some embodiments, the cancer is a bladder cancer (e.g., bladder urothelial carcinoma), and the additional therapy or therapeutic agent is sacituzumab govitecan (e.g., sacituzumab govitecan-hziy, or a biosimilar thereof). In some embodiments, the cancer is triple negative breast cancer, and the additional therapy or therapeutic agent is sacituzumab govitecan (e.g., sacituzumab govitecan-hziy, or a biosimilar thereof). In some embodiments, folate receptor positivity status can be detected in a sample from the subject (e.g., via immunohistochemistry (IHC) and/or fluorescent in situ hybridization (FISH)). In some embodiments, the subject was determined (e.g., prior to administration of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea- 1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I- F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a))) to have a cancer that is folate receptor positive. In some embodiments, the cancer is folate receptor positive ovarian cancer (e.g., folate receptor positive HGSOC), and the additional therapy or therapeutic agent is mirvetuximab soravtansine (e.g., mirvetuximab soravtansine-gynx, or a biosimilar thereof). In some embodiments the immunomodulatory imide drug is thalidomide, lenalidomide, pomalidomide, iberdomide, avadomide, CC-99282, or a combination thereof. In some embodiments, the anti-PD1 therapy is balstilimab, budigalimab, cadonilimab, camrelizumab, cemiplimab (e.g., cemiplimab-rwlc, or a biosimilar thereof), cetrelimab, dostarlimab (e.g., dostarlimab-gxly, or a biosimilar thereof), ezabenlimab, geptanolimab, ivonescimab, nivolumab (e.g., OPDIVO® (nivolumab), or a biosimilar thereof), nofazinlimab, pembrolizumab (e.g., KEYTRUDA® (pembrolizumab), or a biosimilar thereof), penpulimab, pidilizumab, pimivalimab, prolgolimab, pucotenlimab, retifanlimab (e.g., retifanlimab-dlwr, or a biosimilar thereof), rilvegostomig, rosnilimab, rulonilimab, sasanlimab, serplulimab, sintilimab (e.g., TYVYT® (sintilimab), or a biosimilar thereof), spartalizumab, tebotelimab, tislelizumab, toripalimab, volrustomig, vudalimab, zimberelimab, QL-1604, HX-009, INCB- 086550, RG-6139, BAT-1306, SG-001, biosimilars thereof, or a combination thereof. In some embodiments, the PD-L1 inhibitor is INCB-086550. In some embodiments, the anti-PD-L1 therapy is adebrelimab, atezolizumab (e.g., TECENTRIQ® (atezolizumab), or a biosimilar thereof), avelumab (e.g., BAVENCIO® (avelumab), or a biosimilar thereof), bintrafusp alfa, cosibelimab, danburstotug, durvalumab (e.g., IMFINZI® (durvalumab), or a biosimilar thereof), envafolimab (e.g., ENWEIDA® (envafolimab), or a biosimilar thereof), erfonrilimab, pacmilimab, socazolimab, sugemalimab (e.g., CEJEMLY® (sugemalimab), or a biosimilar thereof), A-167, APL-502, AUPM-170, BNT-311, SHR-1701, biosimilars thereof, or a combination thereof. In some embodiments, the additional therapy is radiotherapy. In some embodiments, the cancer is head and neck cancer (e.g., head and neck squamous cell carcinoma), and the additional therapy is radiotherapy. In some embodiments, the additional therapy includes a BRaf inhibitor and a MEK inhibitor. For example, the additional therapy can include dabrafenib and trametinib, vemurafenib and cobimetinib, or encorafenib and binimetinib. Exemplary descriptions of agents in combination with BCL-2 family inhibitors can be found in: Hikita, Hayato, et al. Hepatology 52.4 (2010): 1310-1321; Chen, Jun, et al. Molecular Cancer Therapeutics 10.12 (2011): 2340-2349; Inuzuka, Hiroyuki, et al. Nature 471.7336 (2011): 104-109; Wertz, Ingrid E., et al. Nature 471.7336 (2011): 110-114; Tan, Nguyen, et al. Clinical Cancer Research 17.6 (2011): 1394-1404; Wong, Maureen, et al. Molecular Cancer Therapeutics 11.4 (2012): 1026-1035; Corcoran, Ryan B., et al. Cancer Cell 23.1 (2013): 121- 128; Waibel, Michaela, et al. Cell Reports 5.4 (2013): 1047-1059; Frederick, Dennie T., et al. PloS One 9.7 (2014): e101286; Vlahovic, Gordana, et al. Investigational New Drugs 32.5 (2014): 976-984; Faber, Anthony C., et al. Cancer Discovery 4.1 (2014): 42-52; Leverson, Joel D., et al. Science Translational Medicine 7.279 (2015): 279ra40-279ra40; Guo, Jun, et al. PLoS One 10.3 (2015): e0114363; Lheureux, Stéphanie, et al. International Journal of Cancer 136.5 (2015): E340-E350; Zoeller, Jason J., et al. Cancer Research 76.14_Supplement (2016): 4358- 4358; Weeden, Clare E., et al. Oncogene 37.32 (2018): 4475-4488; Lucantoni, Federico, et al. Cell Death & Disease 9.2 (2018): 1-13; Iavarone, Claudia, et al. Molecular Cancer Therapeutics 18.3 (2019): 642-655; Fleury, Hubert, et al. Nature Communications 10.1 (2019): 1-15; Lohard, Steven, et al. Nature Communications 11.1 (2020): 1-16; Bertino, Erin M., et al. Clinical Cancer Research 27.6 (2021): 1604-1611; Guo, Ting, et al. Aging 13.15 (2021): 19750; Puglisi, Martina, et al. Future Oncology 17.21 (2021): 2747-2758; Harrison, Claire N., et al. Journal of Clinical Oncology 40.15 (2022): 1671; Köhler, Jens, et al. Molecular Cancer Therapeutics 20.4 (2021): 641-654; Jaaks, Patricia, et al. Nature 603.7899 (2022): 166-173; Sobol, Benjamin, et al. International Journal of Molecular Sciences 23.14 (2022): 7850; Passamonti, J Clin Oncol 40, (2022) (suppl 16; abstr 7015); Qin, J Clin Oncol 40, (2022) (suppl 16; abstr e20612); Potter, Danielle S., et al. Cancer Research 82.12_Supplement (2022): 3691- 3691; and Shebl, Bassem, et al. Cancer Research 82.12_Supplement (2022): 3888-3888. Also provided herein is a method of treating cancer, comprising administering to a subject in need thereof (a) a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I- A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I- E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb- 3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I- H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, and (b) an additional therapeutic agent, for simultaneous, separate or sequential use for the treatment of cancer, wherein the amounts of the compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A- 2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E- 2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H- 2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, and the additional therapeutic agent are together effective in treating the cancer. In some embodiments, the method comprises administering (c) at least one pharmaceutically acceptable carrier. These additional therapeutic agents may be administered with one or more doses of the compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea- 1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I- F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, or pharmaceutical composition thereof, as part of the same or separate dosage forms, via the same or different routes of administration, and/or on the same or different administration schedules according to standard pharmaceutical practice known to one skilled in the art. In some embodiments, the compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I- Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I- Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, and the additional therapeutic agent are administered simultaneously as separate dosages. In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, and the additional therapeutic agent are administered as separate dosages sequentially in any order, in jointly therapeutically effective amounts, e.g., in daily or intermittently dosages. In some embodiments, the compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb- 1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, and the additional therapeutic agent are administered simultaneously as a combined dosage. Also provided herein is (i) a pharmaceutical combination for treating a cancer in a subject in need thereof, which comprises (a) a compound of Formula (I) (e.g., (I-A) (e.g., (I- A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I- E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H- 1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, and (b) at least one additional therapeutic agent (e.g., any of the exemplary additional therapeutic agents described herein or known in the art), for simultaneous, separate or sequential use for the treatment of cancer, wherein the amounts of the compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I- C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea- 3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I- G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, and of the additional therapeutic agent are together effective in treating the cancer; (ii) a pharmaceutical composition comprising such a combination; (iii) the use of such a combination for the preparation of a medicament for the treatment of cancer; and (iv) a commercial package or product comprising such a combination as a combined preparation for simultaneous, separate or sequential use; and to a method of treatment of cancer in a subject in need thereof. In some embodiments, the pharmaceutical combination comprises (c) at least one pharmaceutically acceptable carrier. As used herein, terms “treat” or “treatment” refer to therapeutic or palliative measures. Beneficial or desired clinical results include, but are not limited to, alleviation, in whole or in part, of symptoms associated with a disease or disorder or condition, diminishment of the extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state (e.g., one or more symptoms of the disease), and remission (whether partial or total), whether detectable or undetectable. “Treatment” can also mean prolonging survival as compared to expected survival if not receiving treatment. As used herein, the terms “subject,” “individual,” or “patient,” are used interchangeably, refers to any animal, including mammals such as mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, primates, and humans. In some embodiments, the subject is a human. In some embodiments, the subject has experienced and/or exhibited at least one symptom of the disease, disorder, or condition to be treated and/or prevented. In some embodiments, the subject is a pediatric subject. The term “pediatric subject” as used herein refers to a subject under the age of 21 years at the time of diagnosis or treatment. The term “pediatric” can be further be divided into various subpopulations including: neonates (from birth through the first month of life); infants (1 month up to two years of age); children (two years of age up to 12 years of age); and adolescents (12 years of age through 21 years of age (up to, but not including, the twenty-second birthday)). Berhman RE, Kliegman R, Arvin AM, Nelson WE. Nelson Textbook of Pediatrics, 15th Ed. Philadelphia: W.B. Saunders Company, 1996; Rudolph AM, et al. Rudolph’s Pediatrics, 21st Ed. New York: McGraw-Hill, 2002; and Avery MD, First LR. Pediatric Medicine, 2nd Ed. Baltimore: Williams & Wilkins; 1994. In some embodiments, a pediatric subject is from birth through the first 28 days of life, from 29 days of age to less than two years of age, from two years of age to less than 12 years of age, or 12 years of age through 21 years of age (up to, but not including, the twenty-second birthday). In some embodiments, a pediatric subject is from birth through the first 28 days of life, from 29 days of age to less than 1 year of age, from one month of age to less than four months of age, from three months of age to less than seven months of age, from six months of age to less than 1 year of age, from 1 year of age to less than 2 years of age, from 2 years of age to less than 3 years of age, from 2 years of age to less than seven years of age, from 3 years of age to less than 5 years of age, from 5 years of age to less than 10 years of age, from 6 years of age to less than 13 years of age, from 10 years of age to less than 15 years of age, or from 15 years of age to less than 22 years of age. The term “preventing” as used herein means to delay the onset, recurrence or spread, in whole or in part, of the disease or condition as described herein, or a symptom thereof. The term “regulatory agency” refers to a country's agency for the approval of the medical use of pharmaceutical agents with the country. For example, a non-limiting example of a regulatory agency is the U.S. Food and Drug Administration (FDA). The phrase “therapeutically effective amount” means an amount of compound that, when administered to a subject in need thereof, is sufficient to (i) treat a disease, disorder, or condition, (ii) attenuate, ameliorate, or eliminate one or more symptoms of the particular disease, disorder, or condition, or (iii) delay the onset of one or more symptoms of the particular disease, disorder, or condition described herein. The amount of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I- Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I- Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, that will correspond to such an amount will vary depending upon factors such as the particular compound, disease condition and its severity, the identity (e.g., weight) of the subject in need of treatment, but can nevertheless be routinely determined by one skilled in the art. The phrase “effective amount” of a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb- 2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I- H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), as used herein, means an amount of the compound that, when administered to a cell, in vitro or in vivo, is sufficient to reduce proliferation of the cell or to kill the cell. The amount of a compound of Formula (I) (e.g., (I- A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I- E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, that will correspond to such an amount will vary depending upon factors such as the particular compound and genetics of the cell to be treated, but can nevertheless be routinely determined by one skilled in the art. Pharmaceutical Compositions and Administration General In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, is administered as a pharmaceutical composition that includes the compound, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable excipients, and optionally one or more additional therapeutic agents as described herein. In some embodiments, the compounds can be administered in combination with one or more conventional pharmaceutical excipients. Pharmaceutically acceptable excipients include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d-α-tocopherol polyethylene glycol 1000 succinate, surfactants used in pharmaceutical dosage forms such as Tweens, poloxamers or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, tris, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium-chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylates, waxes, polyethylene- polyoxypropylene-block polymers, and wool fat. Cyclodextrins such as α-, ^-, and γ- cyclodextrin, or chemically modified derivatives such as hydroxyalkylcyclodextrins, including 2- and 3-hydroxypropyl-β-cyclodextrins, or other solubilized derivatives can also be used to enhance delivery of compounds described herein. Dosage forms or compositions containing a compound as described herein in the range of 0.005% to 100% with the balance made up from non-toxic excipient may be prepared. The contemplated compositions may contain 0.001%- 100% of a compound provided herein, in one embodiment 0.1-95%, in another embodiment 75-85%, in a further embodiment 20-80%. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington: The Science and Practice of Pharmacy, 22 nd Edition (Pharmaceutical Press, London, UK.2012). Routes of Administration and Composition Components In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, as described herein, or a pharmaceutical composition thereof, can be administered to a subject in need thereof by any accepted route of administration. Acceptable routes of administration include, but are not limited to, buccal, cutaneous, endocervical, endosinusial, endotracheal, enteral, epidural, interstitial, intra-abdominal, intra-arterial, intrabronchial, intrabursal, intracerebral, intracisternal, intracoronary, intradermal, intraductal, intraduodenal, intradural, intraepidermal, intraesophageal, intragastric, intragingival, intraileal, intralymphatic, intramedullary, intrameningeal, intramuscular, intraovarian, intraperitoneal, intraprostatic, intrapulmonary, intrasinal, intraspinal, intrasynovial, intratesticular, intrathecal, intratubular, intratumoral, intrauterine, intravascular, intravenous, intravitreal, nasal, nasogastric, oral, parenteral, percutaneous, peridural, rectal, respiratory (inhalation), subcutaneous, sublingual, submucosal, topical, transdermal, transmucosal, transtracheal, ureteral, urethral and vaginal. In certain embodiments, a preferred route of administration is parenteral (e.g., intratumoral). In some embodiments, a compound of Formula (I) (e.g., (I-A) (e.g., (I-A-1), (I-A-2), or (I-A-3)), (I-B) (e.g., (I-B-1), (I-B-2), or (I-B-3)), (I-C), (I-D), (I-E) (e.g., (I-E-1), (I-E-2), or (I-E-3)), (I-Ea) (e.g., (I-Ea-1), (I-Ea-2), or (I-Ea-3)), (I-Eb) (e.g., (I-Eb-1), (I-Eb-2), or (I-Eb-3)), (I-F) (e.g., (I-F-1), (I-F-2), or (I-F-3)), (I-G), (I-Ga), (I-H) (e.g., (I-H-1), (I-H-2), or (I-H-3))) or Formula (II) (e.g., (II-a)), or a pharmaceutically acceptable salt thereof, as described herein, or a pharmaceutical composition thereof, can be administered orally to a subject in need thereof. Without being bound by any particular theory, it is believed that oral dosing (e.g., versus IV dosing) can be preferred by patients for convenience, perception of efficacy, and/or past experience. Compositions can be formulated for parenteral administration, e.g., formulated for injection via the intravenous, intramuscular, sub-cutaneous, or even intraperitoneal routes. Typically, such compositions can be prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for use to prepare solutions or suspensions upon the addition of a liquid prior to injection can also be prepared; and the preparations can also be emulsified. The preparation of such formulations will be known to those of skill in the art in light of the present disclosure. The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil, or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases the form must be sterile and must be fluid to the extent that it may be easily injected. It also should be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier also can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion, and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin. Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying techniques, which yield a powder of the active ingredient, plus any additional desired ingredient from a previously sterile-filtered solution thereof. Intratumoral injections are discussed, e.g., in Lammers, et al., “Effect of Intratumoral Injection on the Biodistribution and the Therapeutic Potential of HPMA Copolymer-Based Drug Delivery Systems” Neoplasia.2006, 10, 788–795. Pharmacologically acceptable excipients usable in the rectal composition as a gel, cream, enema, or rectal suppository, include, without limitation, any one or more of cocoa butter glycerides, synthetic polymers such as polyvinylpyrrolidone, PEG (like PEG ointments), glycerine, glycerinated gelatin, hydrogenated vegetable oils, poloxamers, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol Vaseline, anhydrous lanolin, shark liver oil, sodium saccharinate, menthol, sweet almond oil, sorbitol, sodium benzoate, anoxid SBN, vanilla essential oil, aerosol, parabens in phenoxyethanol, sodium methyl p-oxybenzoate, sodium propyl p-oxybenzoate, diethylamine, carbomers, carbopol, methyloxybenzoate, macrogol cetostearyl ether, cocoyl caprylocaprate, isopropyl alcohol, propylene glycol, liquid paraffin, xanthan gum, carboxy-metabisulfite, sodium edetate, sodium benzoate, potassium metabisulfite, grapefruit seed extract, methyl sulfonyl methane (MSM), lactic acid, glycine, vitamins, such as vitamin A and E and potassium acetate. In certain embodiments, suppositories can be prepared by mixing the compound described herein with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum and release the active compound. In other embodiments, compositions for rectal administration are in the form of an enema. In other embodiments, the compounds described herein, or a pharmaceutical composition thereof, are suitable for local delivery to the digestive or GI tract by way of oral administration (e.g., solid or liquid dosage forms.). Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the compound is mixed with one or more pharmaceutically acceptable excipients, such as sodium citrate or dicalcium phosphate and/or: a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. In one embodiment, the compositions will take the form of a unit dosage form such as a pill or tablet and thus the composition may contain, along with a compound, or pharmaceutically acceptable salt thereof, as provided herein, a diluent such as lactose, sucrose, dicalcium phosphate, or the like; a lubricant such as magnesium stearate or the like; and a binder such as starch, gum acacia, polyvinylpyrrolidine, gelatin, cellulose, cellulose derivatives, or the like. In another solid dosage form, a powder, marume, solution or suspension (e.g., in propylene carbonate, vegetable oils, PEGs, poloxamer 124 or triglycerides) is encapsulated in a capsule (gelatin or cellulose base capsule). Unit dosage forms in which one or more compounds provided herein or additional active agents are physically separated are also contemplated; e.g., capsules with granules (or tablets in a capsule) of each drug; two-layer tablets; two-compartment gel caps, etc. Enteric coated or delayed release oral dosage forms are also contemplated. Other physiologically acceptable compounds include wetting agents, emulsifying agents, dispersing agents or preservatives that are particularly useful for preventing the growth or action of microorganisms. Various preservatives are well known and include, for example, phenol and ascorbic acid. In certain embodiments the excipients are sterile and generally free of undesirable matter. These compositions can be sterilized by conventional, well-known sterilization techniques. For various oral dosage form excipients such as tablets and capsules sterility is not required. The USP/NF standard is usually sufficient. In certain embodiments, solid oral dosage forms can further include one or more components that chemically and/or structurally predispose the composition for delivery of the compounds to the stomach or the lower GI; e.g., the ascending colon and/or transverse colon and/or distal colon and/or small bowel. Exemplary formulation techniques are described in, e.g., Filipski, K.J., et al., Current Topics in Medicinal Chemistry, 2013, 13, 776-802. Examples include upper-GI targeting techniques, e.g., Accordion Pill (Intec Pharma), floating capsules, and materials capable of adhering to mucosal walls. Other examples include lower-GI targeting techniques. For targeting various regions in the intestinal tract, several enteric/pH-responsive coatings and excipients are available. These materials are typically polymers that are designed to dissolve or erode at specific pH ranges, selected based upon the GI region of desired drug release. These materials also function to protect acid labile drugs from gastric fluid or limit exposure in cases where the active ingredient may be irritating to the upper GI (e.g., hydroxypropyl methylcellulose phthalate series, Coateric (polyvinyl acetate phthalate), cellulose acetate phthalate, hydroxypropyl methylcellulose acetate succinate, Eudragit series (methacrylic acid–methyl methacrylate copolymers), and Marcoat). Other techniques include dosage forms that respond to local flora in the GI tract, Pressure-controlled colon delivery capsule, and Pulsincap. Ocular compositions can include, without limitation, one or more of any of the following: viscogens (e.g., Carboxymethylcellulose, Glycerin, Polyvinylpyrrolidone, Polyethylene glycol); Stabilizers (e.g., Pluronic (triblock copolymers), Cyclodextrins); Preservatives (e.g., Benzalkonium chloride, ETDA, SofZia (boric acid, propylene glycol, sorbitol, and zinc chloride; Alcon Laboratories, Inc.), Purite (stabilized oxychloro complex; Allergan, Inc.)). Topical compositions can include ointments and creams. Ointments are semisolid preparations that are typically based on petrolatum or other petroleum derivatives. Creams containing the selected active agent are typically viscous liquid or semisolid emulsions, often either oil-in-water or water-in-oil. Cream bases are typically water-washable, and contain an oil phase, an emulsifier, and an aqueous phase. The oil phase, also sometimes called the “internal” phase, is generally comprised of petrolatum and a fatty alcohol such as cetyl or stearyl alcohol; the aqueous phase usually, although not necessarily, exceeds the oil phase in volume, and generally contains a humectant. The emulsifier in a cream formulation is generally a nonionic, anionic, cationic, or amphoteric surfactant. As with other carriers or vehicles, an ointment base should be inert, stable, nonirritating, and non-sensitizing. In any of the foregoing embodiments, pharmaceutical compositions described herein can include one or more one or more of the following: lipids, interbilayer crosslinked multilamellar vesicles, biodegradable poly(D,L-lactic-co-glycolic acid) [PLGA]-based or poly anhydride-based nanoparticles or microparticles, and nanoporous particle-supported lipid bilayers. Dosages The dosages may be varied depending on the requirement of the patient, the severity of the condition being treated, and the particular compound being employed. Determination of the proper dosage for a particular situation can be determined by one skilled in the medical arts. The total daily dosage may be divided and administered in portions throughout the day or by means providing continuous delivery. In some embodiments, the compounds described herein are administered at a dosage of from about 0.001 mg/kg to about 500 mg/kg (e.g., from about 0.001 mg/kg to about 200 mg/kg; from about 0.01 mg/kg to about 200 mg/kg; from about 0.01 mg/kg to about 150 mg/kg; from about 0.01 mg/kg to about 100 mg/kg; from about 0.01 mg/kg to about 50 mg/kg; from about 0.01 mg/kg to about 10 mg/kg; from about 0.01 mg/kg to about 5 mg/kg; from about 0.01 mg/kg to about 1 mg/kg; from about 0.01 mg/kg to about 0.5 mg/kg; from about 0.01 mg/kg to about 0.1 mg/kg; from about 0.1 mg/kg to about 200 mg/kg; from about 0.1 mg/kg to about 150 mg/kg; from about 0.1 mg/kg to about 100 mg/kg; from about 0.1 mg/kg to about 50 mg/kg; from about 0.1 mg/kg to about 10 mg/kg; from about 0.1 mg/kg to about 5 mg/kg; from about 0.1 mg/kg to about 1 mg/kg; from about 0.1 mg/kg to about 0.5 mg/kg). Regimens The foregoing dosages can be administered on a daily basis (e.g., as a single dose or as two or more divided doses) or non-daily basis (e.g., every other day, every two days, every three days, once weekly, twice weeks, once every two weeks, once a month). In some embodiments, the period of administration of a compound described herein is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, or more. In a further embodiment, a period of during which administration is stopped is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, or more. In an embodiment, a therapeutic compound is administered to an individual for a period of time followed by a separate period of time. In another embodiment, a therapeutic compound is administered for a first period and a second period following the first period, with administration stopped during the second period, followed by a third period where administration of the therapeutic compound is started and then a fourth period following the third period where administration is stopped. In an aspect of this embodiment, the period of administration of a therapeutic compound followed by a period where administration is stopped is repeated for a determined or undetermined period of time. In a further embodiment, a period of administration is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, or more. In a further embodiment, a period of during which administration is stopped is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, or more. The term “acceptable” with respect to a formulation, composition, or ingredient, as used herein, means having no persistent detrimental effect on the general health of the subject being treated. “API” refers to an active pharmaceutical ingredient. The term “excipient” or “pharmaceutically acceptable excipient” means a pharmaceutically acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, carrier, solvent, or encapsulating material. In one embodiment, each component is “pharmaceutically acceptable” in the sense of being compatible with the other ingredients of a pharmaceutical formulation, and suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenicity, or other problems or complications, commensurate with a reasonable benefit/risk ratio. See, e.g., Remington: The Science and Practice of Pharmacy, 21st ed.; Lippincott Williams & Wilkins: Philadelphia, PA, 2005; Handbook of Pharmaceutical Excipients, 6th ed.; Rowe et al., Eds.; The Pharmaceutical Press and the American Pharmaceutical Association: 2009; Handbook of Pharmaceutical Additives, 3rd ed.; Ash and Ash Eds.; Gower Publishing Company: 2007; Pharmaceutical Preformulation and Formulation, 2nd ed.; Gibson Ed.; CRC Press LLC: Boca Raton, FL, 2009. The term “pharmaceutically acceptable salt” refers to a formulation of a compound that does not cause significant irritation to an organism to which it is administered and does not abrogate the biological activity and properties of the compound. In certain instances, pharmaceutically acceptable salts are obtained by reacting a compound described herein, with acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like. In some instances, pharmaceutically acceptable salts are obtained by reacting a compound having acidic group described herein with a base to form a salt such as an ammonium salt, an alkali metal salt, such as a sodium or a potassium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt, a salt of organic bases such as dicyclohexylamine, N-methyl-D- glucamine, tris(hydroxymethyl)methylamine, and salts with amino acids such as arginine, lysine, and the like, or by other methods previously determined. The term “pharmacologically acceptable salts” is not specifically limited as far as it can be used in medicaments. Examples of a salt that the compounds described herein form with a base include the following: salts thereof with inorganic bases such as sodium, potassium, magnesium, calcium, and aluminum; salts thereof with organic bases such as methylamine, ethylamine, and ethanolamine; salts thereof with basic amino acids such as lysine and ornithine; and ammonium salt. The salts may be acid addition salts, which are specifically exemplified by acid addition salts with the following: mineral acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, nitric acid, and phosphoric acid:organic acids such as formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, methanesulfonic acid, and ethanesulfonic acid; acidic amino acids such as aspartic acid and glutamic acid. The term “pharmaceutical composition” refers to a mixture of a compound described herein with other chemical components (referred to collectively herein as “excipients”), such as carriers, stabilizers, diluents, dispersing agents, suspending agents, and/or thickening agents. The pharmaceutical composition facilitates administration of the compound to a subject. Multiple techniques of administering a compound exist in the art including, but not limited to: rectal, oral, intravenous, aerosol, parenteral, ophthalmic, pulmonary, and topical administration. Compound Preparation The compounds disclosed herein can be prepared in a variety of ways using commercially available starting materials, compounds known in the literature, or from readily prepared intermediates, by employing standard synthetic methods and procedures either known to those skilled in the art, or in light of the teachings herein. Standard synthetic methods and procedures for the preparation of organic molecules and functional group transformations and manipulations can be obtained from the relevant scientific literature or from standard textbooks in the field. Although not limited to any one or several sources, classic texts such as R. Larock, Comprehensive Organic Transformations, VCH Publishers (1989); L. Fieser and M. Fieser, Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); Smith, M. B., March, J., March' s Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 5th edition, John Wiley & Sons: New York, 2001 ; and Greene, T.W., Wuts, P.G. M., Protective Groups in Organic Synthesis, 3rd edition, John Wiley & Sons: New York, 1999, are useful and recognized reference textbooks of organic synthesis known to those in the art. The following descriptions of synthetic methods are designed to illustrate, but not to limit, general procedures for the preparation of compounds of the present disclosure. The synthetic processes disclosed herein can tolerate a wide variety of functional groups; therefore, various substituted starting materials can be used. The processes generally provide the desired final compound at or near the end of the overall process, although it may be desirable in certain instances to further convert the compound to a pharmaceutically acceptable salt thereof. Scheme 1 depicts the synthesis of Compound IA-3 which is a compound of Formula (I-A), wherein R 1 is C(O)OH; and each remaining variable is defined in accordance with Formula (I-A). Compound IA-1 is reacted with Compound I-L1 under standard conditions for Suzuki coupling to provide Compound IA-2, wherein one of X A and X T is B(OH)2 or Bpin; the other of X A and X T is -Br; and each remaining variable in IA-1 and I-L1 is defined in accordance with Formula (I-A). Hydrolysis of the C1-2 alkyl ester in IA-2 provides the corresponding carboxylic acid which is then reacted with I-C1 under standard conditions for amide bond formation, wherein each variable in I-C1 is defined in accordance with Formula (I-A). Removal of the tert-butyl group under standard conditions for deprotection then provides Compound IA-3. Scheme 2 Compound IB-3 which is a compound of Formula (I-B), wherein R 1 is C(O)OH; and each remaining variable is defined in accordance with Formula (I-B), is prepared from IA-1 and I-L2 using methods depicted for Compound IA-3 in Scheme 1, above. One of X A and X T is B(OH) 2 or Bpin; the other of X A and X T is -Br; and each remaining variable in IA-1 and I- L2 is defined in accordance with Formula (I-B).

Scheme 3 Analogously, as shown in Scheme 3, Compound IF-3 (a compound of Formula (I-F), wherein R 1 is C(O)OH; and each remaining variable is defined in accordance with Formula (I- F)) and Compound IH-3 (a compound of Formula (I-H), wherein R 1 is C(O)OH; and each remaining variable is defined in accordance with Formula (I-H)) are prepared with methods similar to those depicted in Schemes 1 and 2 by using I-C2 (wherein each variable is defined in accordance with Formula (I-F) or (I-H)) instead of I-C1. Also provided herein are compounds of Formula (SI-A) and Formula (SI-B): Formula (SI-A) Formula (SI-B) or salts thereof, wherein: each R 2 and R 4 is independently selected from: halo, CN, C 1-3 alkyl, C 1-3 haloalkyl, C 1- 3 alkoxy, C1-3 haloalkoxy, OH, and NR d R e ; m2 is 0, 1, or 2; m4 is 0, 1, 2, or 3; m6 is 0 or 1; R e1 is H or C1-6 alkyl optionally substituted with 1-3 -F; R e2 is H or C 1-6 alkyl optionally substituted with 1-3 -F; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5; each R L is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C1-6 haloalkoxy; -NR d R e ; C(=O)N(R f )2; S(O)0-2(C1-6 alkyl); S(O)0-2(C1-6 haloalkyl); S(O) 1-2 N(R f ) 2 ; -R b ; and C 1-6 alkyl optionally substituted with 1-6 R c ; each R a is independently selected from the group consisting of: (a) halo; (b) cyano; (c) -OH; (d) oxo; (e) -C1-6 alkoxy; (f) -C 1-6 haloalkoxy; (g) -NR d R e ; (h) C(=O)C1-6 alkyl; (i) C(=O)C 1-6 haloalkyl; (j) C(=O)OH; (k) C(=O)OC1-6 alkyl; (l) C(=O)OC 1-6 haloalkyl; (m)C(=O)N(R f ) 2 ; (n) S(O)0-2(C1-6 alkyl); (o) S(O)0-2(C1-6 haloalkyl); (p) S(O) 1-2 N(R f ) 2 ; and (q) C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted with 1-6 R c ; each R b is independently selected from the group consisting of: -(L b )b-R b1 and -R b1 , wherein: each b is independently 1, 2, or 3; each -L b is independently selected from the group consisting of: -O-; -N(H)-; -N(C 1-3 alkyl)-; -S(O) 0-2 -; C(=O); and C 1-3 alkylene; and each R b1 is independently selected from the group consisting of: C3-10 cycloalkyl, 4-10 membered heterocyclyl, C6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 R g ; each R c is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C 1-6 haloalkoxy; -NR d R e ; C(=O)C 1-6 alkyl; C(=O)C 1-6 haloalkyl; C(=O)OC 1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)OH; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); and S(O)1-2N(R f )2; each R d and R e is independently selected from the group consisting of: H; C(=O)C1-6 alkyl; C(=O)C 1-6 haloalkyl; C(=O)OC 1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)N(R f ) 2 ; S(O) 1- 2(C1-6 alkyl); S(O)1-2(C1-6 haloalkyl); S(O)1-2N(R f )2; and C1-6 alkyl optionally substituted with 1-3 R h ; each R f is independently selected from the group consisting of: H and C 1-6 alkyl optionally substituted with 1-3 R h ; and each R h is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C 1-6 haloalkoxy; -NH 2 , -N(H)(C 1-3 alkyl); and -N(C 1-3 alkyl) 2 . In some embodiments of Formula (SI-A) and Formula (SI-B), m2 is 0; m4 is 0; and m6 is 0. In some embodiments of Formula (SI-A) and Formula (SI-B), each R a is independently C1-6 alkyl optionally substituted with 1-3 -F (e.g., CH3 or CF3). In some embodiments of Formula (SI-A) and Formula (SI-B), m2 is 0; m4 is 0; and m6 is 0; and R a is C 1-6 alkyl optionally substituted with 1-3 -F (e.g., CH 3 or CF 3 ). In some embodiments of Formula (SI-A) and Formula (SI-B), a3 is 1. In some embodiments of Formula (SI-A) and Formula (SI-B), L A3 is -O-. In some embodiments of Formula (SI-A) and Formula (SI-B), a3 is 1; and L A3 is -O-. In some embodiments of Formula (SI-A) and Formula (SI-B), a1a + a1b is 3 or 4. For example, a1a + a1b can be 3. For example, a1a + a1b can be 4. In some embodiments of Formula (SI-A) and Formula (SI-B), a1a + a1b is 2 or 5. For example, a1a + a1b can be 5. In some embodiments of Formula (SI-A) and Formula (SI-B), each occurrence of L A1a and L A1b is -CH 2 -. In some embodiments of Formula (SI-A) and Formula (SI-B), one occurrence of L A1a is -CHR L - or -C(R L ) 2 -; each remaining occurrence of L A1a is -CH 2 -; and each occurrence of L A1b is -CH2-. In some embodiments of Formula (SI-A) and Formula (SI- B), one occurrence of L A1b is -CHR L - or -C(R L )2-; each remaining occurrence of L A1b is -CH2- ; and each occurrence of L A1a is -CH 2 -. In some embodiments of Formula (SI-A) and Formula (SI-B), each R L is independently selected from the group consisting of: -F and -C1-3 alkyl optionally substituted with 1-3 F (e.g., CH3 or CF3). In some embodiments, the compounds of Formula (SI-A) and Formula (SI-B) are compounds of Formula (SI-A-1) and (SI-B-1), respectively:

Formula (SI-B-1) or salts thereof, wherein: each R 2 and R 4 is independently selected from: halo, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, and C1-3 haloalkoxy; m2 is 0 or 1; m4 is 0 or 1; a1a is 0, 1, 2, or 3; n2 is 0 or 1; n3 is 0 or 1; m8 is 0, 1, or 2; each R a is independently selected from the group consisting of: -F and -C1-3 alkyl optionally substituted with 1-3 F (e.g., CH3 or CF3); R e1 is H or C 1-6 alkyl optionally substituted with 1-3 -F; R e2 is H or C 1-6 alkyl optionally substituted with 1-3 -F; L A3 is selected from the group consisting of: -O- and -N(H)-; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L ) 2 -; a1a is an integer from 0 to 3; and each R L is independently selected from the group consisting of: -F and -C 1-3 alkyl optionally substituted with 1-3 F (e.g., CH3 or CF3). In some embodiments of Formula (SI-A-1) and (SI-B-1), m2 is 0; m4 is 0; 0 to 1 occurrence of L A1a is -CHR L - or -C(R L ) 2 -; each remaining occurrence of L A1a is -CH 2 -; L A3 is -O-; and L A1b is -CH2-. For example, compounds of Formulas (SI-A-1) and (SI-B-1) can be: In some embodiments, the compounds of Formula (SI-A) and Formula (SI-B) are compounds of Formula (SI-A-2) or Formula (SI-B-2), respectively: Formula (SI-A-2)

Formula (SI-B-2) or salts thereof, wherein: each R 2 and R 4 is independently selected from: halo, C1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, and C1-3 haloalkoxy; m2 is 0 or 1; m4 is 0 or 1; a1a is 0, 1, 2, or 3; m8 is 0, 1, or 2; each R a is independently selected from the group consisting of: -F and -C 1-3 alkyl optionally substituted with 1-3 F (e.g., CH3 or CF3); R e1 is H or C1-6 alkyl optionally substituted with 1-3 -F; R e2 is H or C 1-6 alkyl optionally substituted with 1-3 -F; L A3 is selected from the group consisting of: -O- and -N(H)-; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; a1a is an integer from 0 to 3; and each R L is independently selected from the group consisting of: -F and -C1-3 alkyl optionally substituted with 1-3 F (e.g., CH3 or CF3). In some embodiments of Formula (SI-A-2) and Formula (SI-B-2), m2 is 0; m4 is 0; 0 to 1 occurrence of L A1a is -CHR L - or -C(R L )2-; each remaining occurrence of L A1a is -CH2-; L A3 is -O-; and L A1b is -CH2-. For example, compounds of Formula (SI-A-2) and Formula (SI- B-2) can be:

In some embodiments, the compounds of Formula (SI-A) or Formula (SI-B) are compounds of Formula (SI-A-3) or Formula (SI-B-3), respectively: Formula (SI-A-3)

Formula (SI-B-3) or salts thereof, wherein: each R 2 and R 4 is independently selected from: halo, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, and C 1-3 haloalkoxy; m2 is 0 or 1; m4 is 0 or 1; a1a is 0, 1, 2, or 3; n2 is 0 or 1; n3 is 0 or 1; n4 is 0, 1, or 2; m8 is 0, 1, or 2; R a is independently selected from the group consisting of: -F and -C1-3 alkyl optionally substituted with 1-3 F (e.g., CH3 or CF3); R e1 is H or C 1-6 alkyl optionally substituted with 1-3 -F; R e2 is H or C1-6 alkyl optionally substituted with 1-3 -F; L A3 is selected from the group consisting of: -O- and -N(H)-; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; a1a is an integer from 0 to 3; and each R L is independently selected from the group consisting of: -F and -C 1-3 alkyl optionally substituted with 1-3 F (e.g., CH 3 or CF 3 ). In some embodiments of Formula (SI-A-3) and Formula (SI-B-3) , m2 is 0; m4 is 0; m8 is 0; 0 to 2 occurrences of L A1a is -CH2-; L A3 is -O-; and L A1b is -CH2-. For example, compounds of Formula (SI-A-3) and Formula (SI-B-3) can be:

Exemplary embodiments of compounds of one or more of Formulas (SI-A), (SI-B), (SI-A-1), (SI-B-1), (SI-A-2), (SI-B-2), (SI-A-3), and (SI-B-3) include certain compounds described in Examples 1, 3, 4, 6, 7, 9, 10, 11, 12, 13, 14, 15, 17, 18, 19, 20, 21, 22, 25, 26, 27, 31, 32, 33, 35, 36, 38, 39, 40, 41, 42, 43, 45, 46, 47, 48, 50, 52, 54, 55, 56, 57, 60, 62, 63, 64, 67, 68, 69, 70, 71, 72, 77, 78, 80, 81, 84, 85, 86, 88, 89, 91, 92, 93, 94, 96, 97, 99, 100, 102, 103, 106, 107, 109, 110, 116, 117, 118, 119, 120, 121, 122, 131, 132, 133, 134, 135, 136, 138, 140, 141, 142, 143, 144, 147, 148, 149, 150, 151, 152, 153, 160, 161, 163, 168, 174, 175, 177, 181, 183, 187, 189, 190, 192, 193, 194, 199, 200, 201, 202, 203, 204, 205, 206, 215, 216, 223, 224, and 229. Also provided herein are compounds of Formula (SI-E) and Formula (SI-G):

Formula (SI-G) or salts thereof, wherein: each R 2 and R 4 is independently selected from: H, C 1-3 alkyl, and halo; a3 is 0 or 1; m2 is 0, 1, or 2; m4 is 0, 1, 2, or 3; m6 is 0 or 1; R e1 is H or C1-6 alkyl optionally substituted with 1-3 -F; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; a1a is an integer from 0 to 5; a1d is an integer from 0 to 4, provided that a1a + a1d is from 1 to 4; each R L is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C1-6 haloalkoxy; -NR d R e ; C(=O)N(R f )2; S(O)0-2(C1-6 alkyl); S(O)0-2(C1-6 haloalkyl); S(O)1-2N(R f )2; -R b ; and C1-6 alkyl optionally substituted with 1-6 R c ; each R a is independently selected from the group consisting of: (a) halo; (b) cyano; (c) -OH; (d) oxo; (e) -C 1-6 alkoxy; (f) -C 1-6 haloalkoxy; (g) -NR d R e ; (h) C(=O)C1-6 alkyl; (i) C(=O)C 1-6 haloalkyl; (j) C(=O)OH; (k) C(=O)OC1-6 alkyl; (l) C(=O)OC 1-6 haloalkyl; (m)C(=O)N(R f ) 2 ; (n) S(O)0-2(C1-6 alkyl); (o) S(O) 0-2 (C 1-6 haloalkyl); (p) S(O) 1-2 N(R f ) 2 ; and (q) C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted with 1-6 R c ; each R b is independently selected from the group consisting of: -(L b )b-R b1 and -R b1 , wherein: each b is independently 1, 2, or 3; each -L b is independently selected from the group consisting of: -O-; -N(H)-; -N(C 1-3 alkyl)-; -S(O) 0-2 -; C(=O); and C 1-3 alkylene; each R b1 is independently selected from the group consisting of: C3-10 cycloalkyl, 4-10 membered heterocyclyl, C6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 R g ; each R c is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C1-6 haloalkoxy; -NR d R e ; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC1-6 haloalkyl; C(=O)OH; C(=O)N(R f )2; S(O)0-2(C1-6 alkyl); S(O)0-2(C1-6 haloalkyl); and S(O) 1-2 N(R f ) 2 ; each R d and R e is independently selected from the group consisting of: H; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC1-6 haloalkyl; C(=O)N(R f )2; S(O)1- 2 (C 1-6 alkyl); S(O) 1-2 (C 1-6 haloalkyl); S(O) 1-2 N(R f ) 2 ; and C 1-6 alkyl optionally substituted with 1-3 R h ; each R f is independently selected from the group consisting of: H and C1-6 alkyl optionally substituted with 1-3 R h ; each R g is independently selected from the group consisting of: R h ; C 1-3 alkyl; and C 1- 3 haloalkyl; and each R h is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C 1-6 haloalkoxy; -NH 2 , -N(H)(C 1-3 alkyl); and -N(C 1-3 alkyl) 2 . In some embodiments of Formula (SI-E) and Formula (SI-G), m2 is 0; m4 is 0; and m6 is 0. In some embodiments of Formula (SI-E) and Formula (SI-G), R a is C 1-6 alkyl optionally substituted with 1-3 -F (e.g., CH3 or CF3). In some embodiments of Formula (SI-E) and Formula (SI-G), m2 is 0; m4 is 0; and m6 is 0; and R a is C 1-6 alkyl optionally substituted with 1-3 -F (e.g., CH 3 or CF 3 ). In some embodiments of Formula (SI-E) and Formula (SI-G), a3 is 1. In some embodiments, L A3 is -O-. In some embodiments of Formula (SI-E) and Formula (SI-G), a3 is 1; and L A3 is -O-. In some embodiments of Formula (SI-E) and Formula (SI-G), a1a + a1d is 2 or 5. For example, a1a + a1b can be 5. In some such embodiments each occurrence of L A1a and L A1b is -CH 2 -. In some embodiments of Formula (SI-E) and Formula (SI-G), a1a is 0; and a1d is 1, 2 or 3. In some embodiments of Formula (SI-E) and Formula (SI-G), a1a is 0; and each occurrence of L A1b is -CH2-. In some embodiments of Formula (SI-E) and Formula (SI-G), a1a is 0; one occurrence of L A1b is -CHR L - or -C(R L )2-; and each remaining occurrence of L A1b is -CH 2 -. In some embodiments of Formula (SI-E) and Formula (SI-G), each R L is independently selected from the group consisting of: -F and -C1-3 alkyl optionally substituted with 1-3 F (e.g., CH3 or CF3). In some embodiments, the compounds of Formula (SI-E) are compounds of Formula (SI-E-1): or salts thereof, wherein: each R 2 and R 4 is independently selected from: halo, C1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, and C 1-3 haloalkoxy; m2 is 0 or 1; m4 is 0 or 1; a1a is 1, 2, or 3; R a is independently selected from the group consisting of: -F and -C 1-3 alkyl optionally substituted with 1-3 F (e.g., CH3 or CF3); R e1 is H or C1-6 alkyl optionally substituted with 1-3 -F; L A3 is selected from the group consisting of: -O- and -N(H)-; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; a1a is an integer from 0 to 3; and each R L is independently selected from the group consisting of: -F and -C 1-3 alkyl optionally substituted with 1-3 F (e.g., CH3 or CF3). In some embodiments, of Formula (SI-E-1) m2 is 0; m4 is 0; 1-3 occurrences of L A1a are -CH 2 -; L A3 is -O-; and L A1b is -CH 2 -. For example, compounds of Formula (SI-E-1): can be:

In some embodiments, the compounds of Formula (SI-E) and Formula (SI-G) are compounds of Formula (SI-E-2) and Formula (SI-G-2), respectively: Formula (SI-G-2) or salts thereof, wherein: each R 2 and R 4 is independently selected from: halo, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, and C1-3 haloalkoxy; m2 is 0 or 1; m4 is 0 or 1; a1d is 1, 2, or 3; L A4 is cyclohexylene or phenylene (e.g., 1,4-cyclohexylene or 1,4-phenylene); R a is independently selected from the group consisting of: -F and -C 1-3 alkyl optionally substituted with 1-3 F (e.g., CH3 or CF3); R e1 is H or C1-6 alkyl optionally substituted with 1-3 -F; L A3 is selected from the group consisting of: -O- and -N(H)-; L A1b is independently selected from the group consisting of: -CH 2 -, -CHR L -, and - C(R L )2-; and each R L is independently selected from the group consisting of: -F and -C1-3 alkyl optionally substituted with 1-3 F (e.g., CH 3 or CF 3 ). In some embodiments, of Formula (SI-E-2) and (SI-G-2) m2 is 0; m4 is 0; L A3 is -O-; and each occurrence of L A1b is -CH 2 -. For example, compounds of Formula (SI-E-2) and Formula (SI-G-2) can be: Exemplary embodiments of compounds of one or more of Formulas (SI-E), (SI-G), (SI-E-1), (SI-E-2), and (SI-G-2) include certain compounds described in Examples 8, 16, 28, 29, 34, 44, 53, 65, 66, 73, 74, 75, 76, 87, 95, 98, 111, 137, 154, 156, 158, 162, 164, 167, 169, 170, 171, 178, 182, 191, 195, 196, 197, 198, 212, 213, and 214. Also provided herein are compounds of Formulas (I-Ind-1), (I-Ind-2), and (I-Ind-3): or salts thereof, wherein: each R Y is independently selected from the group consisting of: R a and R b ; c1 is 0, 1, 2, or 3; n2 and n3 are independently 0, 1, or 2; R aN is H or C1-6 alkyl optionally substituted with 1-3 R c ; m8 is 0 to 4; each R a is independently selected from the group consisting of: (a) halo; (b) cyano; (c) -OH; (d) oxo; (e) -C 1-6 alkoxy; (f) -C 1-6 haloalkoxy; (g) -NR d R e ; (h) C(=O)C 1-6 alkyl; (i) C(=O)C 1-6 haloalkyl; (j) C(=O)OH; (k) C(=O)OC1-6 alkyl; (l) C(=O)OC 1-6 haloalkyl; (m)C(=O)N(R f ) 2 ; (n) S(O)0-2(C1-6 alkyl); (o) S(O)0-2(C1-6 haloalkyl); (p) S(O) 1-2 N(R f ) 2 ; and (q) C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted with 1-6 R c ; each R b is independently selected from the group consisting of: -(L b )b-R b1 and -R b1 , wherein: each b is independently 1, 2, or 3; each -L b is independently selected from the group consisting of: -O-; -N(H)-; -N(C 1-3 alkyl)-; -S(O) 0-2 -; C(=O); and C 1-3 alkylene; each R b1 is independently selected from the group consisting of: C3-10 cycloalkyl, 4-10 membered heterocyclyl, C6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 R g ; each R c is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C 1-6 haloalkoxy; -NR d R e ; C(=O)C 1-6 alkyl; C(=O)C 1-6 haloalkyl; C(=O)OC 1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)OH; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); and S(O)1-2N(R f )2; each R d and R e is independently selected from the group consisting of: H; C(=O)C 1-6 alkyl; C(=O)C 1-6 haloalkyl; C(=O)OC 1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)N(R f ) 2 ; S(O) 1- 2(C1-6 alkyl); S(O)1-2(C1-6 haloalkyl); S(O)1-2N(R f )2; and C1-6 alkyl optionally substituted with 1-3 R h ; each R f is independently selected from the group consisting of: H and C 1-6 alkyl optionally substituted with 1-3 R h ; each R g is independently selected from the group consisting of: R h ; C1-3 alkyl; and C1- 3 haloalkyl; and each R h is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C1-6 haloalkoxy; -NH2, -N(H)(C1-3 alkyl); and -N(C1-3 alkyl)2. In some embodiments of Formulas (I-Ind-1), (I-Ind-2), and (I-Ind-3), c1 is 0. In some embodiments of Formula (I-Ind-1), n2 and n3 are independently 1. In some embodiments of Formulas (I-Ind-1), (I-Ind-2), and (I-Ind-3), R aN is C1-6 alkyl optionally substituted with 1-3 R c . In some embodiments of Formulas (I-Ind-1), (I-Ind-2), and (I-Ind-3), c1 is 0; and R aN is C1-6 alkyl optionally substituted with 1-3 R c . In some embodiments of Formula (I-Ind-2) and Formula (I-Ind-3), c1 is 0; R aN is - CH 3 ; and R a is selected from the group consisting of: -F and -C 1-3 alkyl optionally substituted with 1-3 F (e.g., CH3 or CF3). For example, compounds of Formula (I-Ind-2) and Formula (I- Ind-3) can be: Exemplary embodiments of compounds of one or more of Formulas (I-Ind-1), (I-Ind- 2), and (I-Ind-3) include certain compounds described in Examples 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 25, 26, 27, 28, 29, 34, 38, 44, 49, 53, 58, 59, 61, 66, 67, 73, 74, 75, 76, 85, 86, 87, 90, 95, 96, 98, 101, 113, 137, 145, 147, 149, 150, 151, 152, 154, 156, 157, 158, 162, 163, 164, 167, 168, 169, 170, 171, 172, 175, 177, 178, 182, 184, 185, 191, 195, 196, 197, 207, 208, 209, 210, 211, 212, 213, and 214. Also provided herein are compounds of Formula (SI-J) and Formula (SI-K): Formula (SI-J) Formula (SI-K) or salts thereof, wherein: X is selected from the group consisting of: halo; -B(OH) 2 ; -BF3-; -B(OR T )2, wherein each R T is independently C1-3 alkyl optionally substituted with 1-3 R c ; and , wherein L B is C 2-6 alkylene optionally substituted with 1-4 R a ; R a1 is H or C 1-3 alkyl optionally substituted with 1-3 F; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a3 is 0 or 1; a3b is 0 or 1; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; a1a is an integer from 0 to 5; a1b is an integer from 0 to 4, provided that a1a + a1b is from 1 to 4; each R L is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C1-6 haloalkoxy; -NR d R e ; C(=O)N(R f )2; S(O)0-2(C1-6 alkyl); S(O)0-2(C1-6 haloalkyl); S(O)1-2N(R f )2; -R b ; and C1-6 alkyl optionally substituted with 1-6 R c ; each R Y is independently selected from the group consisting of: R a and R b ; c1 is 0, 1, 2, or 3; R aN is H or C1-6 alkyl optionally substituted with 1-3 R c ; m8 is 0, 1, or 2. each R a is independently selected from the group consisting of: (a) halo; (b) cyano; (c) -OH; (d) oxo; (e) -C 1-6 alkoxy; (f) -C1-6 haloalkoxy; (g) -NR d R e ; (h) C(=O)C 1-6 alkyl; (i) C(=O)C 1-6 haloalkyl; (j) C(=O)OH; (k) C(=O)OC 1-6 alkyl; (l) C(=O)OC 1-6 haloalkyl; (m)C(=O)N(R f )2; (n) S(O)0-2(C1-6 alkyl); (o) S(O) 0-2 (C 1-6 haloalkyl); (p) S(O)1-2N(R f )2; and (q) C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted with 1-6 R c ; each R b is independently selected from the group consisting of: -(L b )b-R b1 and -R b1 , wherein: each b is independently 1, 2, or 3; each -L b is independently selected from the group consisting of: -O-; -N(H)-; -N(C 1-3 alkyl)-; -S(O)0-2-; C(=O); and C1-3 alkylene; each R b1 is independently selected from the group consisting of: C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 R g ; each R c is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C1-6 haloalkoxy; -NR d R e ; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)OH; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); and S(O)1-2N(R f )2; each R d and R e is independently selected from the group consisting of: H; C(=O)C1-6 alkyl; C(=O)C 1-6 haloalkyl; C(=O)OC 1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)N(R f ) 2 ; S(O) 1- 2 (C 1-6 alkyl); S(O) 1-2 (C 1-6 haloalkyl); S(O) 1-2 N(R f ) 2 ; and C 1-6 alkyl optionally substituted with 1-3 R h ; each R f is independently selected from the group consisting of: H and C1-6 alkyl optionally substituted with 1-3 R h ; each R g is independently selected from the group consisting of: R h ; C1-3 alkyl; and C1- 3 haloalkyl; and each R h is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C 1-6 haloalkoxy; -NH 2 , -N(H)(C 1-3 alkyl); and -N(C 1-3 alkyl) 2 . In some embodiments of Formula (SI-J) and Formula (SI-K), a3 is 1. In some embodiments of Formula (SI-J) and Formula (SI-K), L A3 is -O-. In some embodiments of Formula (SI-J) and Formula (SI-K), a3 is 1; and L A3 is -O-. In some embodiments of Formula (SI-J) and Formula (SI-K), a1a + a1b is 2 or 5. For example, a1a + a1b can be 5. In some embodiments of Formula (SI-J) and Formula (SI-K), a1a is 0; a3b is 0 or 1; and each occurrence of L A1b is -CH2-. In some embodiments of Formula (SI-J) and Formula (SI-K), a1a is 0; a3b is 0 or 1; one occurrence of L A1b is -CHR L - or -C(R L )2-; and each remaining occurrence of L A1b is -CH 2 -. In some embodiments of Formula (SI-J) and Formula (SI-K), each R L is independently selected from the group consisting of: -F and -C 1-3 alkyl optionally substituted with 1-3 F (e.g., CH3 or CF3). In some embodiments, the compounds of Formula (SI-J) are compounds of Formula (SI-J-1): Formula (SI-J-1) or salts thereof, wherein: m8 is 0 or 1; R a1 is C1-3 alkyl optionally substituted with 1-3 F; R a is independently selected from the group consisting of: -F and -C1-3 alkyl optionally substituted with 1-3 F (e.g., CH 3 or CF 3 ); L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; a1a is an integer from 1 to 4; each R L is independently selected from the group consisting of: -F and -C 1-3 alkyl optionally substituted with 1-3 F (e.g., CH3 or CF3); each R Y is H or C1-3 alkyl optionally substituted with 1-3 F; c1 is 0, or 1; and R aN is C1-6 alkyl optionally substituted with 1-3 F. In some embodiments of Formula (SI-J-1), c1 is 0; m8 is 0; each occurrence of L A1a is -CH 2 -; and L A1b is -CH 2 -. For example, compounds of Formula (SI-J-1) can be:

In some embodiments, the compounds of Formula (SI-J) or Formula (SI-K) are compounds of Formula (SI-J-2) or Formula (SI-K-2), respectively: or salts thereof, wherein: m8 is 0 or 1; R a1 is C 1-3 alkyl optionally substituted with 1-3 F; L A4 is cyclohexylene or phenylene (e.g., 1,4-cyclohexylene or 1,4-phenylene); L A1b is independently selected from the group consisting of: -CH2-, -CHR L -, and - C(R L ) 2 -; a1b is an integer from 1 to 4; a3b is 0 or 1; each R L is independently selected from the group consisting of: -F and -C 1-3 alkyl optionally substituted with 1-3 F (e.g., CH3 or CF3); each R Y is H or C1-3 alkyl optionally substituted with 1-3 F; c1 is 0 or 1; and R aN is C 1-6 alkyl optionally substituted with 1-3 F. In some embodiments of Formula (SI-J-2) and Formula (SI-K-2), c1 is 0; m8 is 0; 0 to 1 occurrence of L A1b are -CHR L - or -C(R L ) 2 -; and each remaining occurrence of L A1b is -CH 2 - . For example, compounds of Formula (SI-J-2) and Formula (SI-K-2) can be: For example, compounds of Formula (SI-J-2) and Formula (SI-K-2) can be: In some embodiments of Formula (SI-J-2) and Formula (SI-K-2), X is , is 0; m8 is 0; 0 to 1 occurrence of L A1b are -CHR L - or -C(R L )2-; and each remaining occurrence of L A1b is -CH 2 -. For example, compounds of Formula (SI-J-2) and Formula (SI-K-2) can be:

, salts thereof. Exemplary embodiments of compounds of one or more of Formulas (SI-J), (SI-K), (SI-J-1), and (SI-J-2), and (SI-K-2) include certain compounds described in Examples 49, 58, 59, 61, 90, 101, 113, 145, 157, 172, 184, 185, 207, 208, 209, 210, and 211. Also provided herein are methods of preparing compounds of Formula (I-A), comprising reacting compounds of Formula (SI-AA) with compounds of Formula (SII) to provide compounds of Formula (I-A), as depicted in Scheme A: Scheme A

wherein: m4 is 0, 1, 2, or 3 (e.g., 0); m2 is 0, 1, or 2 (e.g., 0); each R 2 and R 4 is independently selected from the group consisting of: halo, CN, C 1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, C1-3 haloalkoxy, OH, and NR d R e ; m6 is 0 or 1; R 1 is selected from the group consisting of: (a) C(O)OH, (b) C(O)NR d R e , and (c) C(O)OC 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R c ; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; and L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; each R L is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C1-6 haloalkoxy; -NR d R e ; C(=O)N(R f )2; S(O)0-2(C1-6 alkyl); S(O)0-2(C1-6 haloalkyl); S(O)1-2N(R f )2; -R b ; and C1-6 alkyl optionally substituted with 1-6 R c ; R e2 is H or C1-6 alkyl optionally substituted with 1-3 -F; Ring C is selected from the group consisting of: ; c1 is 0, 1, 2, or 3; each R Y is independently selected from the group consisting of: R a and R b ; R aN is H or C 1-6 alkyl optionally substituted with 1-3 R c ; Y 1 and Y 2 are independently N, CH, or CR Y ; yy represents the point of attachment to the -C(O)NH- moiety; X is CH or N; L C is selected from the group consisting of: a bond, -CH2-, -CHR a -, -C(R a )2-, -N(R d )-, and O, provided that when X is N, then L C is other than O; each R a is independently selected from the group consisting of: (a) halo; each R b is independently selected from the group consisting of: -(L b )b-R b1 and -R b1 , wherein: each b is independently 1, 2, or 3; each -L b is independently selected from the group consisting of: -O-; -N(H)-; -N(C 1-3 alkyl)-; -S(O)0-2-; C(=O); and C1-3 alkylene; and each R b1 is independently selected from the group consisting of: C3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 R g ; each R c is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C 1-6 haloalkoxy; -NR d R e ; C(=O)C 1-6 alkyl; C(=O)C 1-6 haloalkyl; C(=O)OC 1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)OH; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); and S(O)1-2N(R f )2; each R d and R e is independently selected from the group consisting of: H; C(=O)C1-6 alkyl; C(=O)C 1-6 haloalkyl; C(=O)OC 1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)N(R f ) 2 ; S(O) 1- 2(C1-6 alkyl); S(O)1-2(C1-6 haloalkyl); S(O)1-2N(R f )2; and C1-6 alkyl optionally substituted with 1-3 R h ; each R f is independently selected from the group consisting of: H and C 1-6 alkyl optionally substituted with 1-3 R h ; each R g is independently selected from the group consisting of: R h ; C1-3 alkyl; and C1- 3 haloalkyl; and each R h is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C1-6 haloalkoxy; -NH2, -N(H)(C1-3 alkyl); and -N(C1-3 alkyl)2. In the methods of preparing compounds of Formula (I-A) as depicted in Scheme A, the variables m4, R 4 , m2, R 2 , R 1 , R a , m6, L A3 , a3, L A1a , a1a, L A4 , L A1b , a1b, Ring C, L C , and X can be as defined herein for Formula (I-A). In some embodiments of the methods, the compounds of Formula (I-A) are compounds of Formula (I-A-1). In some embodiments of the methods, the compounds of Formula (I-A) are compounds of Formula (I-A-2). In some embodiments of the methods, the compounds of Formula (I-A) are compounds of Formula (I- A-3). In some embodiments of the methods of preparing compounds of Formula (I-A), the compounds of Formula (SI-AA) are compounds of Formula (SI-A). In some embodiments of the methods of preparing compounds of Formula (I-A), the compounds of Formula (SI-AA) are compounds of Formula (SI-A-1). In some embodiments of the methods of preparing compounds of Formula (I-A), the compounds of Formula (SI-AA) are compounds of Formula (SI-A-2). In some embodiments of the methods of preparing compounds of Formula (I-A), the compounds of Formula (SI-AA) are compounds of Formula (SI-A-3). In some embodiments of the methods, Ring C is or , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 F; and R aN is C 1-3 alkyl. In some embodiments of the methods, a3 is 1; L A3 is -O-; a1b is 1; L A1b is -CH2-; each L A1a is independently selected from the group consisting of: -CH2- and -CH(Me)- , provided that 0-1 L A1a is -CH(Me)-; and L A4 is 4-10 membered nitrogen-containing heterocyclylene optionally substituted with 1-2 substituents independently selected from the group consisting of: -F and C1-3 alkyl optionally substituted with 1-3 -F. In some embodiments of the methods, the moiety is selected from the groups depicted in Table (L-I-A). In some embodiments of the methods, R e2 is H; and the methods comprise reacting compounds of Formula (SI-AA) with compounds of Formula (SII) under standard conditions for amide bond formation to provide compounds of Formula (I-A). Examples of standard conditions for amide bond formation include those described in Chem. Soc. Rev., 2009,38, 606-631, which is incorporated herein by reference in its entirety. In some embodiments, the methods comprise reacting compounds of Formula (SI-AA) with compounds of Formula (SII) in the presence of the carboxyl activating agent (e.g., HATU, HOBT, EDCI, DCC, DIC, etc.) and optionally a base (e.g., triethylamine or diisopropylethylamine) to provide compounds of Formula (I-A). Also provided herein are methods of preparing compounds of Formula (I-B), comprising reacting compounds of Formula (SI-BB) with compounds of Formula (SII) to provide compounds of Formula (I-B), as depicted in Scheme B: Scheme B

wherein: m4 is 0, 1, 2, or 3 (e.g., 0); m2 is 0, 1, or 2 (e.g., 0); each R 2 and R 4 is independently selected from the group consisting of: halo, CN, C1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, OH, and NR d R e ; m6 is 0 or 1; R 1 is selected from the group consisting of: (a) C(O)OH, (b) C(O)NR d R e , and (c) C(O)OC1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with 1-3 R c ; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; and L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; each R L is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C1-6 haloalkoxy; -NR d R e ; C(=O)N(R f )2; S(O)0-2(C1-6 alkyl); S(O)0-2(C1-6 haloalkyl); S(O)1-2N(R f )2; -R b ; and C1-6 alkyl optionally substituted with 1-6 R c ; R e2 is H or C1-6 alkyl optionally substituted with 1-3 -F; Ring C is selected from the group consisting of: ; c1 is 0, 1, 2, or 3; each R Y is independently selected from the group consisting of: R a and R b ; R aN is H or C1-6 alkyl optionally substituted with 1-3 R c ; Y 1 and Y 2 are independently N, CH, or CR Y ; yy represents the point of attachment to the -C(O)NH- moiety; X is CH or N; L C is selected from the group consisting of: a bond, -CH2-, -CHR a -, -C(R a )2-, -N(R d )-, and O, provided that when X is N, then L C is other than O; each R a is independently selected from the group consisting of: (a) halo; (b) cyano; each R b is independently selected from the group consisting of: -(L b )b-R b1 and -R b1 , wherein: each b is independently 1, 2, or 3; each -L b is independently selected from the group consisting of: -O-; -N(H)-; -N(C 1-3 alkyl)-; -S(O) 0-2 -; C(=O); and C 1-3 alkylene; and each R b1 is independently selected from the group consisting of: C3-10 cycloalkyl, 4-10 membered heterocyclyl, C6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 R g ; each R c is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C1-6 haloalkoxy; -NR d R e ; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)OH; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); and S(O) 1-2 N(R f ) 2 ; each R d and R e is independently selected from the group consisting of: H; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC1-6 haloalkyl; C(=O)N(R f )2; S(O)1- 2 (C 1-6 alkyl); S(O) 1-2 (C 1-6 haloalkyl); S(O) 1-2 N(R f ) 2 ; and C 1-6 alkyl optionally substituted with 1-3 R h ; each R f is independently selected from the group consisting of: H and C 1-6 alkyl optionally substituted with 1-3 R h ; each R g is independently selected from the group consisting of: R h ; C1-3 alkyl; and C1- 3 haloalkyl; and each R h is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C1-6 haloalkoxy; -NH2, -N(H)(C1-3 alkyl); and -N(C1-3 alkyl)2. In the methods of preparing compounds of Formula (I-B) as depicted in Scheme B, the variables m4, R 4 , m2, R 2 , R 1 , R a , m6, L A3 , a3, L A1a , a1a, L A4 , L A1b , a1b, Ring C, L C , and X can be as defined herein for Formula (I-B). In some embodiments of the methods, the compounds of Formula (I-B) are compounds of Formula (I-B-1). In some embodiments of the methods, the compounds of Formula (I-B) are compounds of Formula (I-B-2). In some embodiments of the methods, the compounds of Formula (I-B) are compounds of Formula (I- B-3). In some embodiments of the methods of preparing compounds of Formula (I-B), the compounds of Formula (SI-BB) are compounds of Formula (SI-B). In some embodiments of the methods of preparing compounds of Formula (I-B), the compounds of Formula (SI-BB) are compounds of Formula (SI-B-1). In some embodiments of the methods of preparing compounds of Formula (I-B), the compounds of Formula (SI-BB) are compounds of Formula (SI-B-2). In some embodiments of the methods of preparing compounds of Formula (I-B), the compounds of Formula (SI-BB) are compounds of Formula (SI-B-3). In some embodiments of the methods, Ring C is , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 F; and R aN is C 1-3 alkyl. In some embodiments of the methods, a3 is 1; L A3 is -O-; a1b is 1; L A1b is -CH 2 -; each L A1a is independently selected from the group consisting of: -CH2- and -CH(Me)- , provided that 0-1 L A1a is -CH(Me)-; and L A4 is 4-10 membered nitrogen-containing heterocyclylene optionally substituted with 1-2 substituents independently selected from the group consisting of: -F and C1-3 alkyl optionally substituted with 1-3 -F. In some embodiments of the methods, the moiety is selected from the groups depicted in Table (L-I-B). In some embodiments of the methods, R e2 is H; and the methods comprise reacting compounds of Formula (SI-BB) with compounds of Formula (SII) under standard conditions for amide bond formation to provide compounds of Formula (I-A). Examples of standard conditions for amide bond formation include those described in Chem. Soc. Rev., 2009,38, 606-631, which is incorporated herein by reference in its entirety. In some embodiments, the methods comprise reacting compounds of Formula (SI-BB) with compounds of Formula (SII) in the presence of the carboxyl activating agent (e.g., HATU, HOBT, EDCI, DCC, DIC, etc.) and optionally a base (e.g., triethylamine or diisopropylethylamine) to provide compounds of Formula (I-A). Also provided herein are methods of preparing compounds of Formula (I-E), comprising reacting compounds of Formula (SI-EE) with compounds of Formula (SIII) to provide compounds of Formula (I-E), as depicted in Scheme E:

Scheme E wherein: m4 is 0, 1, 2, or 3 (e.g., 0); m2 is 0, 1, or 2 (e.g., 0); each R 2 and R 4 is independently selected from the group consisting of: halo, CN, C 1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, C1-3 haloalkoxy, OH, and NR d R e ; m6 is 0 or 1; R 1 is selected from the group consisting of: (a) C(O)OH, (b) C(O)NR d R e , and (c) C(O)OC 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R c ; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; a1d is an integer from 0 to 4, provided that a1a + a1d is from 1 to 4; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; each R L is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C 1-6 haloalkoxy; -NR d R e ; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); S(O)1-2N(R f )2; -R b ; and C1-6 alkyl optionally substituted with 1-6 R c ; R e3 is H or R L ; m8 is 0, 1, or 2; Ring C is selected from the group consisting of: ; c1 is 0, 1, 2, or 3; each R Y is independently selected from the group consisting of: R a and R b ; R aN is H or C1-6 alkyl optionally substituted with 1-3 R c ; Y 1 and Y 2 are independently N, CH, or CR Y ; yy represents the point of attachment to the -C(O)NH- moiety; X is CH or N; L C is selected from the group consisting of: a bond, -CH 2 -, -CHR a -, -C(R a ) 2 -, -N(R d )-, and O, provided that when X is N, then L C is other than O; each R a is independently selected from the group consisting of: each R b is independently selected from the group consisting of: -(L b )b-R b1 and -R b1 , wherein: each b is independently 1, 2, or 3; each -L b is independently selected from the group consisting of: -O-; -N(H)-; -N(C1-3 alkyl)-; -S(O)0-2-; C(=O); and C1-3 alkylene; and each R b1 is independently selected from the group consisting of: C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 R g ; each R c is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C 1-6 haloalkoxy; -NR d R e ; C(=O)C 1-6 alkyl; C(=O)C 1-6 haloalkyl; C(=O)OC 1-6 alkyl; C(=O)OC1-6 haloalkyl; C(=O)OH; C(=O)N(R f )2; S(O)0-2(C1-6 alkyl); S(O)0-2(C1-6 haloalkyl); and S(O)1-2N(R f )2; each R d and R e is independently selected from the group consisting of: H; C(=O)C 1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC1-6 haloalkyl; C(=O)N(R f )2; S(O)1- 2 (C 1-6 alkyl); S(O) 1-2 (C 1-6 haloalkyl); S(O) 1-2 N(R f ) 2 ; and C 1-6 alkyl optionally substituted with 1-3 R h ; each R f is independently selected from the group consisting of: H and C1-6 alkyl optionally substituted with 1-3 R h ; each R g is independently selected from the group consisting of: R h ; C1-3 alkyl; and C1- 3 haloalkyl; and each R h is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C1-6 haloalkoxy; -NH2, -N(H)(C1-3 alkyl); and -N(C1-3 alkyl)2. In the methods of preparing compounds of Formula (I-E) as depicted in Scheme E, the variables m4, R 4 , m2, R 2 , R 1 , R a , m6, L A3 , a3, L A1a , a1a, L A4 , L A1b , a1b, m8, Ring C, L C , and X can be as defined herein for Formula (I-E). In some embodiments of the methods, the compounds of Formula (I-E) are compounds of Formula (I-E-1). In some embodiments of the methods, the compounds of Formula (I-E) are compounds of Formula (I-E-2). In some embodiments of the methods, the compounds of Formula (I-E) are compounds of Formula (I- E-3). In some embodiments of the methods of preparing compounds of Formula (I-E), the compounds of Formula (SI-EE) are compounds of Formula (SI-E). In some embodiments, the compounds of Formula (SI-EE) are compounds of Formula (SI-E-1). In some embodiments, the compounds of Formula (SI-EE) are compounds of Formula (SI-E-2). In some embodiments of the methods of preparing compounds of Formula (I-E), the compounds of Formula (SIII) are compounds of Formula (I-Ind-2) or Formula (I-Ind-3). In some embodiments of the methods, Ring C is , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C1-3 alkyl optionally substituted with 1-3 F; and R aN is C1-3 alkyl. In some embodiments of the methods, a3 is 1; L A3 is -O-; a1a is 0; and L A4 is selected from the group consisting of: C 3-10 cycloalkylene optionally substituted with 1-3 R a ; and phenylene optionally substituted with 1-3 R a . In some embodiments of the methods, a3 is 1; L A3 is -O-; a1a is 0; a1b is 3 or 4; each L A1b is independently selected from the group consisting of: -CH2- and -CH(Me)- , provided that 0-1 of L A1b is -CH(Me)-; and L A4 is selected from the group consisting of: ) and . In some embodiments of the methods, a3 is 1; L A3 is -O-; a1a is 2, 3, or 4; and L A4 is 4-10 membered heterocyclylene optionally substituted with 1-3 R a . In some embodiments of the methods, R e3 is H; and the methods comprising reacting compounds of Formula (SII-EE) with compounds of Formula (SIII) under standard conditions for reductive amination. In some embodiments, the methods comprise reacting compounds of Formula (SII-EE) with compounds of Formula (SIII) in the presence of a reducing agent (e.g., sodium triacetoxyborohydride, sodium cyanoborohydride, or sodium borohydride) optionally in the presence of an acid (e.g., acetic acid). Also provided herein are methods of preparing compounds of Formula (I-E), comprising reacting compounds of Formula (SV) with compounds of Formula (SI-EE2) to provide compounds of Formula (I-E), as depicted in Scheme E2:

Scheme E2 wherein: m4 is 0, 1, 2, or 3 (e.g., 0); m2 is 0, 1, or 2 (e.g., 0); each R 2 and R 4 is independently selected from the group consisting of: halo, CN, C 1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, C1-3 haloalkoxy, OH, and NR d R e ; R 1 is selected from the group consisting of: (a) C(O)OH, (b) C(O)NR d R e , and (c) C(O)OC 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R c ; one of X T and X E is halo, and the other of X T and X E is selected from the group consisting of: -B(OH) 2 ; -BF3-; -B(OR T )2, wherein each R T is independently C1-3 alkyl optionally substituted with 1-3 R c ; and , wherein L B is C 2-6 alkylene optionally substituted with 1-4 R a ; m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L ) 2 -; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; each R L is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C 1-6 haloalkoxy; -NR d R e ; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); S(O)1-2N(R f )2; -R b ; and C1-6 alkyl optionally substituted with 1-6 R c ; m8 is 0, 1, or 2; Ring C is selected from the group consisting of: ; c1 is 0, 1, 2, or 3; each R Y is independently selected from the group consisting of: R a and R b ; R aN is H or C 1-6 alkyl optionally substituted with 1-3 R c ; Y 1 and Y 2 are independently N, CH, or CR Y ; yy represents the point of attachment to the -C(O)NH- moiety; X is CH or N; L C is selected from the group consisting of: a bond, -CH 2 -, -CHR a -, -C(R a ) 2 -, -N(R d )-, and O, provided that when X is N, then L C is other than O; each R a is independently selected from the group consisting of: y y y y y each R b is independently selected from the group consisting of: -(L b )b-R b1 and -R b1 , wherein: each b is independently 1, 2, or 3; each -L b is independently selected from the group consisting of: -O-; -N(H)-; -N(C1-3 alkyl)-; -S(O) 0-2 -; C(=O); and C 1-3 alkylene; and each R b1 is independently selected from the group consisting of: C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 R g ; each R c is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C1-6 haloalkoxy; -NR d R e ; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)OH; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); and S(O)1-2N(R f )2; each R d and R e is independently selected from the group consisting of: H; C(=O)C1-6 alkyl; C(=O)C 1-6 haloalkyl; C(=O)OC 1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)N(R f ) 2 ; S(O) 1- 2(C1-6 alkyl); S(O)1-2(C1-6 haloalkyl); S(O)1-2N(R f )2; and C1-6 alkyl optionally substituted with 1-3 R h ; each R f is independently selected from the group consisting of: H and C 1-6 alkyl optionally substituted with 1-3 R h ; each R g is independently selected from the group consisting of: R h ; C1-3 alkyl; and C1- 3 haloalkyl; and each R h is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C1-6 haloalkoxy; -NH2, -N(H)(C1-3 alkyl); and -N(C1-3 alkyl)2. In the methods of preparing compounds of Formula (I-E) as depicted in Scheme E2, the variables m4, R 4 , m2, R 2 , R 1 , R a , m6, L A3 , a3, L A1a , a1a, L A4 , L A1b , a1b, m8, Ring C, L C , and X can be as defined herein for Formula (I-E). In some embodiments of the methods, the compounds of Formula (I-E) are compounds of Formula (I-E-1). In some embodiments of the methods, the compounds of Formula (I-E) are compounds of Formula (I-E-2). In some embodiments of the methods, the compounds of Formula (I-E) are compounds of Formula (I- E-3). In some embodiments of the methods, Ring C is , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 F; and R aN is C 1-3 alkyl. In some embodiments of the methods, a3 is 1; L A3 is -O-; a1a is 0; and L A4 is selected from the group consisting of: C 3-10 cycloalkylene optionally substituted with 1-3 R a ; and phenylene optionally substituted with 1-3 R a . In some embodiments of the methods, a3 is 1; L A3 is -O-; a1a is 0; a1b is 3 or 4; each L A1b is independently selected from the group consisting of: -CH2- and -CH(Me)- , provided that 0-1 of L A1b is -CH(Me)-; and L A4 is selected from the group consisting of: and . In some embodiments of the methods, a3 is 1; L A3 is -O-; a1a is 2, 3, or 4; and L A4 is 4-10 membered heterocyclylene optionally substituted with 1-3 R a . In some embodiments of the methods, X T is -B(OH) 2 or -Bpin; and X E is halo. In some embodiments of the methods, X T is halo; and X E is -B(OH)2 or -Bpin. In some embodiments of the methods, R e3 is H; and the methods comprising reacting compounds of Formula (SII-EE2) with compounds of Formula (SV) under standard conditions for Suzuki coupling. In some embodiments, the methods comprise reacting compounds of Formula (SII-EE2) with compounds of Formula (SV) in the presence of a palladium catalyst (e.g., CataCXium A Pd G3). Also provided herein are methods of preparing compounds of Formula (I-Eb), comprising reacting compounds of Formula (SI-EE) with compounds of Formula (SIV) to provide compounds of Formula (I-Eb), as depicted in Scheme Eb:

Scheme Eb wherein: m4 is 0, 1, 2, or 3 (e.g., 0); m2 is 0, 1, or 2 (e.g., 0); each R 2 and R 4 is independently selected from the group consisting of: halo, CN, C 1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, C1-3 haloalkoxy, OH, and NR d R e ; m6 is 0 or 1; R 1 is selected from the group consisting of: (a) C(O)OH, (b) C(O)NR d R e , and (c) C(O)OC1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with 1-3 R c ; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; a1d is an integer from 0 to 4, provided that a1a + a1d is from 1 to 4; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; each R L is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C 1-6 haloalkoxy; -NR d R e ; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); S(O)1-2N(R f )2; -R b ; and C1-6 alkyl optionally substituted with 1-6 R c ; R e3 is H or R L ; n2 and n3 are independently 0, 1, or 2; m8 is 0, 1, or 2; Ring C is selected from the group consisting of: ; c1 is 0, 1, 2, or 3; each R Y is independently selected from the group consisting of: R a and R b ; R aN is H or C1-6 alkyl optionally substituted with 1-3 R c ; Y 1 and Y 2 are independently N, CH, or CR Y ; yy represents the point of attachment to the -C(O)NH- moiety; X is CH or N; L C is selected from the group consisting of: a bond, -CH 2 -, -CHR a -, -C(R a ) 2 -, -N(R d )-, and O, provided that when X is N, then L C is other than O; each R a is independently selected from the group consisting of: y y y y y each R b is independently selected from the group consisting of: -(L b )b-R b1 and -R b1 , wherein: each b is independently 1, 2, or 3; each -L b is independently selected from the group consisting of: -O-; -N(H)-; -N(C1-3 alkyl)-; -S(O) 0-2 -; C(=O); and C 1-3 alkylene; and each R b1 is independently selected from the group consisting of: C3-10 cycloalkyl, 4-10 membered heterocyclyl, C6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 R g ; each R c is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C1-6 haloalkoxy; -NR d R e ; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC1-6 haloalkyl; C(=O)OH; C(=O)N(R f )2; S(O)0-2(C1-6 alkyl); S(O)0-2(C1-6 haloalkyl); and S(O) 1-2 N(R f ) 2 ; each R d and R e is independently selected from the group consisting of: H; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC1-6 haloalkyl; C(=O)N(R f )2; S(O)1- 2 (C 1-6 alkyl); S(O) 1-2 (C 1-6 haloalkyl); S(O) 1-2 N(R f ) 2 ; and C 1-6 alkyl optionally substituted with 1-3 R h ; each R f is independently selected from the group consisting of: H and C1-6 alkyl optionally substituted with 1-3 R h ; each R g is independently selected from the group consisting of: R h ; C1-3 alkyl; and C1- 3 haloalkyl; and each R h is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C1-6 haloalkoxy; -NH2, -N(H)(C1-3 alkyl); and -N(C1-3 alkyl)2. In the methods of preparing compounds of Formula (I-Eb) as depicted in Scheme Eb, the variables m4, R 4 , m2, R 2 , R 1 , R a , m6, L A3 , a3, L A1a , a1a, L A4 , L A1b , a1b, n2, n3, m8, Ring C, L C , and X can be as defined herein for Formula (I-Eb). In some embodiments of the methods, the compounds of Formula (I-Eb) are compounds of Formula (I-Eb-1). In some embodiments of the methods, the compounds of Formula (I-Eb) are compounds of Formula (I-Eb-2). In some embodiments of the methods, the compounds of Formula (I-Eb) are compounds of Formula (I-Eb-3). In some embodiments of the methods of preparing compounds of Formula (I-Eb), the compounds of Formula (SI-EE) are compounds of Formula (SI-E). In some embodiments, the compounds of Formula (SI-EE) are compounds of Formula (SI-E-1). In some embodiments, the compounds of Formula (SI-EE) are compounds of Formula (SI-E-2). In some embodiments of the methods of preparing compounds of Formula (I-Eb), the compounds of Formula (SIV) are compounds of Formula (I-Ind-1). In some embodiments of the methods, Ring C is or , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C1-3 alkyl optionally substituted with 1-3 F; and R aN is C1-3 alkyl. In some embodiments of the methods, a3 is 1; L A3 is -O-; a1a is 0; and L A4 is selected from the group consisting of: C 3-10 cycloalkylene optionally substituted with 1-3 R a ; and phenylene optionally substituted with 1-3 R a . In some embodiments of the methods, a3 is 1; L A3 is -O-; a1a is 0; a1b is 3 or 4; each L A1b is independently selected from the group consisting of: -CH2- and -CH(Me)- , provided that 0-1 of L A1b is -CH(Me)-; and L A4 is selected from the group consisting of: ) and . In some embodiments of the methods, a3 is 1; L A3 is -O-; a1a is 2, 3, or 4; and L A4 is 4-10 membered heterocyclylene optionally substituted with 1-3 R a . In some embodiments of the methods, R e3 is H; and the methods comprising reacting compounds of Formula (SII-EE) with compounds of Formula (SIV) under standard conditions for reductive amination. In some embodiments, the methods comprise reacting compounds of Formula (SII-EE) with compounds of Formula (SIV) in the presence of a reducing agent (e.g., sodium triacetoxyborohydride, sodium cyanoborohydride, or sodium borohydride) optionally in the presence of an acid (e.g., acetic acid). Also provided herein are methods of preparing compounds of Formula (I-Eb), comprising reacting compounds of Formula (SV) with compounds of Formula (SI-Eb2) to provide compounds of Formula (I-Eb), as depicted in Scheme Eb2:

Scheme Eb2 m4 is 0, 1, 2, or 3 (e.g., 0); m2 is 0, 1, or 2 (e.g., 0); each R 2 and R 4 is independently selected from the group consisting of: halo, CN, C 1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, C1-3 haloalkoxy, OH, and NR d R e ; R 1 is selected from the group consisting of: (a) C(O)OH, (b) C(O)NR d R e , and (c) C(O)OC 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R c ; one of X T and X E is halo, and the other of X T and X E is selected from the group consisting of: -B(OH)2; -BF 3 -; -B(OR T )2, wherein each R T is independently C1-3 alkyl optionally substituted with 1-3 R c ; and , wherein L B is C 2-6 alkylene optionally substituted with 1-4 R a ; m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L ) 2 -; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; each R L is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C 1-6 haloalkoxy; -NR d R e ; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); S(O)1-2N(R f )2; -R b ; and C1-6 alkyl optionally substituted with 1-6 R c ; m8 is 0, 1, or 2; Ring C is selected from the group consisting of: ; c1 is 0, 1, 2, or 3; each R Y is independently selected from the group consisting of: R a and R b ; R aN is H or C 1-6 alkyl optionally substituted with 1-3 R c ; Y 1 and Y 2 are independently N, CH, or CR Y ; yy represents the point of attachment to the -C(O)NH- moiety; X is CH or N; L C is selected from the group consisting of: a bond, -CH 2 -, -CHR a -, -C(R a ) 2 -, -N(R d )-, and O, provided that when X is N, then L C is other than O; each R a is independently selected from the group consisting of: each R b is independently selected from the group consisting of: -(L b )b-R b1 and -R b1 , wherein: each b is independently 1, 2, or 3; each -L b is independently selected from the group consisting of: -O-; -N(H)-; -N(C1-3 alkyl)-; -S(O) 0-2 -; C(=O); and C 1-3 alkylene; and each R b1 is independently selected from the group consisting of: C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 R g ; each R c is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C1-6 haloalkoxy; -NR d R e ; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)OH; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); and S(O)1-2N(R f )2; each R d and R e is independently selected from the group consisting of: H; C(=O)C1-6 alkyl; C(=O)C 1-6 haloalkyl; C(=O)OC 1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)N(R f ) 2 ; S(O) 1- 2(C1-6 alkyl); S(O)1-2(C1-6 haloalkyl); S(O)1-2N(R f )2; and C1-6 alkyl optionally substituted with 1-3 R h ; each R f is independently selected from the group consisting of: H and C 1-6 alkyl optionally substituted with 1-3 R h ; each R g is independently selected from the group consisting of: R h ; C1-3 alkyl; and C1- 3 haloalkyl; and each R h is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C1-6 haloalkoxy; -NH2, -N(H)(C1-3 alkyl); and -N(C1-3 alkyl)2. In the methods of preparing compounds of Formula (I-Eb) as depicted in Scheme Eb2, the variables m4, R 4 , m2, R 2 , R 1 , R a , m6, L A3 , a3, L A1a , a1a, L A4 , L A1b , a1b, n2, n3, m8, Ring C, L C , and X can be as defined herein for Formula (I-Eb). In some embodiments of the methods, the compounds of Formula (I-Eb) are compounds of Formula (I-Eb-1). In some embodiments of the methods, the compounds of Formula (I-Eb) are compounds of Formula (I-Eb-2). In some embodiments of the methods, the compounds of Formula (I-Eb) are compounds of Formula (I-Eb-3). In some embodiments of the methods, Ring C is , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 F; and R aN is C 1-3 alkyl. In some embodiments of the methods, a3 is 1; L A3 is -O-; a1a is 0; and L A4 is selected from the group consisting of: C 3-10 cycloalkylene optionally substituted with 1-3 R a ; and phenylene optionally substituted with 1-3 R a . In some embodiments of the methods, a3 is 1; L A3 is -O-; a1a is 0; a1b is 3 or 4; each L A1b is independently selected from the group consisting of: -CH2- and -CH(Me)- , provided that 0-1 of L A1b is -CH(Me)-; and L A4 is selected from the group consisting of: ) and . In some embodiments of the methods, a3 is 1; L A3 is -O-; a1a is 2, 3, or 4; and L A4 is 4-10 membered heterocyclylene optionally substituted with 1-3 R a . In some embodiments of the methods, X T is -B(OH) 2 or -Bpin; and X E is halo. In some embodiments of the methods, X T is halo; and X E is -B(OH) 2 or -Bpin. In some embodiments of the methods, R e3 is H; and the methods comprising reacting compounds of Formula (SII-Eb2) with compounds of Formula (SV) under standard conditions for Suzuki coupling. In some embodiments, the methods comprise reacting compounds of Formula (SII-Eb2) with compounds of Formula (SV) in the presence of a palladium catalyst (e.g., CataCXium A Pd G3). Also provided herein are methods of preparing compounds of Formula (I) wherein R 1 is C(O)OH, as depicted in Scheme I, the methods comprising: reacting compounds of Formula (I) wherein R 1 is C(O)OC1-6 alkyl with an acid or a base to provide compounds of Formula (I) wherein R 1 is C(O)OH:

Scheme I wherein: Ring A is selected from the group consisting of: (a) C 3-15 cycloalkylene or 3-15 membered heterocyclylene, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; and (b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 substituents independently selected from the group consisting of: R a and R b ; each R 2 , R 3 , R 4 , and R 5 is independently selected from the group consisting of: halo, CN, C1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, C1-3 haloalkoxy, OH, and NR d R e ; m2 is 0, 1, or 2; m3 and m4 are independently 0, 1, 2, or 3; m5 is 0, 1, 2, 3, or 4; L is –(L A )n1–, wherein L A and n1 are defined according to (AA) or (BB): (AA) n1 is an integer from 3 to 15; and each L A is independently selected from the group consisting of: L A1 , L A3 , and L A4 , provided that 1-3 occurrences of L A is L A4 ; n1 is an integer from 0 to 20; and each L A is independently selected from the group consisting of: L A1 and L A3 ; each L A1 is independently selected from the group consisting of: -CH 2 -, -CHR L -, and - C(R L )2-; each L A3 is independently selected from the group consisting of: -N(R d )-, -N(R b )-, -O- , -S(O) 0-2 -, and C(=O); each L A4 is independently selected from the group consisting of: (a) C 3-15 cycloalkylene or 3-15 membered heterocyclylene, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; and (b) C 6-15 arylene or 5-15 membered heteroarylene, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; provided that L does not contain any N-O, O-O, N-N, N-S(O) 0 , or O-S(O) 0-2 bonds; wherein each R L is independently selected from the group consisting of: halo; cyano; - OH; -C 1-6 alkoxy; -C 1-6 haloalkoxy; -NR d R e ; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); S(O) 1-2 N(R f ) 2 ; -R b ; and C 1-6 alkyl optionally substituted with 1-6 R c ; Ring C is selected from the group consisting ; c1 is 0, 1, 2, or 3; each R Y is independently selected from the group consisting of: R a and R b ; R aN is H or C1-6 alkyl optionally substituted with 1-3 R c ; Y 1 and Y 2 are independently N, CH, or CR Y ; yy represents the point of attachment to L; X is CH, C, or N; the is a single bond or a double bond; L C is selected from the group consisting of: a bond, -CH 2 -, -CHR a -, -C(R a ) 2 -, -N(R d )-, and O, provided that when X is N, then L C is other than O; each R a is independently selected from the group consisting of: (q) y , y , y y , p y ; each R b is independently selected from the group consisting of: -(L b )b-R b1 and -R b1 , wherein: each b is independently 1, 2, or 3; each -L b is independently selected from the group consisting of: -O-; -N(H)-; -N(C 1-3 alkyl)-; -S(O)0-2-; C(=O); and C1-3 alkylene; and each R b1 is independently selected from the group consisting of: C3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 R g ; each R c is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C1-6 haloalkoxy; -NR d R e ; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC1-6 haloalkyl; C(=O)OH; C(=O)N(R f )2; S(O)0-2(C1-6 alkyl); S(O)0-2(C1-6 haloalkyl); and S(O) 1-2 N(R f ) 2 ; each R d and R e is independently selected from the group consisting of: H; C(=O)C 1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC1-6 haloalkyl; C(=O)N(R f )2; S(O)1- 2(C1-6 alkyl); S(O)1-2(C1-6 haloalkyl); S(O)1-2N(R f )2; and C1-6 alkyl optionally substituted with 1-3 R h ; each R f is independently selected from the group consisting of: H and C1-6 alkyl optionally substituted with 1-3 R h ; each R g is independently selected from the group consisting of: R h ; C 1-3 alkyl; and C 1- 3 haloalkyl; and each R h is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C 1-6 haloalkoxy; -NH 2 , -N(H)(C 1-3 alkyl); and -N(C 1-3 alkyl) 2 . In the methods of preparing compounds of Formula (I) wherein R 1 is C(O)OH, as depicted in Scheme I, m5, R 5 , m4, R 4 , m3, R 3 , m2, R 2 , Ring A, L, Ring C, L C , and X can be as defined for Formula (I) herein. In some embodiments of the methods, L is (L-1). In some embodiments of the methods, L is (L-2). In some embodiments of the methods, L is (L-3). In some embodiments of the methods, L is (L-3a). In some embodiments of the methods, L is (L- 3b). In some embodiments of the methods, L is (L-3c). In some embodiments of the methods, L is (L-3d). In some embodiments of the methods, L is (L-4). In some embodiments, the methods provide compounds of Formula (I-A), wherein R 1 is C(O)OH. In some embodiments, the methods provide compounds of Formula (I-B), wherein R 1 is C(O)OH. In some embodiments, the methods provide compounds of Formula (I-C), wherein R 1 is C(O)OH. In some embodiments, the methods provide compounds of Formula (I-D), wherein R 1 is C(O)OH. In some embodiments, the methods provide compounds of Formula (I-E), wherein R 1 is C(O)OH. In some embodiments, the methods provide compounds of Formula (I-Ea), wherein R 1 is C(O)OH. In some embodiments, the methods provide compounds of Formula (I-Eb), wherein R 1 is C(O)OH. In some embodiments, the methods provide compounds of Formula (I-F), wherein R 1 is C(O)OH. In some embodiments, the methods provide compounds of Formula (I-G), wherein R 1 is C(O)OH. In some embodiments, the method comprising reacting compounds of Formula (I) wherein R 1 is C(O)O t Bu with an acid (e.g., TFA or HCl) to provide compounds of Formula (I) wherein R 1 is C(O)OH. EXAMPLES In some of the examples disclosed herein, the final product of a described chemical reaction sequence is structurally depicted with an enhanced stereochemical or1 notation at one stereogenic center. In some such examples, in the chemical name of the same compound, this stereogenic center is assigned a tentative configuration (e.g., (R)- or (S)-) based on the wedge/dash representation of the structural formula. However, this stereogenic center should be understood to have a configuration consistent with the or1 notation. Specifically, this stereogenic center has been resolved, but its specific configuration has not been determined. Accordingly, unless otherwise specified, starting materials and intermediates leading to this compound incorporate the or1 notation at this stereogenic center, notwithstanding the tentative assignments provided in their chemical names. For example, Compound 289a in Example 116 is a single stereoisomer selected from: 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2- ((1S)-6-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)amino)-2-oxoethyl)-6- azaspiro[2.5]octan-1-yl)ethoxy)-2-methylphenyl)picolinic acid; and 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2- ((1R)-6-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)amino)-2-oxoethyl)-6- azaspiro[2.5]octan-1-yl)ethoxy)-2-methylphenyl)picolinic acid. The intermediate product provided in Step A of the same example incorporates the or1 notation. It is therefore a single stereoisomer selected from: tert-butyl (S)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquino lin-2(1H)- yl)-3-(3-(2-(6-(2-ethoxy-2-oxoethyl)-6-azaspiro[2.5]octan-1- yl)ethoxy)-2- methylphenyl)picolinate; and tert-butyl (R)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquino lin-2(1H)- yl)-3-(3-(2-(6-(2-ethoxy-2-oxoethyl)-6-azaspiro[2.5]octan-1- yl)ethoxy)-2- methylphenyl)picolinate. For further clarification, a chemical name that takes into account the or1 notation is provided for the final product having an enhanced stereochemical or1 notation at one stereogenic center. This chemical name is enclosed in brackets (i.e., “[]”). In these chemical names, the prefix “rel” means that the stereochemical configuration shown in a chemical name is relative. To illustrate, when a compound contains one stereogenic center, and its chemical name starts with the prefix “rel,” then this stereogenic center is resolved, but its absolute configuration is either (R)- or (S)-. As such, it should be labelled with an or1 enhanced stereochemical notation in its corresponding structure. For example, the chemical name that takes into account the or1 notation for Compound 289a is: [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(3-(2- ((1S)-6-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)amino)-2-oxoethyl)-6- azaspiro[2.5]octan-1-yl)ethoxy)-2-methylphenyl)picolinic acid], as included in Example 116. When a stereogenic center is labelled with an asterisk (“*”) in a chemical name of a compound having more than one stereogenic center, the stereogenic center labeled with the asterisk is resolved, but its absolute configuration is either (R)- or (S)-. As such, in a chemical name wherein one stereogenic center is labelled with the asterisk, that stereocenter should be labelled with an or1 enhanced stereochemical notation in its corresponding structure. For example, the chemical name containing an asterisk for Compound 282b is: [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3- (((1S,4r)-4-((2S*)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-6-yl)piperazin-1- yl)-2-methylpropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid], as included in Example 185. Example 1. Preparation of Compound 121 6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2- oxoethyl)piperidin-4- yl)propoxy)-2-methylphenyl)picolinic acid Method 1 Step A. tert-Butyl 4-(3-(3-bromo-2-methylphenoxy)propyl)piperidine-1-carboxylat e Combined 3-bromo-2-methylphenol (1 equiv.), tert-butyl 4-(3-bromopropyl)piperidine-1- carboxylate (1 equiv.), and potassium carbonate (3 equiv.) in DMF and heated at 50 °C for 16 h. Diluted reaction mixture with ethyl acetate and brine. Separated layers, dried organic layer over anhydrous magnesium sulfate, filtered, and concentrated to an oil that was purified by silica gel chromatography (heptane/ethyl acetate gradient). Combined and concentrated product containing fractions to afford tert-butyl 4-(3-(3-bromo-2- methylphenoxy)propyl)piperidine-1-carboxylate as an oil that was used without further purification assuming theoretical yield. MS (ESI) m/z: 412.2 [M + H] + . Step B.4-(3-(3-Bromo-2-methylphenoxy)propyl)piperidine, HCl Combined tert-butyl 4-(3-(3-bromo-2-methylphenoxy)propyl)piperidine-1-carboxylat e (1 equiv.) and 4 M HCl in dioxane (10 equiv.) in DCM and let mix under ambient temperature for 40 h. Concentrated mixture to dryness, triturated residue with diethyl ether and hexanes, re- evaporated, and dried the resulting solids overnight under vacuum to afford 4-(3-(3-bromo-2- methylphenoxy)propyl)piperidine, HCl. MS (ESI) m/z: 312.2 [M + H] + . Step C. Methyl 2-(4-(3-(3-bromo-2-methylphenoxy)propyl)piperidin-1-yl)aceta te Combined 4-(3-(3-bromo-2-methylphenoxy)propyl)piperidine, HCl (1 equiv.), methyl 2- bromoacetate (1 equiv.), and potassium carbonate (4 equiv.) in DMF and heated overnight at 50 °C for 16 h. Diluted reaction mixture with ethyl acetate and brine. Separated layers, dried organic over anhydrous magnesium sulfate, filtered, and concentrated to an oil that was purified by silica gel chromatography (heptane/ethyl acetate (w/ 10% of a 7N NH3 solution in methanol)). Combined and concentrated product containing fractions to afford methyl 2-(4-(3- (3-bromo-2-methylphenoxy)propyl)piperidin-1-yl)acetate. MS (ESI) m/z: 384.2 [M+H] + . Step D. Methyl 2-(4-(3-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2- yl)phenoxy)propyl)piperidin-1-yl)acetate Methyl 2-(4-(3-(3-bromo-2-methylphenoxy)propyl)piperidin-1-yl)aceta te (1 equiv.), bis(pinacolato)diboron (1.7 equiv.), and PdCl2(dppf)-CH2Cl2adduct (0.1 equiv.) were combined in 1,4-dioxane. Potassium acetate (3 equiv.) was then added, and the mixture was stirred at 80 °C for 16 h. Diluted reaction mixture with ethyl acetate, water, and brine. Separated layers and washed organic layer with an additional brine. Dried organic layer over anhydrous magnesium sulfate, filtered, and concentrated to dryness under reduced pressure. Purified residue on silica gel (heptane/ethyl acetate gradient). Product containing fractions were combined, concentrated, and dried under vacuum to afford methyl 2-(4-(3-(2-methyl-3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)propyl) piperidin-1-yl)acetate which was used without further purification. MS (ESI) m/z: 432.4 [M+H] + . Step E. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-(1-(2-methoxy-2-oxoethyl)piperidin-4-yl)propoxy) -2-methylphenyl)picolinate Combined tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)- 3-bromopicolinate and methyl 2-(4-(3-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2- yl)phenoxy)propyl)piperidin-1-yl)acetate in 1,4-dioxane. Added potassium phosphate (4 equiv.) and CataCxium Pd G3 (0.05 equiv.). Let mixture stir at 100 °C for 1 hr under microwave heating. Diluted reaction mixture with ethyl acetate, water, and brine. Separated layers and washed organic layer with an additional brine. Dried over anhydrous magnesium sulfate, filtered, and concentrated to dryness under reduced pressure to afford tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-(1-(2-methoxy-2- oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinate which was used without further purification assuming theoretical yield. MS (ESI) m/z: 790.4 [M+H] + . Step F. 2-(4-(3-(3-(6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)pro pyl)piperidin-1-yl)acetic acid tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1- (2-methoxy-2-oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl )picolinate (1 equiv.) was dissolved in a mixture of THF, methanol, and water. Lithium hydroxide (10 equiv.) was added and the mixture was stirred under ambient temperature for 16 h. The pH was adjusted to ~ 5-6 with formic acid and the volatiles were removed under reduced pressure. The residue was triturated in water to provide solids that were filtered, rinsed with water, and dried under vacuum to afford 2-(4-(3-(3-(6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)piperidin-1- yl)acetic acid that was used without further purification. MS (ESI) m/z: 776.4 [M+H] + . Step G. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2- oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinate 2-(4-(3-(3-(6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin-2(1H)-yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)propyl)piperidi n-1-yl)acetic acid (1 equiv.) was dissolved in a mixture of DMF and DIEA (5 equiv.). HATU (1.2 equiv.) was added to the solution and let mix at ambient temperature for 5 min. 3-(6-Amino-1-methyl-1H-indazol-3- yl)piperidine-2,6-dione (1 equiv.) was added to the solution and the mixture was stirred at ambient temperature for 16 h. Additional amounts of DIEA and HATU (1/3 amount originally used) to the reaction mixture and stirred for 5 min. Added 150 mg additional of 3-(6-amino-1- methyl-1H-indazol-3-yl)piperidine-2,6-dione (1 equiv.) to the solution and let continue to mix at ambient temperature for 1 h. Diluted reaction mixture with ethyl acetate and brine. Separated layers, dried organic over anhydrous magnesium sulfate, filtered, and concentrated to an oil affording tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)- 3-(3-(3-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-inda zol-6-yl)amino)-2- oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinate that was used without further purification assuming theoretical yield. MS (ESI) m/z: 1016.4 [M+H] + . Step H. 6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3- (1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl )amino)-2- oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinic acid Dissolved tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)- 3-(3-(3-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-inda zol-6-yl)amino)-2- oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinate (1 equiv.) in a mixture of DCM and trifluoroacetic acid (50 equiv.). Let reaction stir for 16 h at ambient temperature. Concentrated reaction mixture to dryness under reduced pressure and purified by preparative RP-HPLC (water/acetonitrile, 0.1% formic acid in each). Combined and lyophilized pure product containing fractions to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)propoxy)-2-met hylphenyl)picolinic acid. MS (ESI) m/z: 960.4 [M+H] + ; H NMR (500 MHz, DMSO) δ 12.87 (s, 1H), 10.88 (s, 1H), 9.97 – 9.63 (m, 1H), 8.05 – 8.00 (m, 2H), 7.81 – 7.76 (m, 1H), 7.65 – 7.59 (m, 2H), 7.50 – 7.42 (m, 3H), 7.41 – 7.31 (m, 2H), 7.24 – 7.15 (m, 1H), 7.13 – 7.06 (m, 1H), 7.00 – 6.94 (m, 1H), 6.91 – 6.85 (m, 1H), 6.65 – 6.60 (m, 1H), 4.98 (s , 2H), 4.36 – 4.29 (m, 1H), 3.99 – 3.94 (m, 2H), 3.92 (s , 5H), 3.14 – 3.11 (m, 2H), 3.06 – 3.00 (m, 2H), 2.90 – 2.87 (m, 2H), 2.68 – 2.56 (m, 1H), 2.40 – 2.07 (m, 1H), 1.90 (s , 3H), 1.82 – 1.60 (m, 5H), 1.48 – 1.02 (m, 8H). Method 2: Step A. tert-butyl 4-(3-(3-bromo-2-methylphenoxy)propyl)piperidine-1-carboxylat e A mixture of 3-bromo-2-methyl-phenol (2 g, 10.69 mmol, 1 equiv.), K2CO3 (4.43 g, 32.08 mmol, 3 eq.), tert-butyl 4-(3-hydroxypropyl)piperidine-1-carboxylate (3.27 g, 10.69 mmol, 1 equiv.) in MeCN (20 mL) was degassed and purged with N2 three times. The mixture was stirred at 60 °C for 12 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~7% ethyl acetate/petroleum ether) to give tert-butyl 4-(3-(3-bromo-2- methylphenoxy)propyl)piperidine-1-carboxylate (1.8 g, 4.37 mmol, 40.82% yield) as a colorless oil. 1 H NMR (400 MHz, CDCl 3 ) δ = 7.09 - 7.07 (m, 1H), 6.91 (t, J = 8.0 Hz, 1H), 6.68 (s, 1H), 3.86 (t, J = 6.4 Hz, 2H), 2.63 (t, J = 12.0 Hz, 2H), 2.23 (s, 3H), 1.79 - 1.69 (m, 2H), 1.69 - 1.50 (m, 4H), 1.39 (s, 9H), 1.38 - 1.30 (m, 3H), 1.11 - 0.98 (m, 2H) Step B.4-(3-(3-bromo-2-methylphenoxy)propyl)piperidine A mixture of tert-butyl 4-(3-(3-bromo-2-methylphenoxy)propyl)piperidine-1-carboxylat e (1.8 g, 4.37 mmol, 1 equiv.) in HCl/EtOAc (20 mL) was stirred at 20 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give 4-(3-(3-bromo-2- methylphenoxy)propyl)piperidine (1.5 g, crude) as a white solid. MS (ESI) m/z: 313.9 [M+3] + . Step C. Ethyl 2-(4-(3-(3-bromo-2-methylphenoxy)propyl)piperidin-1-yl)aceta te A mixture of 4-(3-(3-bromo-2-methylphenoxy)propyl)piperidine (1 g, 3.20 mmol, 1 equiv.), ethyl 2-bromoacetate (534.84 mg, 3.20 mmol, 354.20 uL, 1 equiv.), and K 2 CO 3 (885.24 mg, 6.41 mmol, 2 equiv.) in MeCN (15 mL) was degassed and purged with N2 three times. The mixture was stirred at 60 °C for 2 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~45% ethyl acetate/petroleum ether) to give ethyl 2-(4-(3-(3-bromo-2- methylphenoxy)propyl)piperidin-1-yl)acetate (0.5 g, 1.26 mmol, 39.19% yield) as a yellow oil MS (ESI) m/z: 399.9 [M+3] + . 1 H NMR (400 MHz, CD3OD) δ = 7.14 (d, J = 8.0 Hz, 1H), 6.98 (t, J = 8.0 Hz, 1H), 6.75 (d, J = 8.0 Hz, 1H), 4.19 (q, J = 7.2 Hz, 2H), 3.93 (t, J = 6.4 Hz, 2H), 3.23 - 3.18 (m, 2H), 2.95 (d, J = 11.6 Hz, 2H), 2.31 (s, 3H), 2.18 - 2.11 (m, 2H), 1.84 - 1.79 (m, 2H), 1.72 - 1.64 (m, 4H), 1.59 - 1.53 (m, 2H), 1.46 - 1.40 (m, 4H), 1.35 (dd, J = 3.2, 10.8 Hz, 2H), 1.28 (t, J = 7.2 Hz, 3H) Step D. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-(1-(2-ethoxy-2-oxoethyl)piperidin-4-yl)propoxy)- 2-methylphenyl)picolinate A mixture of ethyl 2-(4-(3-(3-bromo-2-methylphenoxy)propyl)piperidin-1-yl)aceta te (500 mg, 1.26 mmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)picolinate (768.89 mg, 1.26 mmol, 1 equiv.), [2-(2-aminophenyl)phenyl]palladium(1+);bis(1- adamantyl)-butyl-phosphane;methanesulfonate (91.41 mg, 125.52 umol, 0.1 equiv.), and K 3 PO 4 (1.5 M, 2.51 mL, 3 equiv.) in 1,4-dioxane (5 mL) was degassed and purged with N 2 three times. The mixture was stirred at 80 °C for 12 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~5% ethyl acetate/petroleum ether) to give tert-butyl 6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2( 1H)-yl)-3-(3-(3-(1-(2-ethoxy- 2-oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinate (800 mg, 696.51 umol, 55.49% yield, 70% purity) as a yellow solid. MS (ESI) m/z: 804.5 [M+H] + . Step E. 2-(4-(3-(3-(6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)pro pyl)piperidin-1-yl)acetic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-ethoxy-2-oxoethyl)piperidin-4-yl)pro poxy)-2- methylphenyl)picolinate (342.86 mg, 298.51 umol, 70% purity, 1 equiv.) in THF (3 mL) was added LiOH (1 M, 895.52 uL, 3 equiv.). The mixture was stirred at 20 °C for 1 hour. The reaction mixture was adjusted pH to 5, filtered, and concentrated under reduced pressure to give 2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin-2(1H)-yl)-2- (tert-butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)propyl)pi peridin-1-yl)acetic acid (150 mg, 193.31 umol, 64.76% yield) as a yellow solid. MS (ESI) m/z: 776.6 [M+H] + . Step F. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2- oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinate A mixture of 2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)pro pyl)piperidin-1-yl)acetic acid (70 mg, 90.21 umol, 1 equiv.), 3-(6-amino-1-methyl-1H-indazol-3-yl)piperidine-2,6-dione (23.30 mg, 90.21 umol, 1 equiv.), HATU (37.73 mg, 99.23 umol, 1.1 equiv.), and TEA (27.39 mg, 270.64 umol, 37.67 uL, 3 equiv.) in DMF (1 mL) was degassed and purged with N 2 three times. The mixture was stirred at 25 °C for 12 hours under N 2 atmosphere. The solution was poured into water (2 mL), filtered, and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2- oxoethyl)piperidin-4- yl)propoxy)-2-methylphenyl)picolinate (60 mg, 59.04 umol, 65.45% yield) as a yellow solid. MS (ESI) m/z: 508.9 [M/2+H] + . Step G. 6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3- (1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl )amino)-2- oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-meth yl-1H-indazol-6-yl)amino)-2- oxoethyl)piperidin-4-yl) propoxy)-2-methylphenyl)picolinate (60 mg, 59.04 umol, 1 equiv.) in DCM (0.5 mL) was added TFA (0.5 mL). The mixture was stirred at 20 °C for 0.5 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex luna C18 150×25mm×10um; mobile phase: [water (formic acid)-MeCN]; B%: 27%-57%,15min) to give 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2- ((3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)prop oxy)-2- methylphenyl)picolinic acid (35.4 mg, 35.93 umol, 60.86% yield, 97.45% purity) as an off- white solid. MS (ESI) m/z: 960.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.99 - 12.69 (m, 1H), 10.86 (s, 1H), 10.11 - 9.72 (m, 1H), 8.07 - 7.98 (m, 2H), 7.82 - 7.76 (m, 1H), 7.67 - 7.59 (m, 2H), 7.55 - 7.25 (m, 6H), 7.24 - 7.17 (m, 1H), 7.13 - 7.06 (m, 1H), 7.00 - 6.93 (m, 1H), 6.88 (d, J = 8.4 Hz, 1H), 6.63 (d, J = 8.0 Hz, 1H), 5.03 - 4.94 (m, 2H), 4.32 (dd, J = 5.2, 9.2 Hz, 1H), 4.00 - 3.94 (m, 2H), 3.92 (s, 5H), 3.05 - 3.01 (m, 4H), 2.69 - 2.60 (m, 3H), 2.23 - 2.12 (m, 3H), 1.90 (s, 3H), 1.83 - 1.67 (m, 5H), 1.46 - 1.25 (m, 6H) Example 2. Preparation of Compound 108 2-(5-(1-(((3r,5r,7r)-adamantan-1-yl)methyl)-5-methyl-1H-pyra zol-4-yl)-6-((6-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino)-6-oxohe xyl)carbamoyl)pyridin-2-yl)- N-(benzo[d]thiazol-2-yl)-1,2,3,4-tetrahydroisoquinoline-8-ca rboxamide Step A. Methyl 6-(3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6-( 8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) - yl)picolinamido)hexanoate 3-(1-(Adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-( benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)picolinic acid (1 g, 1.52 mmol, 1 equiv.) and methyl 6-aminohexanoate (551.48 mg, 3.04 mmol, 2 equiv., HCl) were combined in DMF (10 mL). To this mixture was added HATU (692.58 mg, 1.82 mmol, 1.2 equiv.) and DIEA (588.53 mg, 4.55 mmol, 793.17 mL, 3 equiv.). The reaction mixture was stirred at 25 °C for 1 hour and then added dropwise to water 30 (mL), stirred briefly, and filtered to give a solid that was purified by silica gel chromatography (heptane/ethyl acetate gradient). Combined and concentrated product containing fractions to afford methyl 6-(3-(1-(adamantan-1-ylmethyl)-5- methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl) -3,4-dihydroisoquinolin- 2(1H)-yl)picolinamido)hexanoate (690 mg, 816.42 mmol, 53.79 % yield). MS (ESI) m/z: 786.9 [M+H] + . Step B. 6-(3-(1-(Adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6-( 8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) - yl)picolinamido)hexanoic acid To a solution of methyl 6-(3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6-( 8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)picolinamido)hexanoate (670 mg, 852.42 mmol, 1 equiv.) in THF (7 mL) was added an aqueous solution of LiOH•H2O (1 M, 2.56 mL, 3 equiv.). The mixture was stirred at 25 °C for 12 hours. The pH of the reaction mixture was adjusted to 6 using an aqueous solution of HCl (1 M). The resulting solids were stirred briefly, filtered to give a crude product that was triturated with MeCN (5 mL) at 25 °C for 0.5 hour and then filtered to afford 6-(3-(1-(Adamantan-1-ylmethyl)-5-methyl-1H-pyrazol- 4-yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoqui nolin-2(1H)- yl)picolinamido)hexanoic acid (552.07 mg, 642.20 mmol, 75.34 % yield). MS (ESI) m/z: 772.1 [M+H] + ; 1 H NMR (400 MHz, DMSO-d6) δ 8.10 (t, J = 5.2 Hz, 1H), 7.90 (d, J = 7.6 Hz, 1H), 7.66 (t, J = 6.8 Hz, 2H), 7.45 (d, J = 8.8 Hz, 1H), 7.41 - 7.26 (m, 4H), 7.26 - 7.21 (m, 2H), 6.93 (d, J = 8.8 Hz, 1H), 5.05 (s, 2H), 3.89 (t, J = 5.6 Hz, 2H), 3.04 - 2.98 (m, 4H), 2.10 (t, J = 7.2 Hz, 2H), 2.05 (s, 3H), 1.92 (s, 3H), 1.68 - 1.61 (m, 4H), 1.58 - 1.51 (m, 10H), 1.41 - 1.36 (m, 2H), 1.33 - 1.27 (m, 2H), 1.15 - 1.08 (m, 2H). Step C.2-(5-(1-(((3r,5r,7r)-adamantan-1-yl)methyl)-5-methyl-1H-py razol-4-yl)-6-((6-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino)-6- oxohexyl)carbamoyl)pyridin-2-yl)-N-(benzo[d]thiazol-2-yl)-1, 2,3,4- tetrahydroisoquinoline-8-carboxamide To a solution of 6-(3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6-( 8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)picolinamido)hexanoic acid (80 mg, 103.63 mmol, 1 equiv.) and HATU (47.28 mg, 124.36 mmol, 1.2 equiv.) in DMF (1 mL) was added DIEA (40.18 mg, 310.89 mmol, 54.15 mL, 3 equiv.) and 3-(7-amino-1- methyl-1H-indazol-3-yl)piperidine-2,6-dione (29.44 mg, 113.99 mmol, 1.1 equiv.). The mixture was stirred at 25 °C for 12 hours, diluted with DMF (1.5 mL), and then purified by preparative RP HPLC (water/ACN, both with 0.1% FA modifier) to afford 2-(5-(1-(((3r,5r,7r)- adamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-((6-((3-( 2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)amino)-6-oxohexyl)carbamoyl)pyridin-2 -yl)-N-(benzo[d]thiazol-2- yl)-1,2,3,4-tetrahydroisoquinoline-8-carboxamide (24.94 mg, 23.73 mmol, 22.90 % yield, 96.3% purity). MS (ESI) m/z: 1013.2 [M+H] + ; 1 H NMR (400 MHz, DMSO-d6) δ 12.94 - 12.76 (m, 1H), 10.89 (s, 1H), 9.78 (s, 1H), 8.10 (t, J = 5.6 Hz, 1H), 8.02 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 7.6 Hz, 1H), 7.59 (d, J = 7.6 Hz, 2H), 7.50 - 7.43 (m, 2H), 7.42 - 7.38 (m, 1H), 7.37 - 7.30 (m, 2H), 7.25 (s, 1H), 7.11 - 7.02 (m, 2H), 6.95 (d, J = 8.8 Hz, 1H), 4.99 (s, 2H), 4.44 - 4.30 (m, 1H), 4.00 (s, 3H), 3.89 (t, J = 5.2 Hz, 2H), 3.68 (s, 2H), 3.09 - 2.97 (m, 4H), 2.65 - 2.61 (m, 1H), 2.32 - 2.30 (m, 1H), 2.20 - 2.15 (m, 1H), 2.06 (s, 3H), 1.91 (s, 4H), 1.68 - 1.58 (m, 4H), 1.58 - 1.48 (m, 12H), 1.40 - 1.34 (m, 2H), 1.22 (d, J = 9.2 Hz, 2H). Example 3. Preparation of Compound 125a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(3- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino )-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Ethyl (1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexane-1-carboxylate To a solution of ethyl 4-hydroxycyclohexanecarboxylate (10 g, 58.06 mmol, 1 equiv.), 3- bromo-2-methyl-phenol (11.95 g, 63.87 mmol, 1.1 equiv.), PPh 3 (24.37 g, 92.90 mmol, 1.6 equiv.) in THF (100 mL) was added DIAD (18.79 g, 92.90 mmol, 18.06 mL, 1.6 equiv.) under 0 °C. Then the mixture was stirred at 25 °C for 16 h. The reaction mixture was concentrated under reduced pressure to give a residue that was purified by silica gel chromatography (ethyl acetate / petroleum ether gradient) to afford ethyl (1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexane-1-carboxylate (3.6 g, 10.55 mmol, 18.17 % yield). 1 H NMR (400 MHz, CDCl3) δ 7.14 (d, J = 8.0 Hz, 1H), 6.97 (t, J = 8.0 Hz, 1H), 6.80 (d, J = 8.0 Hz, 1H), 4.20 - 4.11 (m, 3H), 2.39 - 2.35 (m, 2H), 2.30 (s, 3H), 2.19 - 2.14 (m, 2H), 2.09 - 2.04 (m, 2H), 1.62 - 1.52 (m, 5H), 1.26 (t, J = 6.8 Hz, 3H). Step B. ((1r,4r)-4-(3-Bromo-2-methylphenoxy)cyclohexyl)methanol To a solution of ethyl 4-(3-bromo-2-methyl-phenoxy)cyclohexanecarboxylate (3.6 g, 10.55 mmol, 1 equiv.) in THF (15 mL) was added LiAlH4 (480.49 mg, 12.66 mmol, 1.2 equiv.) under 0 °C. The resulting mixture was then stirred at 25 °C for 1.5 h. The reaction was quenched by addition of a saturated aqueous solution of sodium sulfate (10 mL), and then extracted with ethyl acetate (10 mL × 3). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to afford ((1r,4r)-4-(3-Bromo-2- methylphenoxy)cyclohexyl)methanol (2.96 g, crude) that was used without further purification. 1 H NMR (400 MHz, CDCl3) δ 7.13 (d, J = 8.0 Hz, 1H), 6.97 (t, J = 8.0 Hz, 1H), 6.81 (d, J = 8.4 Hz, 1H), 4.13 - 4.08(m, 1H), 3.50 (d, J = 6.4 Hz, 2H), 2.30 (s, 3H), 2.20 - 2.14 (m, 2H), 1.95 - 1.86 (m, 2H), 1.53 - 1.45 (m, 3H), 1.14 - 1.04 (m, 2H). Step C. (1r,4r)-4-(3-Bromo-2-methylphenoxy)cyclohexane-1-carbaldehyd e To a solution of DMSO (3.09 g, 39.57 mmol, 3.09 mL, 4 equiv.) in DCM (10 mL) was added dropwise a solution of oxalyl chloride (2.51 g, 19.79 mmol, 1.73 mL, 2 equiv.) in DCM (2 mL) at -70 °C under a nitrogen atmosphere. The mixture was stirred at -70 °C for 1 h. ((1r,4r)-4-(3- Bromo-2-methylphenoxy)cyclohexyl)methanol (2.96 g, 9.89 mmol, 1 equiv.) in DCM (10 mL) was then added dropwise at -70 °C. The solution was stirred for 1 h at -70 °C. TEA (6.01 g, 59.36 mmol, 8.26 mL, 6 equiv.) was added, and the mixture was stirred at -70 °C for 0.5 h under a nitrogen atmosphere. The reaction mixture was quenched by addition water (10 mL), diluted with DCM (10 mL), and extracted with DCM (10 mL × 3). The combined organic layers were washed with water (5 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to afford (1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexane-1-carbaldehyde (3 g, crude) that was used without further purification. 1 H NMR (400 MHz, DMSO-d6) δ 9.62 - 9.59 (m, 1H), 7.16 - 7.01 (m, 3H), 4.36 - 4.23 (m, 1H), 2.39 - 2.31 (m, 1H), 2.25 - 2.18 (m, 3H), 2.03 - 1.92 (m, 4H), 1.50 - 1.38 (m, 4H). Step D. Ethyl (E)-3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)acrylat e To a solution of NaH (56.52 mg, 1.41 mmol, 60% purity, 2.1 equiv.) in THF (3 mL) at 0 °C was added ethyl 2-diethoxyphosphorylacetate (301.75 mg, 1.35 mmol, 267.04 mL, 2 equiv.). (1r,4r)-4-(3-Bromo-2-methylphenoxy)cyclohexane-1-carbaldehyd e (200 mg, 672.98 mmol, 1 equiv.) was then added, and the mixture was stirred at 25 °C for 6 h. The reaction mixture was quenched by addition of a saturated aqueous solution of ammonium chloride (5 mL) at 0 °C, and then extracted with ethyl acetate (10 mL × 3). The combined organic layers were washed with water (10 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue which was purified by silica gel chromatography (ethyl acetate / petroleum ether gradient) to afford the title compound (140 mg, 362.12 mmol, 53.81 % yield). 1 H NMR (400 MHz, CDCl3) δ 7.14 (d, J = 8.0 Hz, 1H), 7.01 - 6.87 (m, 2H), 6.80 (d, J = 8.4 Hz, 1H), 5.82 (dd, J = 1.2, 15.6 Hz, 1H), 4.21 - 4.15 (m, 2H), 4.16 - 4.07 (m, 1H), 2.30 (s, 3H), 2.26 - 2.16 (m, 3H), 1.97 - 1.89 (m, 2H), 1.58 - 1.48 (m, 2H), 1.33 - 1.26 (m, 5H). Step E. Ethyl 3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propanoate A mixture of ethyl (E)-3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]prop-2-enoate (1.6 g, 4.36 mmol, 1 equiv.) and PtO 2 (98.92 mg, 435.64 mmol, 0.1 equiv.) in EtOH (15 mL) was degassed and purged with hydrogen three times. The mixture was stirred at 25 °C for 3 hours under a hydrogen atmosphere (balloon, ~ 15 psi). The reaction mixture was filtered and concentrated under reduced pressure to give a residue that was purified by preparative RP- HPLC (water/acetonitrile with 0.1% formic acid in each). Combined and lyophilized pure product containing fractions to afford ethyl 3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propanoate (1.4 g, 3.79 mmol, 87.02 % yield). 1 H NMR (400 MHz, DMSO-d6) δ 7.17 - 7.10 (m, 1H), 7.06 (t, J = 8.0 Hz, 1H), 7.03 - 6.99 (m, 1H), 4.27 - 4.17 (m, 1H), 4.11 - 4.04 (m, 2H), 2.33 - 2.26 (m, 2H), 2.20 (s, 3H), 2.03 (d, J = 10.0 Hz, 2H), 1.75 (d, J = 12.0 Hz, 2H), 1.55 - 1.42 (m, 2H), 1.39 - 1.21 (m, 3H), 1.17 (t, J = 7.2 Hz, 3H), 1.11 - 0.97 (m, 2H). Step F. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1r,4r)-4-(3-ethoxy-3-oxopropyl)cyclohexyl)oxy)-2 -methylphenyl)picolinate A mixture of ethyl 3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propanoate (200 mg, 541.58 mmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (546.40 mg, 758.21 mmol, 1.4 equiv.), [2-(2-aminophenyl)phenyl]palladium(1+);bis(1- adamantyl)-butyl-phosphane;methanesulfonate (78.88 mg, 108.32 mmol, 0.2 equiv.), and an aqueous solution of potassium carbonate (1.5 M, 541.58 mL, 1.5 equiv.) in dioxane (2.5 mL) was degassed and purged with nitrogen three times. The mixture was stirred at 100 °C for 1 h under microwave heating. The reaction mixture was filtered and concentrated under reduced pressure to provide a residue that was purified by silica gel chromatography (ethyl acetate / petroleum ether gradient) to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-ethoxy-3-ox opropyl)cyclohexyl)oxy)-2- methylphenyl)picolinate (440 mg, crude). 1 H NMR (400 MHz, DMSO-d6) δ 8.02 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.60 (d, J = 7.5 Hz, 1H), 7.50 - 7.40 (m, 3H), 7.40 - 7.30 (m, 2H), 7.12 - 7.03 (m, 1H), 6.99 - 6.89 (m, 2H), 6.59 - 6.49 (m, 1H), 5.03 - 4.90 (m, 2H), 4.27 - 4.14 (m, 1H), 4.03 (d, J = 6.8 Hz, 2H), 3.87 (t, J = 5.6 Hz, 2H), 3.29 (s, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.29 (t, J = 7.6 Hz, 2H), 2.05 (d, J = 11.2 Hz, 1H), 1.84 (s, 3H), 1.79 - 1.70 (m, 2H), 1.50 - 1.42 (m, 2H), 1.23 (d, J = 8.4 Hz, 2H), 1.19 - 1.17 (m, 3H), 1.07 (s, 2H), 1.00 (s, 9H). Step G. 3-((1r,4r)-4-(3-(6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-di hydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)cyclohexyl)propanoic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1r,4r)-4-(3-ethoxy-3-oxopropyl)cyclohexyl)oxy)-2 -methylphenyl)picolinate (440 mg, 567.77 mmol, 1 equiv.) and LiOH monohydrate (71.47 mg, 1.70 mmol, 3 equiv.) in THF (1.2 mL) and water (0.4 mL) was stirred at 25 °C for 1.5 h. The mixture was concentrated and redissolved in water (20 mL), and an aqueous solution of 1M HCl was added to adjust the pH to 2. The resulting solids were filtered and dried under reduced pressure to afford 3-((1r,4r)- 4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoqui nolin-2(1H)-yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)cyclohexyl)prop anoic acid (380 mg, 508.76 mmol, 89.61 % yield). Step H. tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1r,4r)-4-(3-((3-(2,6-dioxopiperidin-3-yl)-1-meth yl-1H-indazol-6-yl)amino)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinate A mixture of 3-((1r,4r)-4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-di hydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)cyclohexyl)propanoic acid (120 mg, 160.66 µmol, 1 equiv.), 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (45.64 mg, 176.73 µmol, 1.1 equiv.), HATU (73.31 mg, 192.79 µmol, 1.2 equiv.), and N,N- diisopropylethylamine (62.29 mg, 481.98 µmol, 83.95 uL, 3 equiv.) in DMF (0.5 mL) was stirred at 25 °C for 12 hours. The reaction mixture was diluted with water (1 mL) and extracted with ethyl acetate (1 mL × 3). The combined organic layers were washed with water, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give crude tert- butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)- 4-(3-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl) amino)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (120 mg) as a white solid which was carried forward without further purification. Step I. 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1r,4r)-4-(3-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-ind azol-6-yl)amino)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1r,4r)-4-(3-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-ind azol-6-yl)amino)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (120 mg, 121.56 µmol, 1 equiv.) in trifluoroacetic acid (0.5 mL) and DCM (0.5 mL) was stirred at 40 °C for 10 hours. The reaction mixture was concentrated under reduced pressure. The resulting crude material was purified by prep-HPLC (column: Phenomenex luna C18150 × 25 mm × 10 um; mobile phase: [water (1% formic acid) - acetonitrile]; B%: 56%-86%, 10 min) to give 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r) -4-(3-((3-(2,6-dioxopiperidin-3- yl)-1-methyl-1H-indazol-6-yl)amino)-3-oxopropyl)cyclohexyl)o xy)-2- methylphenyl)picolinic acid (35.76 mg, 37.68 µmol, 31% yield, 98.1% purity) as a white solid. MS (ESI) m/z: 932.3 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ 12.94 - 12.74 (m, 1H), 10.89 - 10.80 (m, 1H), 10.05 (s, 1H), 8.09 - 8.00 (m, 2H), 7.82 - 7.75 (m, 1H), 7.65 - 7.59 (m, 2H), 7.53 - 7.31 (m, 6H), 7.12 - 7.05 (m, 2H), 7.00 - 6.90 (m, 2H), 6.61 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.34 - 4.28 (m, 1H), 4.26 - 4.17 (m, 1H), 3.91 (d, J = 2.0 Hz, 5H), 3.03 (s, 2H), 2.67 - 2.61 (m, 2H), 2.42 - 2.30 (m, 5H), 2.20 - 2.07 (m, 4H), 1.89 - 1.80 (m, 5H), 1.57 (d, J = 6.0 Hz, 2H), 1.39 - 1.32 (m, 2H). Example 4. Preparation of Compound 130 6-[8-(1,3-Benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[4-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]piperazin-1-yl]butyl]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A.4-(3-bromo-2-methyl-phenyl)but-3-yn-1-ol A mixture of 1-bromo-3-iodo-2-methyl-benzene (5 g, 16.84 mmol, 1 equiv.), but-3-yn-1-ol (1.77 g, 25.26 mmol, 1.91 mL, 1.5 equiv.), Pd(PPh3)4 (778.34 mg, 673.56 µmol, 0.04 equiv.), CuI (128.28 mg, 673.56 µmol, 0.04 equiv.) and in TEA (40 mL) was degassed and purged with nitrogen three times, and then the mixture was stirred at 90 °C for 16 hours under a nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure to remove solvent to give a residue which was purified by silica gel chromatography (petroleum ether/ethyl acetate=5/1 to 4/1) to afford 4-(3-bromo-2-methyl-phenyl)but-3-yn-1-ol (3.6 g, 15.06 mmol, 89.4% yield). 1 H NMR (400 MHz, CDCl 3 ) δ = 7.39 (d, J = 8.0 Hz, 1H), 7.26 (d, J = 7.6 Hz, 1H), 6.88 (t, J = 7.6 Hz, 1H), 3.76 (t, J = 6.4 Hz, 2H), 2.66 (t, J = 6.4 Hz, 2H), 2.45 (s, 3H), 1.79 (s, 1H). Step B.4-(3-Bromo-2-methyl-phenyl)butan-1-ol A mixture of 4-(3-bromo-2-methyl-phenyl)but-3-yn-1-ol (3.5 g, 14.64 mmol, 1 equiv.), PtO 2 (332.39 mg, 1.46 mmol, 0.1 equiv.) in EtOH (5 mL) was degassed and purged with hydrogen gas three times, and then the mixture was stirred at 30 °C for 5 hours under a hydrogen atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give 4-(3-bromo-2-methyl-phenyl)butan-1-ol that was used without further purification assuming theoretical yield (4 g, crude). 1 H NMR (400 MHz, CDCl 3 ) δ = 7.32 (d, J = 8.0 Hz, 1H), 6.99 (d, J = 7.6 Hz, 1H), 6.91 - 6.85 (m, 1H), 3.67 - 3.57 (m, 3H), 2.64 - 2.58 (m, 2H), 2.31 (s, 3H), 1.29 (s, 2H), 1.16 (t, J = 7.2 Hz, 2H). Step C.1-Bromo-3-(4-bromobutyl)-2-methyl-benzene To a solution of 4-(3-bromo-2-methyl-phenyl)butan-1-ol (2.0 g, 8.23 mmol, 1 equiv.) in DCM (10 mL) was added PPh 3 (2.37 g, 9.05 mmol, 1.1 equiv.) and CBr 4 (4.09 g, 12.34 mmol, 1.5 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to remove solvent to give a residue which was purified by silica gel chromatography (petroleum ether/ethyl acetate=20/1 to 10/1) to afford 1-bromo-3-(4- bromobutyl)-2-methyl-benzene (1.6 g, 5.23 mmol, 63.6% yield). 1 H NMR (400 MHz, CDCl3) δ = 7.34 (d, J = 8.0 Hz, 1H), 6.99 (d, J = 7.6 Hz, 1H), 6.92 - 6.86 (m, 1H), 3.36 (t, J = 6.8 Hz, 2H), 2.64 - 2.58 (m, 2H), 2.32 (s, 3H), 1.90 - 1.81 (m, 2H), 1.69 - 1.59 (m, 2H). Step D. Ethyl 2-[4-[4-(3-bromo-2-methyl-phenyl)butyl]piperazin-1-yl]acetat e To a solution of 1-bromo-3-(4-bromobutyl)-2-methyl-benzene (1.6 g, 5.23 mmol, 1 equiv.) in MeCN (20 mL) was added ethyl 2-piperazin-1-ylacetate (900.41 mg, 5.23 mmol, 1 equiv.) and K2CO3 (2.17 g, 15.68 mmol, 3 equiv.). The mixture was stirred at 60 °C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to remove solvent to give a residue which was purified by silica gel chromatography (DCM: MeOH = 1/0 to 10:1) to afford ethyl 2-[4-[4-(3-bromo-2-methyl-phenyl)butyl]piperazin-1-yl]acetat e (1.6 g, 4.03 mmol, 77.0% yield). MS (ESI) m/z: 398.9 [M+H] + . Step E. tert-Butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[4-[4-(2-ethoxy-2-oxo-ethyl)piperazin-1-yl]butyl]-2 -methyl-phenyl]pyridine-2- carboxylate A mixture of ethyl 2-[4-[4-(3-bromo-2-methyl-phenyl)butyl]piperazin-1-yl]acetat e (300 mg, 0.755 mmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (462.47 mg, 0.755 mmol, 1 equiv.), K 2 CO 3 (313.04 mg, 2.27 mmol, 3 equiv.), and Ad 2 n BuP Pd G3 (cataCXium® A Pd G3) (54.98 mg, 0.076 mmol, 0.1 equiv.) in dioxane (5 mL) and H2O (2 mL) was degassed and purged with nitrogen gas three times. The mixture was stirred at 100 °C for 2 hours under a nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure to provide a residue which was purified by silica gel chromatography (DCM: MeOH = 1/0 to 10:1) to afford tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[4-[4-(2-ethoxy-2-oxo-ethyl)piperazin -1-yl]butyl]-2-methyl- phenyl]pyridine-2-carboxylate (280 mg, 0.035 mmol, 46.2% yield) as a yellow oil. MS (ESI) m/z: 803.5 [M+H] + . Step F. 2-[4-[4-[3-[6-[8-(1,3-Benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H-isoquinolin-2- yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl-phenyl]butyl]p iperazin-1-yl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[4-[4-(2-ethoxy-2-oxo-ethyl)piperazin-1-yl]butyl] -2-methyl-phenyl]pyridine-2- carboxylate (250 mg, 0.31 mmol, 1 equiv.) in THF (3 mL) and H 2 O (1 mL) was added LiOH•H2O (78.39 mg, 1.87 mmol, 6.0 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue that was diluted with water (10 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were concentrated under reduced pressure to afford 2-[4-[4-[3-[6-[8-(1,3-benzothiazol- 2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butox ycarbonyl-3-pyridyl]-2- methyl-phenyl]butyl]piperazin-1-yl]acetic acid which was used without further purification (200 mg, 0.26 mmol, 82.9% yield). MS (ESI) m/z: 775.5 [M+H] + . Step G. tert-Butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[4-[4-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indaz ol-6-yl]amino]-2-oxo- ethyl]piperazin-1-yl]butyl]-2-methyl-phenyl]pyridine-2-carbo xylate To a solution of 2-[4-[4-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenyl]butyl]piperazin-1- yl]acetic acid (78 mg, 0.10 mmol, 1 equiv.) in DMF (2 mL) was added HATU (38.27 mg, 0.10 mmol, 1 equiv.), 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (26.00 mg, 0.10 mmol, 1 equiv.), and DIEA (39 mg, 0.30 mmol, 1 equiv.). The mixture was stirred at 25 °C for 2 hours, then diluted with water (10 mL), filtered, and concentrated under reduced pressure to afford tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- [3-[4-[4-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-y l]amino]-2-oxo-ethyl]piperazin- 1-yl]butyl]-2-methyl-phenyl]pyridine-2-carboxylate that was used without further purification (100 mg, 0.098 mmol, 97.9% yield). MS (ESI) m/z: 507.5 [M+2H]/2 + . Step H. 6-[8-(1,3-Benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4- [4-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amin o]-2-oxo-ethyl]piperazin-1- yl]butyl]-2-methyl-phenyl]pyridine-2-carboxylic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[4-[4-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indaz ol-6-yl]amino]-2-oxo- ethyl]piperazin-1-yl]butyl]-2-methyl-phenyl]pyridine-2-carbo xylate (70 mg, 0.069 mmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL) was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by preparative RP HPLC (water/MeCN, both with 0.1% formic acid modifier) to afford 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[4-[4-[2-[[3-(2,6-dioxo- 3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]pipera zin-1-yl]butyl]-2-methyl- phenyl]pyridine-2-carboxylic acid (12.87 mg, 0.013 mmol, 19.3% yield, 99.4% purity). MS (ESI) m/z: 959.5 [M+H] + ; 1 H NMR (400 MHz, DMSO-d6) δ = 10.87 (s, 1H), 9.85 (s, 1H), 8.24 (s, 1H), 8.07 - 7.97 (m, 2H), 7.78 (d, J = 8.4 Hz, 1H), 7.63 ( d, J = 9.2 Hz, 2H), 7.46 - 7.41 (m, 2H), 7.39 - 7.33 (m, 2H), 7.20 ( d, J = 8.8 Hz, 1H), 7.10 - 7.02 (m, 2H), 6.90 ( t, J = 9.2 Hz, 2H), 4.99 ( s, 2H), 4.33 (dd, J = 4.8, 9.4 Hz, 1H), 3.93 (s, 5H), 3.15 ( s, 3H), 3.02 ( d, J = 5.6 Hz, 2H), 2.69 - 2.58 (m, 6H), 2.41 - 2.39 (m, 2H), 2.34 ( s, 4H), 2.24 ( s, 1H), 2.02 (s, 3H), 1.56 - 1.46 (m, 4H). Example 5. Preparation of Compound 133 6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4-(3-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)oxy)propy l)piperazin-1-yl)propoxy)-2- methylphenyl)picolinic acid Step A. (3-(3-Bromo-2-methylphenoxy)propoxy)(tert-butyl)dimethylsila ne 3-Bromo-2-methylphenol (2.00 g, 1 equiv., 10.7 mmol), (3-bromopropoxy)(tert- butyl)dimethylsilane (3.39 g, 1.25 equiv., 13.4 mmol), and potassium carbonate (2.22 g, 1.50 equiv., 16.0 mmol) were combined in DMF and heated overnight at 50 °C. Diluted the reaction mixture with ethyl acetate (100 mL) and brine (25 mL). Separated layers, dried organic layer over anhydrous magnesium sulfate, filtered, and concentrated to an oil that was purified by silica gel chromatography (heptane/ethyl acetate gradient). Combined and concentrated product containing fractions to afford (3-(3-bromo-2-methylphenoxy)propoxy)(tert- butyl)dimethylsilane (3.49 g, 9.71 mmol, 90.8 % yield) returned. MS (ESI) m/z: 359.2 [M+H] + . Step B. tert-Butyldimethyl(3-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2- yl)phenoxy)propoxy)silane Combined (3-(3-bromo-2-methylphenoxy)propoxy)(tert-butyl)dimethylsila ne (1.00 g, 1 equiv., 2.78 mmol), bis(pinacolato)diboron (1.20 g, 1.7 equiv., 4.73 mmol), and PdCl2(dppf)- CH2Cl2 adduct (227 mg, 0.1 equiv., 278 µmol) in 1,4-dioxane (15 mL). Added potassium acetate (819 mg, 3 equiv., 8.35 mmol) and let mixture stir at 90 °C overnight. Diluted reaction mixture with 125 mL of ethyl acetate, 25 mL of water, and 25 mL of brine. Separated layers and washed organic layer with an additional 50 mL of brine, dried over anhydrous magnesium sulfate, filtered, and concentrated to dryness under reduced pressure. Purified residue by silica gel chromatography (heptane/ethyl acetate gradient). Combined and concentrated product containing fractions to afford tert-butyldimethyl(3-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)phenoxy)propoxy)silane (0.862 g, 2.12 mmol, 76.2 % yield). MS (ESI) m/z: 407. [M+H] + . Step C. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-((tert-butyldimethylsilyl)oxy)propoxy)-2-methylp henyl)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- bromopicolinate (662 mg, 1 equiv., 1.17 mmol) and tert-butyldimethyl(3-(2-methyl-3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)propoxy)silane (571 mg, 1.2 equiv., 1.40 mmol) from were dissolved in 1,4-dioxane (8.0 mL). Added potassium phosphate (745 mg, 2.34 mL, 1.5 molar, 3 equiv., 3.51 mmol) and CataCxium Pd G3 (42.6 mg, 0.05 equiv., 58.5 µmol). Let mixture stir at 100 °C for 40 minutes under microwave heating. Diluted reaction mixture with 125 mL of ethyl acetate, 25 mL of water, and 25 mL of brine. Separated layers and washed organic layer with an additional 50 mL of brine, dried over anhydrous magnesium sulfate, filtered, and concentrated to dryness under reduced pressure. Purified residue by silica gel chromatography (heptane/ethyl acetate gradient). Combined and concentrated product containing fractions to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((tert-butyldimethylsil yl)oxy)propoxy)-2- methylphenyl)picolinate(530 mg, 693 µmol, 59.2 % yield) returned. MS (ESI) m/z: 765.3 [M+H] + . Step D. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-hydroxypropoxy)-2-methylphenyl)picolinate TBAF (217 mg, 831 µL, 1.0 molar, 1.2 equiv., 831 µmol), as a 1N solution in THF, and tert- butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((tert- butyldimethylsilyl)oxy)propoxy)-2-methylphenyl)picolinate (530 mg, 1 equiv., 693 µmol) were combined in THF (10.0 mL) and mixed at ambient temperature overnight. Added an additional portion of TBAF (217 mg, 831 µL, 1.0 molar, 1.2 equiv., 831 µmol) and let mix again overnight at ambient temperature. Diluted reaction mixture with 125 mL of ethyl acetate, 25 mL of water, and 25 mL of brine. Separated layers and washed organic layer with an additional 50 mL of brine, dried over anhydrous magnesium sulfate, filtered, and concentrated to dryness under reduced pressure. Purified residue by silica gel chromatography (heptane/ethyl acetate gradient). Combined and concentrated product containing fractions to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- (3-(3-hydroxypropoxy)-2-methylphenyl)picolinate (322 mg, 495 µmol, 71.4 % yield). MS (ESI) m/z: 651.2 [M+H] + . Step E. tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-chloropropoxy)-2-methylphenyl)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3- hydroxypropoxy)-2-methylphenyl)picolinate (322 mg, 1 equiv., 495 µmol) was dissolved in DCM (10 mL). Added thionyl chloride (118 mg, 72.2 µL, 2 equiv., 990 µmol) and let reaction mix overnight at ambient temperature. Diluted reaction mixture with additional DCM and a saturated aqueous solution of sodium bicarbonate. Separated layers, dried organic layer over anhydrous magnesium sulfate, filtered, and concentrated to dryness under vacuum. Purified residue by silica gel chromatography (heptane/ethyl acetate gradient). Combined and concentrated product containing fractions to afford tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-chloro propoxy)-2- methylphenyl)picolinate (163 mg, 244 µmol, 49.2 % yield). MS (ESI) m/z: 669.1 [M+H] + . Step F. tert-Butyl 4-(3-((3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazo l-6- yl)oxy)propyl)piperazine-1-carboxylate tert-Butyl 4-(3-hydroxypropyl)piperazine-1-carboxylate (195 mg, 2 equiv., 799 µmol) and 3- (2,6-bis(benzyloxy)pyridin-3-yl)-6-bromo-1-methyl-1H-indazol e (200 mg, 1 equiv., 400 µmol) in dioxane (5 mL). Added tBuBrettPhos Pd G3 (17.1 mg, 0.05 equiv., 20.0 µmol) followed by potassium tert-butoxide (53.8 mg, 480 µL, 1.0 molar, 1.2 equiv., 480 µmol) and let mix at ambient temperature for 48 h. Diluted reaction mixture with 125 mL of ethyl acetate, 25 mL of water, and 25 mL of brine. Separated layers and washed organic layer with an additional 50 mL of brine, dried over anhydrous magnesium sulfate, filtered, and concentrated to dryness under reduced pressure. Purified residue by silica gel chromatography (heptane/ethyl acetate containing 10% of a 7N solution of NH3 in MeOH). Concentrated product containing fractions to afford tert-butyl 4-(3-((3-(2,6-bis(benzyloxy)pyridin-3-yl)-1- methyl-1H-indazol-6-yl)oxy)propyl)piperazine-1-carboxylate (170 mg, 256 µmol, 64.1 % yield). MS (ESI) m/z: 664.3 [M+H] + . Step G. tert-Butyl 4-(3-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)oxy)propyl)piperazine-1-carboxylate Dissolved tert-butyl 4-(3-((3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazo l-6- yl)oxy)propyl)piperazine-1-carboxylate (170 mg, 1 equiv., 256 µmol) in Ethyl acetate (10 mL). Added palladium on carbon (545 mg, 5% wt, 1 equiv., 256 µmol) and degassed reaction under vacuum. Let mix at ambient temperature under an hydrogen atmosphere for 48 h. Filtered reaction through a pad of Celite, rinsed Celite with ethyl acetate, and concentrated reaction to afford tert-butyl 4-(3-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)oxy)propyl)piperazine-1-carboxylate (79.6 mg, 164 µmol, 64.0 % yield). MS (ESI) m/z: 486.3 [M+H] + . Step H.3-(1-Methyl-6-(3-(piperazin-1-yl)propoxy)-1H-indazol-3-yl) piperidine-2,6-dione, 2HCl Dissolved tert-butyl 4-(3-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)oxy)propyl)piperazine-1-carboxylate (79.6 mg, 1 equiv., 164 µmol) in DCM (10 mL). Added HCl in dioxane (239 mg, 1.64 mL, 4 molar, 40.0 equiv., 6.56 mmol) and let mix under ambient temperature overnight. Concentrated material to afford 3-(1-methyl-6-(3-(piperazin- 1-yl)propoxy)-1H-indazol-3-yl)piperidine-2,6-dione, 2HCl (85 mg, 0.19 mmol). MS (ESI) m/z: 386.3 [M+H] + . Step I. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)- 3-(3-(3-(4-(3-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-inda zol-6- yl)oxy)propyl)piperazin-1-yl)propoxy)-2-methylphenyl)picolin ate 3-(1-Methyl-6-(3-(piperazin-1-yl)propoxy)-1H-indazol-3-yl)pi peridine-2,6-dione (80 mg, 2.8 equiv., 0.21 mmol) and tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-chloropropoxy)-2-methyl phenyl)picolinate (50 mg, 1 equiv., 75 µmol) were dissolved in DMF (0.5 mL). Added DIEA (48 mg, 65 µL, 5 equiv., 0.37 mmol). Let mix at 50°C overnight. Added an additional amount of DIEA (same amount as originally used). Let continue to mix overnight at 60 °C. Purified reaction mixture by RP HPLC (water/MeCN, both with 0.1% formic acid modifier) to afford tert-butyl 6-(8-(benzo[d]thiazol- 2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(4-( 3-((3-(2,6-dioxopiperidin-3-yl)- 1-methyl-1H-indazol-6-yl)oxy)propyl)piperazin-1-yl)propoxy)- 2-methylphenyl)picolinate (12 mg, 12 µmol, 16 % yield). MS (ESI) m/z: 1018.2 [M+H] + . Step J. 6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3- (4-(3-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl )oxy)propyl)piperazin-1- yl)propoxy)-2-methylphenyl)picolinic acid tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4- (3-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)ox y)propyl)piperazin-1- yl)propoxy)-2-methylphenyl)picolinate (12 mg, 1 equiv., 12 µmol) was dissolved in a mixture of DCM (30 mL) and HCl in dioxane (0.21 g, 1.5 mL, 4.0 molar, 500 equiv., 5.9 mmol). Let reaction stir at ambient temperature for 72 h. The reaction mixture was concentrated under reduced pressure to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4-(3-((3-(2,6-dioxopiperidin-3-yl)-1-meth yl-1H-indazol-6- yl)oxy)propyl)piperazin-1-yl)propoxy)-2-methylphenyl)picolin ic acid (16.5 mg, 13.9 µmol). MS (ESI) m/z: 962.2 [M+H] + . Example 6. Preparation of Compound 138a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1-yl)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1r,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6- yl)piperazin-1-yl)-3-oxopropyl)cyclohexyl)oxy)-2-methylpheny l)picolinate To a solution of 3-((1r,4r)-4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)cyclohexyl)propanoic acid (50 mg, 66.94 µmol, 1 equiv.), 3-(1-methyl-6- (piperazin-1-yl)-1H-indazol-3-yl)piperidine-2,6-dione (26.30 mg, 80.34 µmol, 1.2 equiv.) and EDCI (16.04 mg, 83.68 µmol, 1.2 equiv.) in pyridine (2 mL) was stirred at 25 °C for 3 hours. Then the mixture was diluted with water (2 mL) and extracted with ethyl acetate (1 mL × 3). The combined organic layers were filtered and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1r,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-6-yl)piperazin-1-yl)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (60 mg, crude) as a yellow oil which was carried forward without further purification. MS (ESI) m/z: 1056.47 [M+H] + . Step B. 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1r,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-6-yl)piperazin-1-yl)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1r,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-6-yl)piperazin-1-yl)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (50 mg, 47.34 µmol, 1 equiv.) in trifluoroacetic acid (0.8 mL) and dichloromethane (0.8 mL) was stirred at 40 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex luna C18150 × 25 mm × 10 µmol; mobile phase: [water (1% formic acid) - acetonitrile]; B%: 55% - 85%, 10 min) to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1r,4r)-4-(3-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin -1-yl)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (20.2 mg, 19.3 µmol, 40.7% yield, 95.2% purity) as a white solid. MS (ESI) m/z: 1000.4 [M+H] + ; 1 H NMR (400 MHz, DMSO-d6) δ 12.86 - 12.84 (m, 2H), 10.85 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.54 - 7.44 (m, 4H), 7.39 - 7.32 (m, 2H), 7.09 - 7.05 (m, 1H), 6.97 - 6.89 (m, 4H), 6.61 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.28 - 4.18 (m, 2H), 3.93 - 3.90 (m, 5H), 3.65 - 3.60 (m, 4H), 3.24 - 3.18 (m, 4H), 3.04 - 2.99 (m, 2H), 2.67 - 2.61(m, 2H), 2.39 (t, J = 7.6 Hz, 2H), 2.33 - 2.28 (m, 1H), 2.10 - 2.07 (m, 2H), 1.87 - 1.80 (m, 5H), 1.49 - 1.44 (m, 2H), 1.35 - 1.31 (m, 4H), 1.13 - 1.04 (m, 2H). Example 7. Preparation of Compound 142 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin -1-yl)-2-oxoethyl)piperidin-4- yl)propoxy)-2-methylphenyl)picolinic acid Step A. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-(1-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1 H-indazol-6-yl)piperazin-1- yl)-2-oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl)picoli nate To a solution of 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]propyl]-1- piperidyl]acetic acid (70 mg, 90.21 µmol, 1 equiv.) in DMF (2 mL) was added HATU (34.30 mg, 90.21 µmol, 1 equiv.), 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (29.53 mg, 90.21 µmol, 1 equiv.), and DIEA (11.66 mg, 90.21 µmol, 15.71 uL, 1 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (10 × 3 mL). The combined organic layers were filtered and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-(4-(3-(2,6-di oxopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)-2-oxoethyl)piperidin-4-yl)propo xy)-2-methylphenyl)picolinate (75 mg, 69.10 µmol, 76% yield) as a yellow oil, which was carried forward without further purification. MS (ESI) m/z 543.4 [M/2+H] + . Step B. 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3- (1-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)piperazin-1-yl)-2- oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)piperazin- 1-yl)-2-oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl)pico linate (70 mg, 64.50 µmol, 1 equiv.) in trifluoroacetic acid (1 mL) and dichloromethane (2 mL). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove solvent to give the crude product. The crude product was purified by reverse-phase HPLC(column: Phenomenex luna C18150 × 25 mm × 10 um; mobile phase: [water (1% formic acid) - acetonitrile]; B%: 30%-60%, 10 min) to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-(4-(3-(2,6-dioxop iperidin-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)-2-oxoethyl)piperidin-4-yl)propo xy)-2-methylphenyl)picolinic acid (25.64 mg, 23.93 µmol, 37% yield, 96% purity) as a white solid. MS (ESI) m/z: (1029.7 [M+H] + ; 1 H NMR (400 MHz, CDCl3) δ 7.82 - 7.72 (m, 2H), 7.54 ( d, J = 7.6 Hz, 1H), 7.45 (dd, J = 2.8, 8.8 Hz, 1H), 7.37 - 7.29 (m, 3H), 7.27 - 7.21 (m, 2H), 7.09 - 7.02 (m, 1H), 6.94 - 6.88 (m, 1H), 6.82 - 6.77 (m, 1H), 6.75 - 6.70 (m, 1H), 6.64 - 6.59 (m, 1H), 6.54 (s, 1H), 5.02 ( s, 1H), 4.18 (t, J = 6.0 Hz, 1H), 3.97 - 3.84 (m, 4H), 3.83 (d, J = 2.0 Hz, 3H), 3.78 - 3.74 (m, 2H), 3.14 ( d, J = 7.6 Hz, 4H), 3.03 ( d, J = 4.0 Hz, 2H), 2.97 - 2.83 (m, 2H), 2.80 ( d, J = 100 Hz, 2H), 2.67 - 2.49 (m, 2H), 2.47 - 2.35 (m, 2H), 2.30 ( dd, J = 4.4, 8.8 Hz, 2H), 2.06 - 1.98 (m, 4H), 1.83 (s, 3H), 1.73 - 1.68 (m, 2H), 1.62 ( d, J = 12.4 Hz, 2H), 1.36 - 1.31 (m, 2H), 1.18 (s, 2H). Example 8. Preparation of Compound 148a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6- yl)piperazin-1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl)pico linate A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[2-methyl-3-[4-(3-oxopropyl)cyclohexoxy]phenyl]pyridin e-2-carboxylate (80 mg, 109.45 µmol, 1 equiv), 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (43.00 mg, 131.34 µmol, 1.2 equiv.), 4A molecular sieves (109.45 µmol, 1 equiv.) and NaBH(OAc) 3 (69.59 mg, 328.35 µmol, 3 equiv.) in DCM (6 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 1.5 hour under nitrogen atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1s,4r)-4-(3-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin -1-yl)propyl)cyclohexyl)oxy)- 2-methylphenyl)picolinate (100 mg, 95.94 µmol, 87% yield) as a white solid. MS (ESI) m/z: 549.0 [M/2+H] + . Step B. 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-6-yl)piperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)piperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (80 mg, 76.75 µmol, 1 equiv) in dichloromethane (1 mL) and trifluoroacetic acid (1 mL) was stirred at 40 °C. After 3 h the reaction solution was concentrated and purified by prep-HPLC (column: Phenomenex luna C18150 × 25mm × 10um; mobile phase: [water (1% formic acid)-acetonitrile]; B%: 30% - 60%, 0min) to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- (3-(((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1 H-indazol-6-yl)piperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (70.56 mg, 66.37 µmol, 86 % yield, 92.7% purity) as a white solid. MS (ESI) m/z: 986.2 [M+H] + ; 1 H NMR (400 MHz, DMSO-d6) δ 12.95 - 12.79 (m, 1H), 10.86 (s, 1H), 8.14 (s, 1H), 8.04 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.53 - 7.50 (m, 1H), 7.48 - 7.45 (m, 3H), 7.40 - 7.34 (m, 2H), 7.10 - 7.06 (m, 1H), 6.98 - 6.89 (m, 5H), 6.62 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.29 - 4.21 (m, 2H), 3.94 - 3.88 (m, 6H), 3.03 (t, J = 5.2 Hz, 3H), 2.90 - 2.79 (m, 2H), 2.68 - 2.57 (m, 5H), 2.35 - 2.26 (m, 2H), 2.20 - 2.08 (m, 4H), 1.88 (s, 3H), 1.83 - 1.79 (m, 2H), 1.59 - 1.54 (m, 2H), 1.25 (d, J = 6.8 Hz, 6H), 1.12 – 1.04 (m, 2H). Example 9. Preparation of Compound 154a 6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(3-((3R,5S)-4- (2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl) piperazin-1-yl)-2-oxoethyl)-3,5- dimethylpiperazin-1-yl)propoxy)-2-methylphenyl)picolinic acid Step A. (3R,5S)-tert-Butyl 4-(2-methoxy-2-oxoethyl)-3,5-dimethylpiperazine-1- carboxylate A mixture of methyl 2-bromoacetate (9.35 g, 56.00 mmol, 6.19 mL, 1.5 equiv.), (3R, 5S)-tert- butyl 3,5-dimethylpiperazine-1-carboxylate (8 g, 37.33 mmol, 1 equiv.), and K2CO3 (15.48 g, 111.99 mmol, 3 equiv.) in MeCN (100 mL) was degassed and purged with N2 three times, and then stirred at 60 °C. After 3 hours, the reaction mixture was diluted with water (100 mL) and extracted with EtOAc (120 mL × 3). The combined organic layers were washed with brine (120 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The resulting crude material was purified by flash silica gel chromatography (0-25% ethyl acetate/petroleum ether) to give (3R,5S)-tert-butyl 4-(2-methoxy-2-oxoethyl)-3,5- dimethylpiperazine-1-carboxylate (10 g, 33.3 mmol, 89.2% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 4.10 - 4.05 (m, 2H), 3.67 (s, 2H), 3.50 (s, 2H), 2.78 - 2.69 (m, 2H), 2.49 - 2.32 (m, 2H), 1.39 (s, 9H), 1.20 - 1.16 (m, 3H), 0.97 (d, J = 6.4 Hz, 6H). Step B.2-((2R,6S)-2,6-Dimethylpiperazin-1-yl)acetate hydrochloride A mixture of (3R,5S)-tert-butyl 4-(2-methoxy-2-oxoethyl)-3,5-dimethylpiperazine-1- carboxylate (10 g, 33.29 mmol, 1 equiv.) and 4 M HCl in dioxane (83.22 mL, 10 equiv.) in dichloromethane (80 mL) was stirred at 25 °C for 16 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give methyl 2-((2R,6S)-2,6- dimethylpiperazin-1-yl)acetate hydrochloride (12.25 g, crude) as a yellow solid which was carried forward without further purification. 1 H NMR (400 MHz,CD3OD) δ = 4.44 (s, 2H), 4.36 (q, J = 7.2 Hz, 2H), 4.29 - 4.21 (m, 2H), 3.74 - 3.67 (m, 2H), 3.58 - 3.50 (m, 2H), 1.49 (d, J = 6.4 Hz, 6H), 1.35 (t, J = 7.2 Hz, 3H). Step C.1-Bromo-4-(3-bromopropoxy)-2-methylbenzene A mixture of 4-bromo-3-methylphenol (3 g, 16.04 mmol, 1 equiv.), 1,3-dibromopropane (16.19 g, 80.20 mmol, 8.18 mL, 5 equiv.) and K2CO3 (6.65 g, 48.12 mmol, 3 equiv.) in MeCN (30 mL) was stirred at 70 °C for 12 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure and purified by flash silica gel chromatography (0-5% ethyl acetate/petroleum ether) to give 1-bromo-4-(3-bromopropoxy)-2-methylbenzene (2.5 g, 8.12 mmol, 50.6% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.44 (d, J = 8.8 Hz, 1H), 6.98 (d, J = 2.8 Hz, 1H), 6.74 (dd, J = 8.8, 2.8 Hz, 1H), 4.08 - 4.02 (m, 2H), 3.65 (t, J = 6.4 Hz, 2H), 2.30 (s, 3H), 2.26 – 2.19 (m, 2H). Step D. Ethyl 2-((2R,6S)-4-(3-(4-bromo-3-methylphenoxy)propyl)-2,6- dimethylpiperazin-1-yl)acetate A mixture of 1-bromo-4-(3-bromopropoxy)-2-methylbenzene (2.34 g, 7.60 mmol, 1.2 equiv.), methyl 2-((2R,6S)-2,6-dimethylpiperazin-1-yl)acetate hydrochloride (1.5 g, 6.34 mmol, 1 equiv.) and N,N-diisopropylethylamine (4.09 g, 31.68 mmol, 5.52 mL, 5 equiv.) in dimethylformamide (40 mL) was stirred at 60 °C for 16 hours under N 2 atmosphere. The reaction mixture was diluted with water (100 mL) and extracted with dichloromethane (100 mL × 3). The combined organic layers were washed with brine (100 mL × 6), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The resulting crude material was purified by flash silica gel chromatography (0~7% dichloromethane / MeOH) to give ethyl 2-((2R,6S)-4-(3-(4-bromo-3-methylphenoxy)propyl)-2,6- dimethylpiperazin-1-yl)acetate (1.3 g, 3.04 mmol, 48% yield) as an orange oil. 1 H NMR (400 MHz, CD 3 OD) δ = 7.37 (d, J = 8.8 Hz, 1H), 6.86 (d, J = 3.0 Hz, 1H), 6.66 (dd, J = 8.8, 3.2 Hz, 1H), 4.16 (q, J = 7.2 Hz, 2H), 3.98 (t, J = 6.0 Hz, 2H), 3.55 (s, 2H), 3.04 - 2.98 (m, 2H), 2.86 - 2.84 (m, 1H), 2.82 (s, 1H), 2.54 - 2.46 (m, 2H), 2.33 (s, 3H), 2.00 - 1.92 (m, 2H), 1.92 - 1.85 (m, 2H), 1.27 (t, J = 7.2 Hz, 3H), 1.08 (d, J = 6.4 Hz, 6H). Step E. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4-(3-((3R,5S)-4-(2-ethoxy-2-oxoethyl)-3,5-dimethylpip erazin-1-yl)propoxy)-2- methylphenyl)picolinate Ethyl 2-((2R,6S)-4-(3-(4-bromo-3-methylphenoxy)propyl)-2,6-dimethy lpiperazin-1-yl)acetate (400 mg, 935.95 µmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)picolinate (687.97 mg, 1.12 mmol, 1.2 equiv.), cataCXium® A Pd G3 (68.16 mg, 93.59 µmol, 0.1 equiv.) and KF (1.5 M, 1.87 mL, 3 equiv.) were taken up into a microwave tube in 1,4-dioxane (11 mL). The sealed tube was heated in a microwave reactor at 100 °C for 60 minutes. The reaction mixture was concentrated under reduced pressure and purified by reverse-phase HPLC to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(3- ((3R,5S)-4-(2-ethoxy-2-oxoethyl)-3,5-dimethylpiperazin-1-yl) propoxy)-2- methylphenyl)picolinate (410 mg, 477.46 umol, 51% yield, 97% purity) as a yellow solid. MS (ESI) m/z: 833.4 [M+H] + ; 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.12 - 12.55 (m, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.77 (d, J = 8.0 Hz, 1H), 7.60 (d, J = 7.6 Hz, 1H), 7.48 - 7.41 (m, 3H), 7.39 - 7.31 (m, 2H), 6.92 (d, J = 8.8 Hz, 1H), 6.85 (d, J = 8.4 Hz, 1H), 6.81 (s, 1H), 6.75 - 6.69 (m, 1H), 4.96 (s, 2H), 4.06 (q, J = 7.2 Hz, 2H), 3.98 (t, J = 6.0 Hz, 2H), 3.86 (t, J = 6.0 Hz, 2H), 3.45 (s, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.87 - 2.80 (m, 2H), 2.72 (d, J = 10.4 Hz, 2H), 2.34 (t, J = 6.8 Hz, 2H), 2.00 (s, 3H), 1.87 -1.78 (m, 2H), 1.66 (t, J = 10.4 Hz, 2H), 1.17 (t, J = 7.2 Hz, 3H), 1.03 (s, 9H), 0.95 (d, J = 6.4 Hz, 6H). Step F.2-((2R,6S)-4-(3-(4-(6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3 ,4-dihydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-3-methylpheno xy)propyl)-2,6- dimethylpiperazin-1-yl)acetic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4-(3-((3R,5S)-4-(2-ethoxy-2-oxoethyl)-3,5-dimethylpip erazin-1-yl)propoxy)-2- methylphenyl)picolinate (400 mg, 480.17 umol, 1 equiv.) and LiOH·H2O (100.75 mg, 2.40 mmol, 5 equiv.) in THF (4 mL) and H 2 O (1 mL) was stirred at 40 °C under N 2 atmosphere. After 7 hours, the reaction mixture was concentrated under reduced pressure to provide 2- ((2R,6S)-4-(3-(4-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-d ihydroisoquinolin-2(1H)-yl)-2- (tert-butoxycarbonyl)pyridin-3-yl)-3-methylphenoxy)propyl)-2 ,6-dimethylpiperazin-1- yl)acetic acid (400 mg, crude) as a yellow solid, which was carried forward without further purification. MS (ESI) m/z: 805.7 [M+H] + . Step G. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4-(3-((3R,5S)-4-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6- yl)piperazin-1-yl)-2-oxoethyl)-3,5-dimethylpiperazin-1-yl)pr opoxy)-2- methylphenyl)picolinate 2-((2R,6S)-4-(3-(4-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4 -dihydroisoquinolin-2(1H)-yl)- 2-(tert-butoxycarbonyl)pyridin-3-yl)-3-methylphenoxy)propyl) -2,6-dimethylpiperazin-1- yl)acetic acid (100 mg, 124.22 µmol, 1 equiv.), 3-(1-methyl-6-(piperazin-1-yl)-1H-indazol-3- yl)piperidine-2,6-dione hydrochloride (54.24 mg, 149.07 µmol, 1.2 equiv.), and N,N- diisopropyl-N-diethylamine (48.17 mg, 372.67 µmol, 64.91 µL, 3 equiv.) in DMF (1.5 mL) was stirred at 25 °C. After 5 minutes, HATU (56.68 mg, 149.07 umol, 1.2 equiv.) was added, and the mixture was stirred at 25 °C for 1 hour under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(4-(3-((3R,5S)-4-(2-(4-(3 -(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-2-oxoethyl)-3,5-dimet hylpiperazin-1-yl)propoxy)-2- methylphenyl)picolinate (130 mg, 116.7 µmol, 93.9% yield) as a brown solid. MS (ESI) m/z: 1114.5 [M+H] + . Step H. 6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(3- ((3R,5S)-4-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-in dazol-6-yl)piperazin-1-yl)-2- oxoethyl)-3,5-dimethylpiperazin-1-yl)propoxy)-2-methylphenyl )picolinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4-(3-((3R,5S)-4-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6- yl)piperazin-1-yl)-2-oxoethyl)-3,5-dimethylpiperazin-1-yl)pr opoxy)-2- methylphenyl)picolinate (120 mg, 107.69 µmol, 1 equiv.) in trifluoroacetic acid (0.6 mL) and dichloromethane (0.6 mL) was stirred at 25 °C. After 16 h, the reaction mixture was concentrated under reduced pressure and purified by prep-HPLC (column: Phenomenex luna C18150 × 25mm × 10um; mobile phase: [water (1% formic acid)-acetonitrile]; B%: 25%- 55%,10 min) to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)- 3-(4-(3-((3R,5S)-4-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-meth yl-1H-indazol-6-yl)piperazin-1- yl)-2-oxoethyl)-3,5-dimethylpiperazin-1-yl)propoxy)-2-methyl phenyl)picolinic acid (80.4 mg, 73.8 µmol, 68.5% yield, 97.1% purity) as a white solid. MS (ESI) m/z: 1058.8 [M+H] + ; 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.38 - 12.38 (m, 1H), 10.85 (s, 1H), 8.14 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.52 (d, J = 8.8 Hz, 1H), 7.50 - 7.31 (m, 6H), 6.96 - 6.88 (m, 4H), 6.78 (s, 1H), 6.73 -6.68 (m, 1H), 4.97 (s, 2H), 4.29 - 4.24 (m, 1H), 3.98 (t, J = 6.0 Hz, 2H), 3.90 (s, 3H), 3.69 (s, 3H), 3.61 (s, 6H), 3.23 (s, 2H), 3.19 (s, 2H), 3.13 (d, J = 2.0 Hz, 2H), 3.02 (t, J = 5.2 Hz, 3H), 2.79 (d, J = 10.0 Hz, 2H), 2.65 - 2.59 (m, 2H), 2.45 (s, 2H), 2.34 - 2.26 (m, 1H), 2.20 - 2.13 (m, 1H), 2.03 (s, 3H), 1.90 - 1.85 (m, 2H), 0.97 (d, J = 6.0 Hz, 6H). Example 10. Preparation of Compound 153 6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(3-(4-(2-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin -1-yl)-2-oxoethyl)piperazin-1- yl)propoxy)-2-methylphenyl)picolinic acid Step A.1-Bromo-4-(3-bromopropoxy)-2-methylbenzene A mixture of 4-bromo-3-methylphenol (2 g, 10.69 mmol, 1 equiv.), 1,3-dibromopropane (10.79 g, 53.47 mmol, 5.45 mL, 5 equiv.), and K2CO3 (4.43 g, 32.08 mmol, 3 equiv.) in MeCN (20 mL) was stirred at 70 °C. After 12 h, the reaction mixture was filtered and concentrated under the reduced pressure. The resulting crude material was purified by flash silica gel chromatography (0-5% ethyl acetate/petroleum ether) to give 1-bromo-4-(3-bromopropoxy)- 2-methylbenzene (1.55 g, 5.03 mmol, 47.1% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.44 (d, J = 8.8 Hz, 1H), 6.97 (d, J = 2.8 Hz, 1H), 6.74 (dd, J = 3.2, 8.8 Hz, 1H), 4.05 (t, J = 6.0 Hz, 2H), 3.65 (t, J = 6.4 Hz, 2H), 2.30 (s, 3H), 2.22 (m, J = 6.4 Hz, 2H). Step B. Ethyl 2-(4-(3-(4-bromo-3-methylphenoxy)propyl)piperazin-1-yl)aceta te A mixture of 1-bromo-4-(3-bromopropoxy)-2-methylbenzene (1.5 g, 4.87 mmol, 1 equiv.), ethyl 2-(piperazin-1-yl)acetate (838.73 mg, 4.87 mmol, 1 equiv.), and K2CO3 (3.37 g, 24.35 mmol, 5 equiv.) in MeCN (15 mL) was stirred at 60 °C. After 14 h, the reaction mixture was filtered and concentrated under the reduced pressure. The resulting crude material was purified by silica gel chromatography (0-100% ethyl acetate/petroleum ether) to give ethyl 2-(4-(3-(4- bromo-3-methylphenoxy)propyl)piperazin-1-yl)acetate (1.44 g, 3.28 mmol, 67.4% yield, 91.0% purity) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.53 (d, J = 8.8 Hz, 1H), 7.05 (d, J = 2.8 Hz, 1H), 6.82 (dd, J = 3.2, 8.8 Hz, 1H), 4.19 (q, J = 7.2 Hz, 2H), 4.07 (t, J = 6.4 Hz, 2H), 3.29 (s, 2H), 2.62 (dd, J = 2.0, 3.6 Hz, 4H), 2.56 - 2.45 (m, 6H), 2.41 (s, 3H), 1.99 - 1.91 (m, 2H), 1.30 (t, J = 7.2 Hz, 3H). Step C. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4-(3-(4-(2-ethoxy-2-oxoethyl)piperazin-1-yl)propoxy)- 2-methylphenyl)picolinate A mixture of ethyl 2-(4-(3-(4-bromo-3-methylphenoxy)propyl)piperazin-1-yl)aceta te (400 mg, 1.00 mmol, 1 equiv.), tert-butyl 6-(8-((3H-indol-2-yl)carbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pic olinate (612.55 mg, 1.00 mmol, 1 equiv.), and K2CO3 (1.5 M, 1.00 mL, 1.5 equiv.) in dioxane (4 mL) was added cataCXium® A Pd G3 (145.65 mg, 200.00 µmol, 0.2 equiv.). The reaction mixture was purged with N2 three times and the reaction mixture was stirred at 80 °C. After 2 h, the reaction solution was partitioned between H 2 O (10 mL) and ethyl acetate (10 mL). The organic phase was separated, washed with aqueous NaCl solution (3 mL × 2), dried over with Na2SO4, filtered, and concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography (0-90% ethyl acetate/petroleum ether) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(4-(3-(4-(2-ethoxy-2- oxoethyl)piperazin-1-yl)propoxy)-2-methylphenyl)picolinate (692 mg, 733.27 µmol, 73.3% yield, 85.3% purity) as a yellow solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.01 - 12.64 (m, 1H), 8.02 (d, J = 7.6 Hz, 1H), 7.77 (d, J = 8.0 Hz, 1H), 7.60 (d, J = 7.6 Hz, 1H), 7.46 - 7.41 (m, 3H), 7.39 - 7.31 (m, 2H), 6.94 (d, J = 2.4 Hz, 1H), 6.86 (d, J = 8.4 Hz, 1H), 6.81 (d, J = 2.4 Hz, 1H), 6.73 (d, J = 2.0 Hz, 1H), 4.96 (s, 2H), 4.04 (s, 2H), 4.02 - 4.00 (m, 2H), 3.86 (t, J = 5.6 Hz, 2H), 3.17 (s, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.39 (d, J = 5.6 Hz, 8H), 2.33 - 2.25 (m, 3H), 2.00 (s, 2H), 1.83 (t, J = 6.8 Hz, 2H), 1.18 (d, J = 1.2 Hz, 3H), 1.03 (s, 9H). Step D. 2-(4-(3-(4-(6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-3-methylphenoxy)pro pyl)piperazin-1-yl)acetic acid tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(3-(4- (2-ethoxy-2-oxoethyl)piperazin-1-yl)propoxy)-2-methylphenyl) picolinate (380 mg, 472.05 µmol, 1 equiv.) and LiOH•H 2 O (1 M, 1.42 mL, 3 equiv.) in THF (4 mL) and H 2 O (1 mL) was stirred at 25 °C. After 13 h, the reaction solution was concentrated, diluted with water (4 mL), and the pH was adjusted to 3 by addition of 1 M HCl. The resulting slurry was filtered, and the filter cake was dried to give 2-(4-(3-(4-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-3- methylphenoxy)propyl)piperazin-1-yl)acetic acid (333 mg, crude) as a yellow solid which was carried forward without further purification. MS (ESI) m/z: 777.5 [M+H] + . Step E. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4-(3-(4-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1 H-indazol-6-yl)piperazin-1- yl)-2-oxoethyl)piperazin-1-yl)propoxy)-2-methylphenyl)picoli nate To a mixture of 2-(4-(3-(4-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-3-methylpheno xy)propyl)piperazin-1- yl)acetic acid (120 mg, 154.45 µmol, 1 equiv.), HATU (64.60 mg, 169.90 µmol, 1.1 equiv.), N,N-diisopropyl-N-ethylamine (59.9 mg, 463.4 µmol, 80.7 µL, 3 equiv.) in DMF (1.2 mL) was added 3-(1-methyl-6-(piperazin-1-yl)-1H-indazol-3-yl)piperidine-2, 6-dione (50.6 mg, 154.5 µmol, 1 equiv.). The reaction solution was stirred at 25 °C. After 2 h, the reaction solution was diluted with water (2 mL) and filtered. The filter cake was dissolved in dichloromethane and methanol and concentrated to provide tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(3-(4-(2-(4-(3-(2,6-dioxop iperidin-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)-2-oxoethyl)piperazin-1-yl)propo xy)-2-methylphenyl)picolinate (215 mg, crude) as a yellow solid, which was carried forward without further purification. MS (ESI) m/z: 543.9 [M/2+H] + . Step F. 6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(3- (4-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)piperazin-1-yl)-2- oxoethyl)piperazin-1-yl)propoxy)-2-methylphenyl)picolinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4-(3-(4-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1 H-indazol-6-yl)piperazin-1-yl)-2- oxoethyl)piperazin-1-yl)propoxy)-2-methylphenyl)picolinate (215 mg, 197.92 µmol, 1 equiv.) in trifluoroacetic acid (1 mL) and CH 2 Cl 2 (3 mL) was stirred at 25 °C. After 70 h, the reaction mixture was concentrated under reduced pressure and purified by prep-HPLC (column: Phenomenex luna C18150 × 25 mm × 10 um; mobile phase: [water (1% formic acid)-MeCN]; B%: 20%-50%, 0 min) to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(3-(4-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)piperazin- 1-yl)-2-oxoethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linic acid (22.5 mg, 21.3 µmol, 10.8% yield, 97.4% purity) as a white solid. MS (ESI) m/z: 1030.7 [M+H] + ; 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.85 (s, 1H), 8.14 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.52 (d, J = 8.8 Hz, 1H), 7.49 - 7.42 (m, 3H), 7.36 (q, J = 8.0 Hz, 2H), 6.97 - 6.88 (m, 4H), 6.78 (d, J = 2.0 Hz, 1H), 6.72 - 6.67 (m, 1H), 4.97 (s, 2H), 4.26 (dd, J = 5.2, 9.3 Hz, 1H), 3.97 (t, J = 6.4 Hz, 2H), 3.92 (s, 6H), 3.77 - 3.69 (m, 3H), 3.62 (s, 4H), 3.18 (d, J = 4.0 Hz, 6H), 3.08 - 2.96 (m, 4H), 2.70 - 2.57 (m, 4H), 2.36 - 2.26 (m, 2H), 2.09 (d, J = 18.0 Hz, 2H), 2.02 (s, 3H), 1.89 - 1.82 (m, 2H). Example 11. Preparation of Compound 152 6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(3-(4-(2-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin -1-yl)-2-oxoethyl)piperazin-1- yl)propoxy)-2-methylphenyl)picolinic acid Step A.1-Bromo-4-(3-bromopropoxy)-2-methylbenzene A mixture of 4-bromo-3-methylphenol (2 g, 10.69 mmol, 1 equiv.), 1,3-dibromopropane (10.79 g, 53.47 mmol, 5.45 mL, 5 equiv.), and K 2 CO 3 (4.43 g, 32.08 mmol, 3 equiv.) in MeCN (20 mL) was stirred at 70 °C. After 12 h, the reaction mixture was filtered and concentrated under the reduced pressure. The resulting crude material was purified by flash silica gel chromatography (0-5% ethyl acetate/petroleum ether) to give 1-bromo-4-(3-bromopropoxy)- 2-methylbenzene (1.55 g, 5.03 mmol, 47.1% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.44 (d, J = 8.8 Hz, 1H), 6.97 (d, J = 2.8 Hz, 1H), 6.74 (dd, J = 3.2, 8.8 Hz, 1H), 4.05 (t, J = 6.0 Hz, 2H), 3.65 (t, J = 6.4 Hz, 2H), 2.30 (s, 3H), 2.22 (m, J = 6.4 Hz, 2H). Step B. Ethyl 2-(4-(3-(4-bromo-3-methylphenoxy)propyl)piperazin-1-yl)aceta te A mixture of 1-bromo-4-(3-bromopropoxy)-2-methylbenzene (1.5 g, 4.87 mmol, 1 equiv.), ethyl 2-(piperazin-1-yl)acetate (838.73 mg, 4.87 mmol, 1 equiv.), and K 2 CO 3 (3.37 g, 24.35 mmol, 5 equiv.) in MeCN (15 mL) was stirred at 60 °C. After 14 h, the reaction mixture was filtered and concentrated under the reduced pressure. The resulting crude material was purified by silica gel chromatography (0-100% ethyl acetate/petroleum ether) to give ethyl 2-(4-(3-(4- bromo-3-methylphenoxy)propyl)piperazin-1-yl)acetate (1.44 g, 3.28 mmol, 67.4% yield, 91.0% purity) as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.53 (d, J = 8.8 Hz, 1H), 7.05 (d, J = 2.8 Hz, 1H), 6.82 (dd, J = 3.2, 8.8 Hz, 1H), 4.19 (q, J = 7.2 Hz, 2H), 4.07 (t, J = 6.4 Hz, 2H), 3.29 (s, 2H), 2.62 (dd, J = 2.0, 3.6 Hz, 4H), 2.56 - 2.45 (m, 6H), 2.41 (s, 3H), 1.99 - 1.91 (m, 2H), 1.30 (t, J = 7.2 Hz, 3H). Step C. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4-(3-(4-(2-ethoxy-2-oxoethyl)piperazin-1-yl)propoxy)- 2-methylphenyl)picolinate A mixture of ethyl 2-(4-(3-(4-bromo-3-methylphenoxy)propyl)piperazin-1-yl)aceta te (400 mg, 1.00 mmol, 1 equiv.), tert-butyl 6-(8-((3H-indol-2-yl)carbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pic olinate (612.55 mg, 1.00 mmol, 1 equiv.), and K 2 CO 3 (1.5 M, 1.00 mL, 1.5 equiv.) in dioxane (4 mL) was added cataCXium® A Pd G3 (145.65 mg, 200.00 µmol, 0.2 equiv.). The reaction mixture was purged with N 2 three times. The reaction mixture was stirred at 80 °C. After 2 h, the reaction solution was partitioned between H2O (10 mL) and ethyl acetate (10 mL). The organic phase was separated, washed with aqueous NaCl solution (3 mL × 2), dried over with Na 2 SO 4 , filtered, and concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography (0- 90% ethyl acetate/petroleum ether) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(4-(3-(4-(2-ethoxy-2-oxoe thyl)piperazin-1-yl)propoxy)- 2-methylphenyl)picolinate (692 mg, 733.27 µmol, 73.3% yield, 85.3% purity) as a yellow solid. 1 H NMR (400 MHz, DMSO-d6) δ = 13.01 - 12.64 (m, 1H), 8.02 (d, J = 7.6 Hz, 1H), 7.77 (d, J = 8.0 Hz, 1H), 7.60 (d, J = 7.6 Hz, 1H), 7.46 - 7.41 (m, 3H), 7.39 - 7.31 (m, 2H), 6.94 (d, J = 2.4 Hz, 1H), 6.86 (d, J = 8.4 Hz, 1H), 6.81 (d, J = 2.4 Hz, 1H), 6.73 (d, J = 2.0 Hz, 1H), 4.96 (s, 2H), 4.04 (s, 2H), 4.02 - 4.00 (m, 2H), 3.86 (t, J = 5.6 Hz, 2H), 3.17 (s, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.39 (d, J = 5.6 Hz, 8H), 2.33 - 2.25 (m, 3H), 2.00 (s, 2H), 1.83 (t, J = 6.8 Hz, 2H), 1.18 (d, J = 1.2 Hz, 3H), 1.03 (s, 9H). Step D. 2-(4-(3-(4-(6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-3-methylphenoxy)pro pyl)piperazin-1-yl)acetic acid tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(3-(4- (2-ethoxy-2-oxoethyl)piperazin-1-yl)propoxy)-2-methylphenyl) picolinate (380 mg, 472.05 umol, 1 equiv.) and LiOH•H2O (1 M, 1.42 mL, 3 equiv.) in THF (4 mL) and H2O (1 ml) was stirred at 25 °C. After 13 h, the reaction solution was concentrated, diluted with water (4 mL), and the pH was adjusted to 3 by the addition of 1 M HCl. The resulting slurry was filtered, and the filter cake was dried to give 2-(4-(3-(4-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-3- methylphenoxy)propyl)piperazin-1-yl)acetic acid (333 mg, crude) as a yellow solid which was carried forward without further purification. MS (ESI) m/z: 777.5 [M+H] + . Step E. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4-(3-(4-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1 H-indazol-7-yl)piperazin-1- yl)-2-oxoethyl)piperazin-1-yl)propoxy)-2-methylphenyl)picoli nate A mixture of 2-(4-(3-(4-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-3-methylphenoxy)pro pyl)piperazin-1-yl)acetic acid (170 mg, 218.81 µmol, 1 equiv.), HATU (91.52 mg, 240.69 umol, 1.1 equiv.), and N,N- diisopropyl-N-ethylamine (84.84 mg, 656.42 µmol, 114.34 µL, 3 equiv.) in DMF (2 mL) was added 3-(1-methyl-7-(piperazin-1-yl)-1H-indazol-3-yl)piperidine-2, 6-dione (71.63 mg, 218.81 µmol, 1 equiv.), and the mixture was stirred at 25 °C. After 12 h, the reaction solution was diluted with water (5 mL) and filtered. The filter cake was washed with dichloromethane (4 mL) and MeOH, and the filtrates were concentrated to provide tert-butyl 6-(8-(benzo[d]thiazol- 2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(4-(3-(4-( 2-(4-(3-(2,6-dioxopiperidin-3- yl)-1-methyl-1H-indazol-7-yl)piperazin-1-yl)-2-oxoethyl)pipe razin-1-yl)propoxy)-2- methylphenyl)picolinate (213 mg, crude) as a yellow solid, which was carried forward without further purification. MS (ESI) m/z: 1086.8 [M+H] + . Step F. 6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(3- (4-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7- yl)piperazin-1-yl)-2- oxoethyl)piperazin-1-yl)propoxy)-2-methylphenyl)picolinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4-(3-(4-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1 H-indazol-7-yl)piperazin-1-yl)-2- oxoethyl)piperazin-1-yl)propoxy)-2-methylphenyl)picolinate (200 mg, 184.11 µmol, 1 equiv.) in CH2Cl2 (3 mL) and trifluoracetic acid (1 mL) was stirred at 40 °C. After 18 h, the reaction solution was concentrated and purified by prep-HPLC (column: Phenomenex luna C18150 × 25 mm × 10 um; mobile phase: [water (1% formic acid)-MeCN]; B%: 25%-55%,10 min) to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(3-(4-(2- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pip erazin-1-yl)-2- oxoethyl)piperazin-1-yl)propoxy)-2-methylphenyl)picolinic acid (64.5 mg, 55.16 µmol, 30.0 % yield, 88.1% purity) as a green solid. MS (ESI) m/z: 1030.7 [M+H] + ; 1 H NMR (400 MHz, DMSO-d6) δ = 10.96 - 10.83 (m, 1H), 8.13 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.4 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.50 - 7.41 (m, 5H), 7.40 - 7.34 (m, 2H), 7.06 - 7.02 (m, 2H), 6.94 (t, J = 9.2 Hz, 2H), 6.79 (s, 1H), 6.72 (d, J = 9.2 Hz, 1H), 4.97 (s, 2H), 4.57 - 4.37 (m, 2H), 4.35 (dd, J = 5.2, 9.2 Hz, 1H), 4.27 (s, 3H), 4.01 (s, 4H), 3.91 (t, J = 5.2 Hz, 2H), 3.02 (t, J = 5.2 Hz, 4H), 2.98 - 2.92 (m, 2H), 2.87 - 2.75 (m, 4H), 2.73 - 2.65 (m, 4H), 2.64 - 2.56 (m, 4H), 2.38 - 2.27 (m, 2H), 2.24 - 2.09 (m, 2H), 2.03 (s, 5H). Example 12. Preparation of Compound 151 6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4-(2-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin -1-yl)-2-oxoethyl)piperazin-1- yl)propoxy)-2-methylphenyl)picolinic acid Step A. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-(4-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1 H-indazol-7-yl)piperazin-1- yl)-2-oxoethyl)piperazin-1-yl)propoxy)-2-methylphenyl)picoli nate To a solution of 3-(1-methyl-7-(piperazin-1-yl)-1H-indazol-3-yl)piperidine-2, 6-dione hydrochloride (50.56 mg, 139.0 µmol, 1.20 equiv.) in DMF (1 mL) was added N,N-diisopropyl- N-ethylamine (44.9 mg, 347.5 µmol, 60.53 µL, 3.00 equiv.), HOBt (20.4 mg, 150.6 µmol, 1.30 equiv.), EDCI (28.9 mg, 150.6 µmol, 1.30 equiv.), and 2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)propyl)piperazin-1-yl)acetic acid (90 mg, 115.8 µmol, 1.00 equiv.). The reaction solution was stirred at 25 °C. After 12 h, the reaction mixture was concentrated under reduced pressure and purified by prep-TLC (SiO2, DCM: MeOH = 20:1) to give tert-butyl 6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2( 1H)-yl)-3-(3-(3-(4-(2-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin -1-yl)-2-oxoethyl)piperazin-1- yl)propoxy)-2-methylphenyl)picolinate (110 mg, 101.3 µmol, 87.4% yield) as a yellow oil. MS (ESI) m/z: 1086.8 [M+H] + . Step B. 6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3- (4-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7- yl)piperazin-1-yl)-2- oxoethyl)piperazin-1-yl)propoxy)-2-methylphenyl)picolinic acid A solution tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)- 3-(3-(3-(4-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-in dazol-7-yl)piperazin-1-yl)-2- oxoethyl)piperazin-1-yl)propoxy)-2-methylphenyl)picolinate (110 mg, 101.3 µmol, 1.00 equiv.) in dichloromethane (1 mL) and trifluoroacetic acid (1 mL) was stirred at 40 °C. After 12 hours, the reaction mixture was concentrated and purified by prep-HPLC (column: Phenomenex luna C18 150*25mm* 10um;mobile phase: [water (1% formic acid )- MeCN];B%: 25%-55%,10min) to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(4-(2-(4-(3-(2,6-dioxop iperidin-3-yl)-1-methyl-1H- indazol-7-yl)piperazin-1-yl)-2-oxoethyl)piperazin-1-yl)propo xy)-2-methylphenyl)picolinic acid (22.8 mg, 21.1 µmol, 20.8% yield, 99.3% purity) as a white solid. MS (ESI) m/z: 1030.7 [M+H] + ; 1 H NMR (400 MHz, DMSO) δ = 10.89 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.49 - 7.33 (m, 7H), 7.09 (t, J = 8.0 Hz, 1H), 7.02 (d, J = 4.8 Hz, 2H), 6.97 (d, J = 8.8 Hz, 1H), 6.88 (d, J = 8.0 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.47 - 4.40 (m, 1H), 4.34 (d, J = 9.6 Hz, 1H), 4.27 (s, 3H), 4.22 - 4.15 (m, 1H), 3.99 (t, J = 5.6 Hz, 2H), 3.91 (t, J = 5.6 Hz, 2H), 3.18 (s, 8H), 3.02 (t, J = 5.6 Hz, 3H), 2.95 - 2.89 (m, 2H), 2.80 - 2.74 (m, 2H), 2.62 (d, J = 5.2 Hz, 2H), 2.54 (s, 4H), 2.36 - 2.29 (m, 2H), 2.16 (d, J = 13.2 Hz, 1H), 1.92 (d, J = 5.6 Hz, 2H) , 1.89 (s, 3H). Example 13. Preparation of Compound 146a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pip erazin-1-yl)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Ethyl (1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexane-1-carboxylate To a solution of ethyl 4-hydroxycyclohexanecarboxylate (10 g, 58.06 mmol, 1 equiv.), 3- bromo-2-methyl-phenol (11.95 g, 63.87 mmol, 1.1 equiv.), and PPh 3 (24.37 g, 92.90 mmol, 1.6 equiv.) in THF (100 mL) was added DIAD (18.79 g, 92.90 mmol, 18.06 mL, 1.6 equiv.) under 0 °C. Then the mixture was stirred at 25 °C for 16 h. The reaction mixture was concentrated under reduced pressure to give a residue that was purified by silica gel chromatography (ethyl acetate / petroleum ether gradient) to afford the title compound (3.6 g, 10.55 mmol, 18.17 % yield). 1 H NMR (400 MHz, CDCl3) δ 7.14 (d, J = 8.0 Hz, 1H), 6.97 (t, J = 8.0 Hz, 1H), 6.80 (d, J = 8.0 Hz, 1H), 4.20 - 4.11 (m, 3H), 2.39 - 2.35 (m, 2H), 2.30 (s, 3H), 2.19 - 2.14 (m, 2H), 2.09 - 2.04 (m, 2H), 1.62 - 1.52 (m, 5H), 1.26 (t, J = 6.8 Hz, 3H). Step B. ((1r,4r)-4-(3-Bromo-2-methylphenoxy)cyclohexyl)methanol To a solution of ethyl 4-(3-bromo-2-methyl-phenoxy)cyclohexanecarboxylate (3.6 g, 10.55 mmol, 1 equiv.) in THF (15 mL) was added LiAlH4 (480.49 mg, 12.66 mmol, 1.2 equiv.) under 0 °C. The resulting mixture was then stirred at 25 °C for 1.5 h. The reaction was quenched by the addition of a saturated aqueous solution of sodium sulfate (10 mL), and then extracted with ethyl acetate (10 mL × 3). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to afford the title compound (2.96 g, crude) that was used without further purification. 1 H NMR (400 MHz, CDCl3) δ 7.13 (d, J = 8.0 Hz, 1H), 6.97 (t, J = 8.0 Hz, 1H), 6.81 (d, J = 8.4 Hz, 1H), 4.13 - 4.08(m, 1H), 3.50 (d, J = 6.4 Hz, 2H), 2.30 (s, 3H), 2.20 - 2.14 (m, 2H), 1.95 - 1.86 (m, 2H), 1.53 - 1.45 (m, 3H), 1.14 - 1.04 (m, 2H). Step C. (1r,4r)-4-(3-Bromo-2-methylphenoxy)cyclohexane-1-carbaldehyd e To a solution of DMSO (3.09 g, 39.57 mmol, 3.09 mL, 4 equiv.) in DCM (10 mL) was added dropwise a solution of oxalyl chloride (2.51 g, 19.79 mmol, 1.73 mL, 2 equiv.) in DCM (2 mL) at -70 °C under a nitrogen atmosphere. The mixture was stirred at -70 °C for 1 h. [4-(3-bromo- 2-methyl-phenoxy)cyclohexyl]methanol (2.96 g, 9.89 mmol, 1 equiv.) in DCM (10 mL) was then added dropwise at -70 °C. The solution was stirred for 1 h at -70 °C. TEA (6.01 g, 59.36 mmol, 8.26 mL, 6 equiv.) was added, and the mixture was stirred at -70 °C for 0.5 h under a nitrogen atmosphere. The reaction mixture was quenched by the addition of water (10 mL), diluted with DCM (10 mL), and extracted with DCM (10 mL × 3). The combined organic layers were washed with water (5 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to afford (1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexane-1-carbaldehyde (3 g, crude) that was used without further purification. 1 H NMR (400 MHz, DMSO-d6) δ 9.62 - 9.59 (m, 1H), 7.16 - 7.01 (m, 3H), 4.36 - 4.23 (m, 1H), 2.39 - 2.31 (m, 1H), 2.25 - 2.18 (m, 3H), 2.03 - 1.92 (m, 4H), 1.50 - 1.38 (m, 4H). Step D. Ethyl (E)-3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)acrylat e To a solution of NaH (56.52 mg, 1.41 mmol, 60% purity, 2.1 equiv.) in THF (3 mL) at 0 °C was added ethyl 2-diethoxyphosphorylacetate (301.75 mg, 1.35 mmol, 267.04 mL, 2 equiv.). (1r,4r)-4-(3-Bromo-2-methylphenoxy)cyclohexane-1-carbaldehyd e (200 mg, 672.98 mmol, 1 equiv.) was then added, and the mixture was stirred at 25 °C for 6 h. The reaction mixture was quenched by addition of a saturated aqueous solution of ammonium chloride (5 mL) at 0 °C, and then extracted with ethyl acetate (10 mL × 3). The combined organic layers were washed with water (10 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue which was purified by silica gel chromatography (ethyl acetate / petroleum ether gradient) to afford ethyl (E)-3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)acrylate (140 mg, 362.12 mmol, 53.81 % yield). 1 H NMR (400 MHz, CDCl3) δ 7.14 (d, J = 8.0 Hz, 1H), 7.01 - 6.87 (m, 2H), 6.80 (d, J = 8.4 Hz, 1H), 5.82 (dd, J = 1.2, 15.6 Hz, 1H), 4.21 - 4.15 (m, 2H), 4.16 - 4.07 (m, 1H), 2.30 (s, 3H), 2.26 - 2.16 (m, 3H), 1.97 - 1.89 (m, 2H), 1.58 - 1.48 (m, 2H), 1.33 - 1.26 (m, 5H). Step E. Ethyl 3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propanoate A mixture of ethyl (E)-3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]prop-2-enoate (1.6 g, 4.36 mmol, 1 equiv.), PtO2 (98.92 mg, 435.64 mmol, 0.1 equiv.) in EtOH (15 mL) was degassed and purged with hydrogen three times. The mixture was stirred at 25 °C for 3 hours under a hydrogen atmosphere (balloon, ~ 15 psi). The reaction mixture was filtered and concentrated under reduced pressure to give a residue that was purified by preparative RP-HPLC (water/acetonitrile with 0.1% formic acid in each). Combined and lyophilized pure product containing fractions to afford ethyl 3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propanoate (1.4 g, 3.79 mmol, 87.02 % yield). 1 H NMR (400 MHz, DMSO-d6) δ 7.17 - 7.10 (m, 1H), 7.06 (t, J = 8.0 Hz, 1H), 7.03 - 6.99 (m, 1H), 4.27 - 4.17 (m, 1H), 4.11 - 4.04 (m, 2H), 2.33 - 2.26 (m, 2H), 2.20 (s, 3H), 2.03 (d, J = 10.0 Hz, 2H), 1.75 (d, J = 12.0 Hz, 2H), 1.55 - 1.42 (m, 2H), 1.39 - 1.21 (m, 3H), 1.17 (t, J = 7.2 Hz, 3H), 1.11 - 0.97 (m, 2H). Step F. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1r,4r)-4-(3-ethoxy-3-oxopropyl)cyclohexyl)oxy)-2 -methylphenyl)picolinate A mixture of ethyl 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]propanoate (200 mg, 541.58 mmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (546.40 mg, 758.21 mmol, 1.4 equiv.), [2-(2-aminophenyl)phenyl]palladium(1+);bis(1- adamantyl)-butyl-phosphane;methanesulfonate (78.88 mg, 108.32 mmol, 0.2 equiv.), and an aqueous solution of potassium carbonate (1.5 M, 541.58 mL, 1.5 equiv.) in dioxane (2.5 mL) was degassed and purged with nitrogen three times. The mixture was stirred at 100 °C for 1 h under microwave heating. The reaction mixture was filtered and concentrated under reduced pressure to provide a residue that was purified by silica gel chromatography (ethyl acetate / petroleum ether gradient) to afford the title compound (440 mg, crude). 1 H NMR (400 MHz, DMSO-d6) δ 8.02 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.60 (d, J = 7.5 Hz, 1H), 7.50 - 7.40 (m, 3H), 7.40 - 7.30 (m, 2H), 7.12 - 7.03 (m, 1H), 6.99 - 6.89 (m, 2H), 6.59 - 6.49 (m, 1H), 5.03 - 4.90 (m, 2H), 4.27 - 4.14 (m, 1H), 4.03 (d, J = 6.8 Hz, 2H), 3.87 (t, J = 5.6 Hz, 2H), 3.29 (s, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.29 (t, J = 7.6 Hz, 2H), 2.05 (d, J = 11.2 Hz, 1H), 1.84 (s, 3H), 1.79 - 1.70 (m, 2H), 1.50 - 1.42 (m, 2H), 1.23 (d, J = 8.4 Hz, 2H), 1.19 - 1.17 (m, 3H), 1.07 (s, 2H), 1.00 (s, 9H). Step G. 3-((1r,4r)-4-(3-(6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-di hydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)cyclohexyl)propanoic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1r,4r)-4-(3-ethoxy-3-oxopropyl)cyclohexyl)oxy)-2 -methylphenyl)picolinate (440 mg, 567.77 mmol, 1 equiv.) and LiOH monohydrate (71.47 mg, 1.70 mmol, 3 equiv.) in THF (1.2 mL) and water (0.4 mL) was stirred at 25 °C for 1.5 h. The mixture was concentrated and redissolved in water (20 mL), and an aqueous solution of 1M HCl was added to adjust the pH to 2. The resulting solids were filtered and dried under reduced pressure to afford the title compound (380 mg, 508.76 mmol, 89.61 % yield). Step H. tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1r,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-7- yl)piperazin-1-yl)-3-oxopropyl)cyclohexyl)oxy)-2-methylpheny l)picolinate A mixture of 3-((1r,4r)-4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-di hydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)cyclohexyl)propanoic acid (100 mg, 133.88 umol, 1 equiv.), 3-(1-methyl-7-piperazin-1-yl-indazol-3-yl)piperidine-2,6- dione (48.21 mg, 147.27 umol, 1.1 equiv.), and EDCI (32.08 mg, 167.35 µmol, 1.25 equiv.) in pyridine (1 mL) was stirred at 25 °C for 12 hours. The mixture was diluted with water (2 mL) and extracted with ethyl acetate (1 mL × 3). The combined organic layers were filtered and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-(4-(3-( 2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)-3-oxopropyl)cyclohexy l)oxy)-2- methylphenyl)picolinate (120 mg, crude) as a yellow oil which was carried forward without further purification. MS(ESI) m/z: 1056.7 [M+H] + . Step I. 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1r,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-7-yl)piperazin-1-yl)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1r,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-7-yl)piperazin-1-yl)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (100 mg, 94.67 umol, 1 eq.) in trifluoroacetic acid (0.5 mL) and dichloromethane (0.5 mL) was stirred at 40 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The resulting residue was purified by prep-HPLC (column: Phenomenex luna C18150 × 25mm × 10um; mobile phase: [water (1% formic acid)-MeCN]; 63% - 93%, 15 min) to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1r,4r)-4-(3-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin -1-yl)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (31.1 mg, 28.2 umol, 29.8% yield) as a yellow solid. MS(ESI) m/z: 1000.6 [M+H] + ; 1 HNMR (400 MHz, DMSO-d 6 ) δ = 12.95 - 12.80 (m, 1H), 12.75 - 12.42 (m, 1H), 10.94 - 10.85 (m, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.66 - 7.59 (m, 1H), 7.48 - 7.34 (m, 6H), 7.11 - 7.01 (m, 3H), 6.94 (dd, J = 8.4, 15.6 Hz, 2H), 6.61 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.37 - 4.31 (m, 1H), 4.27 (s, 3H), 4.23 - 4.14 (m, 1H), 4.07 - 3.81 (m, 3H), 3.25 - 3.14 (m, 2H), 3.10 - 3.00 (m, 2H), 2.96 - 2.84 (m, 1H), 2.84 - 2.73 (m, 1H), 2.70 - 2.60 (m, 3H), 2.41 - 2.31 (m, 3H), 2.21 - 2.02 (m, 4H), 1.89 - 1.79 (m, 5H), 1.50 - 1.43 (m, 2H), 1.42 - 1.25 (m, 4H), 1.14 - 1.03 (m, 2H). Example 14. Preparation of Compound 143

6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(1-(2-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin -1-yl)-2-oxoethyl)piperidin-4- yl)ethoxy)-2-methylphenyl)picolinic acid Step A. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(2-(1-(2-ethoxy-2-oxoethyl)piperidin-4-yl)ethoxy)-2 -methylphenyl)picolinate A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)picolinat e (300 mg, 489.8 µmol, 1.00 equiv.), ethyl 2-(4-(2-(3-bromo-2-methylphenoxy)ethyl)piperidin-1-yl)acetat e (207.0 mg, 538.7 µmol, 1.10 equiv.), cataCXium Pd G3 (35.7 mg, 49.0 µmol, 0.10 equiv.), and KF (85.37 mg, 1.47 mmol, 34.4 µL, 3.00 equiv.) in dioxane (4 mL) and water (1 mL) was stirred at 100 °C under N2 atmosphere. After 1 h, the reaction solution was concentrated and purified by prep- TLC (SiO 2 , petroleum ether : ethyl acetate = 1:1,Rf=0.12) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(2-(1-(2-ethoxy-2- oxoethyl)piperidin-4-yl)ethoxy)-2-methylphenyl)picolinate (360 mg, 455.7 µmol, 93.1% yield) as a yellow solid. MS (ESI) m/z: 790.7 [M+H] + . Step B. 2-(4-(2-(3-(6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)eth yl)piperidin-1-yl)acetic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(1-(2-ethoxy-2-oxoethyl)piperidin-4-yl)eth oxy)-2-methylphenyl)picolinate (360 mg, 455.7 µmol, 1.00 equiv.) in THF (4 mL) and H2O (1 mL) was added LiOH.H2O (57.4 mg, 1.37 mmol, 3.00 equiv.). The mixture was stirred at 25 °C. After 12 h, the reaction solution was acidified to pH = 5 with 1M HCl and extracted with EtOAc (20 mL × 3). The combined organic layers were washed with brine (30 mL × 1), dried over Na2SO4, filtered, and concentrated to provide 2-(4-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)ethyl)piperidin-1-yl)acetic acid (300 mg, 393.74 umol, 86.4% yield) as a yellow solid, which was carried forward without further purification. MS (ESI) m/z: 762.6 [M+H] + . Step C. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(2-(1-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1 H-indazol-6-yl)piperazin-1- yl)-2-oxoethyl)piperidin-4-yl)ethoxy)-2-methylphenyl)picolin ate To a solution of 2-(4-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)ethyl)piperidin-1-yl)acetic acid (100 mg, 131.3 µmol, 1.00 equiv.) in DMF (1 mL) was added N,N-diisopropyl-N- ethylamine (50.9 mg, 393.7 µmol, 68.6 µL, 3.00 equiv.), HATU (59.9 mg, 157.5 µmol, 1.20 equiv.), and 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne hydrochloride (52.5 mg, 144.4 µmol, 1.10 equiv.). The reaction solution was stirred at 25 °C. After 1 h, the reaction solution was diluted with water (20 mL) and extracted with dichloromethane (30 mL × 3). The combined organic layers were washed with brine, dried over Na2SO4, filtered, and concentrated to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(1-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)piperazin- 1-yl)-2-oxoethyl)piperidin-4-yl)ethoxy)-2-methylphenyl)picol inate (120 mg, 112.01 umol, 85.35% yield) as a yellow solid, which was carried forward without further purification. MS (ESI) m/z: 1071.8 [M+H] + . Step D. 6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2- (1-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)piperazin-1-yl)-2- oxoethyl)piperidin-4-yl)ethoxy)-2-methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(1-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)piperazin- 1-yl)-2-oxoethyl)piperidin-4-yl)ethoxy)-2-methylphenyl)picol inate (120 mg, 112.01 µmol, 1.00 equiv.) in DCM (1 mL) was added trifluoroacetic acid (1.54 g, 13.51 mmol, 1 mL, 120.6 equiv.). The reaction mixture was stirred at 40 °C. After 1 h the reaction solution was concentrated and purified by prep-HPLC (column: Phenomenex luna C 18 150×25mm×10um;mobile phase: [water (1% formic acid)-acetonitrile];B%: 25%-55%, 10min) to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (2-(1-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)piperazin-1-yl)-2- oxoethyl)piperidin-4-yl)ethoxy)-2-methylphenyl)picolinic acid (23.1 mg, 22.2 µmol, 19.8% yield, 97.8% purity) as a white solid. MS (ESI) m/z: 1015.8 [M+H] + ; 1 H NMR (400 MHz, DMSO-d6) δ = 10.86 (s, 1H), 8.04 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 7.6 Hz, 1H), 7.63 (d, J = 7.2 Hz, 1H), 7.53 (d, J = 8.8 Hz, 1H), 7.50 - 7.32 (m, 6H), 7.12 - 7.05 (m, 1H), 6.95 (d, J = 8.8 Hz, 2H), 6.91 - 6.86 (m, 2H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.29 - 4.25 (m, 1H), 4.02 - 3.98 (m, 2H), 3.92 (s, 2H), 3.90 (s, 3H), 3.75 (s, 2H), 3.63 (s, 2H), 3.26 - 3.25 (m, 2H), 3.18 (s, 4H), 3.03 (t, J = 5.6 Hz, 2H), 2.84 (d, J = 11.6 Hz, 2H), 2.63 - 2.59 (m, 2H), 2.33 - 2.25 (m, 1H), 2.21 - 2.13 (m, 1H), 2.05 - 1.97 (m, 2H), 1.89 (s, 3H), 1.74 - 1.74 (m, 1H), 1.74 - 1.66 (m, 4H), 1.54 - 1.47 (m, 1H), 1.28 - 1.20 (m, 2H) Example 15. Preparation of Compound 140 6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4-(2-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin -1-yl)-2-oxoethyl)piperazin-1- yl)propoxy)-2-methylphenyl)picolinic acid Step A. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-(4-(2-ethoxy-2-oxoethyl)piperazin-1-yl)propoxy)- 2-methylphenyl)picolinate To a solution of ethyl 2-(4-(3-(3-bromo-2-methylphenoxy)propyl)piperazin-1-yl)aceta te (201.5 mg, 504.6 µmol, 1.00 equiv.) in 1,4-dioxane (6 mL) and H 2 O (0.6 mL) were added cataCXium Pd G3 (36.75 mg, 50.5 µmol, 0.10 equiv.), KF (87.95 mg, 1.51 mmol, 35.46 µL, 3.00 equiv.) and tert-butyl 6-(8-((3H-indol-2-yl)carbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)- 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)picolinate (300 mg, 504.62 µmol, 1 equiv.). The mixture was stirred at 100 °C for 1 hour under nitrogen atmosphere. The reaction mixture was concentrated and purified by prep-TLC (SiO2, DCM: MeOH = 20:1, Rf = 0.4) to give tert- butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4-(2- ethoxy-2-oxoethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pic olinate (220 mg, 273.29 µmol, 54.2% yield) as a yellow oil. MS (ESI) m/z: 805.4 [M+H] + . Step B. 2-(4-(3-(3-(6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy) propyl)piperazin-1-yl)acetic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4-(2-ethoxy-2-oxoethyl)piperazin-1-yl)pro poxy)-2- methylphenyl)picolinate (220 mg, 273.29 µmol, 1.00 equiv.) in THF (4 mL) was added LiOH•H2O (19.6 mg, 819.88 µmol, 3.00 equiv.) and H2O (1 mL). The reaction mixture was stirred at 25 °C. After 12 h, the reaction solution was acidified to pH = 3 with aqueous citric acid, filtered, and concentrated to provide 2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyri din-3-yl)-2-methylphenoxy) propyl)piperazin-1-yl)acetic acid (175 mg, 225.2 µmol, 82.4% yield) as a yellow solid, which was carried forward without further purification. MS (ESI) m/z: 777.6 [M+H] + . Step C. tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-(4-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1 H-indazol-6-yl)piperazin-1- yl)-2-oxoethyl)piperazin-1-yl)propoxy)-2-methylphenyl)picoli nate To a solution of 3-(1-methyl-6-(piperazin-1-yl)-1H-indazol-3-yl)piperidine-2, 6-dione hydrochloride (41.21 mg, 113.27 µmol, 1.10 equiv.) and 2-(4-(3-(3-(6-(8-(Benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy) propyl)piperazin-1-yl)acetic acid (80 mg, 102.97 umol, 1.00 equiv.) in DMF (1 mL) were added N,N-diisopropyl-N-ethylamine (39.9 mg, 308.9 µmol, 53.80 µL, 3.00 equiv.) and HATU (58.7 mg, 154.5 µmol, 1.50 equiv.). The reaction solution was stirred at 25 °C. After 1 h, the reaction solution was concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4-(2-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin -1-yl)-2-oxoethyl)piperazin-1- yl)propoxy)-2-methylphenyl)picolinate (110 mg, 101.26 µmol, 98.3% yield) as a yellow solid, which was carried forward without further purification. MS (ESI) m/z: 1086.7 [M+H] + . Step D. 6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3- (4-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)piperazin-1-yl)-2- oxoethyl)piperazin-1-yl)propoxy)-2-methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)piperazin- 1-yl)-2-oxoethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linate (110 mg, 101.3 µmol, 1.00 equiv.) in DCM (1 mL) was added trifluoroacetic acid (1.99 g, 17.48 mmol, 1.00 mL, 172.61 equiv.). The reaction solution was stirred at 40 °C. After 1 h, the reaction mixture was concentrated and purified by prep-HPLC (column: Phenomenex luna C18 150*25mm* 10um;mobile phase: [water (1% formic acid)-ACN];B%: 22%-52%,10min) to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-(4-(2-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin-1-yl )-2-oxoethyl)piperazin-1- yl)propoxy)-2-methylphenyl)picolinic acid (22.1 mg, 20.0 µmol, 19.7% yield, 97.3% purity) as a white solid. MS (ESI) m/z: 1030.7 [M+H] + ; 1 H NMR (400 MHz, DMSO-d6) δ = 10.84 (s, 1H), 8.16 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.63 (m, 1H),7.53 (d, J = 8.8 Hz, 1H), 7.49 - 7.33 (m, 6H), 7.11 - 7.07 (m, 1H), 6.95 (d, J = 8.8 Hz, 2H), 6.90 - 6.86 (m, 2H), 6.64 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.27 (d, J = 9.2 Hz, 1H), 3.98 (t, J = 5.6 Hz, 2H), 3.94 - 3.90 (m, 2H), 3.90 (s, 3H), 3.75 - 3.73 (m, 2H), 3.63 - 3.61 (m, 2H), 3.28-3.26 (m, 4H), 3.20 - 3.18 (m, 4H), 3.03 (t, J = 5.6 Hz, 2H), 2.62 (t, J = 5.6 Hz, 2H), 2.48 - 2.44 (m, 8H), 2.34 - 2.30 (m, 1H), 2.16 (d, J = 13.2 Hz, 1H), 1.89 (s, 3H), 1.88-1.86 (m, 2H). Example 16. Preparation of Compound 220 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[1-[2-[4-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]piperazin-1-yl ]ethyl]-4-piperidyl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. tert-Butyl 4-[3-(3-bromo-2-methyl-phenoxy)propyl]piperidine-1-carboxyla te To a solution of 3-bromo-2-methyl-phenol (1 g, 5.35 mmol, 1 eq.) and tert-butyl 4-(3- bromopropyl)piperidine-1-carboxylate (1.64 g, 5.35 mmol, 1 equiv.) in MeCN (4 mL) was added K 2 CO 3 (2.22 g, 16.04 mmol, 3 eq.). The mixture was stirred at 60 °C. After 2 h, the reaction solution was concentrated under reduced pressure purified by column chromatography (SiO2, petroleum ether/ethyl acetate=1/0 to 10/1) to give tert-butyl 4-[3-(3-bromo-2-methyl- phenoxy)propyl]piperidine-1-carboxylate (2.2 g, 5.34 mmol, 99.8% yield) as a colorless oil. MS(ESI) m/z: 312.4 [M+H] + ; 1 H NMR (400 MHz, CDCl 3 ) δ = 7.14 (d, J = 8.0 Hz, 1H), 6.99 (t, J = 8.0 Hz, 1H), 6.76 (d, J = 8.4 Hz, 1H), 4.14 - 4.06 (m, 2H), 3.94 (t, J = 6.4 Hz, 2H), 2.69 (t, J = 2.4, 12.8 Hz, 2H), 2.32 (s, 3H), 1.88 - 1.79 (m, 2H), 1.70 (d, J = 12.8 Hz, 2H), 1.48 - 1.43 (m, 12H), 1.18 - 1.08 (m, 2H) Step B.4-[3-(3-bromo-2-methyl-phenoxy)propyl]piperidine To a solution of tert-butyl 4-[3-(3-bromo-2-methyl-phenoxy)propyl]piperidine-1-carboxyla te (2.2 g, 5.34 mmol, 1 equiv.) in 4 M HCl in EtOAc (10 mL) was stirred at 25 °C. After 1 h, the reaction mixture was filtered and concentrated under reduced pressure to give 4-[3-(3-bromo- 2-methyl-phenoxy)propyl]piperidine (1.6 g, 5.12 mmol, 96.05% yield) as a white solid which was carried forward without further purification. MS(ESI) m/z: 314.4 [M+H] + ; 1 HNMR (400 MHz, CDCl 3 ) δ = 7.14 (d, J = 8.0 Hz, 1H), 6.99 (t, J = 8.0 Hz, 1H), 6.76 (d, J = 8.4 Hz, 1H), 4.14 - 4.06 (m, 2H), 3.94 (t, J = 6.4 Hz, 2H), 2.69 (t, J = 2.4, 12.8 Hz, 2H), 2.32 (s, 3H), 1.88 - 1.79 (m, 2H), 1.70 (d, J = 12.8 Hz, 2H), 1.48 - 1.43 (m, 12H), 1.18 - 1.08 (m, 2H). Step C.4-[3-(3-bromo-2-methyl-phenoxy)propyl]-1-(2,2-diethoxyethy l)piperidine To a solution of 4-[3-(3-bromo-2-methyl-phenoxy)propyl]piperidine (1.5 g, 4.80 mmol, 1 equiv.) and 2-bromo-1,1-diethoxy-ethane (946.71 mg, 4.80 mmol, 722.68 uL, 1 equiv.) in MeCN (5 mL) was added K 2 CO 3 (1.99 g, 14.41 mmol, 3 equiv.) and potassium iodide (797.46 mg, 4.80 mmol, 1 equiv.). The mixture was stirred at 80 °C for 8 hours. The reaction mixture was concentrated under reduced pressure and purified by column chromatography (SiO2, Ethyl acetate/MeOH=1/0 to 20/1) to give 4-[3-(3-bromo-2-methyl-phenoxy)propyl]-1-(2,2- diethoxyethyl)piperidine (1.7 g, 3.97 mmol, 82.6% yield) as a yellow oil. MS(ESI) m/z: 430.5 [M+H] + . Step D. tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[1-(2,2-diethoxyethyl)-4-piperidyl]propoxy]-2-me thyl-phenyl]pyridine-2- carboxylate A mixture of 4-[3-(3-bromo-2-methyl-phenoxy)propyl]-1-(2,2-diethoxyethyl) piperidine (800 mg, 1.87 mmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (1.14 g, 1.87 mmol, 1 equiv.), [2-(2-aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-buty l- phosphane;methanesulfonate (136.00 mg, 186.74 umol, 0.1 eq.), and KF (1.5 M, 3.73 mL, 3 equiv.) in dioxane (5 mL) and H 2 O (1 mL) was degassed and purged with N 2 three times. The reaction mixture was then stirred at 100 °C in a microwave reactor. After 1 h, the reaction solution was concentrated under reduced pressure and purified by column chromatography (SiO 2 , ethyl acetate/MeOH=1/0 to 10/1) to give tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-(2, 2-diethoxyethyl)-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate (1.5 g, 1.72 mmol, 91.99% yield, 95.52% purity) as a yellow oil, which was carried forward without further purification. MS(ESI) m/z: 834.8 [M+H] + ; 1 H NMR (400 MHz, CDCl3) δ = 7.85 (d, J = 8.0 Hz, 1H), 7.59 (dd, J = 7.2, 15.6 Hz, 2H), 7.40 - 7.30 (m, 5H), 7.14 - 7.06 (m, 1H), 6.90 (d, J = 8.8 Hz, 1H), 6.80 (d, J = 8.4 Hz, 1H), 6.69 (d, J = 7.6 Hz, 1H), 5.12 - 4.95 (m, 2H), 4.81 - 4.60 (m, 1H), 4.12 - 4.08 (m, 2H), 3.97 (t, J = 6.4 Hz, 2H), 3.72 - 3.68 (m, 2H), 3.61 - 3.54 (m, 2H), 3.07 (t, J = 5.2 Hz, 4H), 2.64 - 2.52 (m, 2H), 2.05 (s, 3H), 1.84 - 1.80 (m, 2H), 1.69 - 1.55 (m, 9H), 1.24 - 1.20 (m, 6H), 1.15 (s, 9H). Step E. 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[2- methyl-3-[3-[1-(2-oxoethyl)-4-piperidyl]propoxy]phenyl]pyrid ine-2-carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[3-[1-(2,2-diethoxyethyl)-4-piperidyl]propoxy]-2- methyl-phenyl]pyridine-2- carboxylate (100 mg, 119.89 umol, 1 equiv.) in formic acid (2 mL) was stirred at 90 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[2-methyl-3-[3-[1-(2- oxoethyl)-4-piperidyl]propoxy]phenyl]pyridine-2-carboxylic acid hydrochloride (70 mg, 99.5 umol, 83% yield) as a dark green oil, which was carried forward without further purification. MS(ESI) m/z: 722.5 [M+H] + . Step F. 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3- [1-[2-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]pi perazin-1-yl]ethyl]-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid To a solution of 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (83.72 mg, 255.74 umol, 0.9 eq.) and NaBH(OAc) 3 (180.67 mg, 852.46 umol, 3.0 eq.) in isopropanol (1.5 mL) and DCM (1.5 mL) was added 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[2-methyl-3-[3-[1-(2-oxoethyl)-4-piperid yl]propoxy]phenyl]pyridine-2- carboxylic acid (200 mg, 284.15 umol, 1.0 eq.) at 0 °C slowly. The mixture was stirred at 0 °C for 1 hour, then the mixture was warmed to 25 °C and stirred at 25 °C for 12 hours. The mixture was concentrated under reduced pressure, diluted with N,N-dimethylformamide (2 mL) and purified by prep-HPLC (column: Phenomenex luna C18150× 25mm× 10um; mobile phase: [water (1% formic acid)-acetonitrile]; B%: 20%-50%, 8min) to give 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-[2- [4-[3-(2,6-dioxo-3-piperidyl)- 1-methyl-indazol-6-yl]piperazin-1-yl]ethyl]-4-piperidyl]prop oxy]-2-methyl-phenyl]pyridine- 2-carboxylic acid (34.9 mg, 32.8 umol, 11.5% yield, 99.6% purity) as a white solid. MS (ESI) m/z: 1016.8 [M+H] + ; 1 H NMR (400 MHz, DMSO-d6) δ = 10.85 (s, 1H), 8.01 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.64 - 7.59 (m, 1H), 7.54 - 7.26 (m, 6H), 7.10 - 7.03 (m, 1H), 6.93 - 6.89 (m, 1H), 6.88 - 6.78 (m, 3H), 6.66 (d, J = 7.6 Hz, 1H), 4.96 (s, 2H), 4.25 (dd, J = 5.2, 9.2 Hz, 1H), 3.95 (s, 4H), 3.91 - 3.87 (m, 5H), 3.19 (s, 4H), 3.03 - 3.95 (m, 4H), 2.65 - 2.58 (m, 4H), 2.58 - 2.54 (m, 4H), 2.34 - 2.26 (m, 1H), 2.22 - 2.10 (m, 3H), 1.90 (s, 3H), 1.77 - 1.66 (m, 2H), 1.65 - 1.52 (m, 2H), 1.37 - 1.22 (m, 3H), 1.21 - 0.94 (m, 2H). Example 17. Preparation of Compound 117a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[(3S,5R)-4- [2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]- 2-oxo-ethyl]-3,5-dimethyl- piperazin-1-yl]propoxy]-2-methyl-phenyl]pyridine-2-carboxyli c acid Step A. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(3S,5R)-4-[2-[[3-( 2,6-dioxo-3-piperidyl)-1- methyl-indazol-7-yl]amino]-2-oxo-ethyl]-3,5-dimethyl-piperaz in-1-yl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylate A mixture of 2-[(2S,6R)-4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3, 4-dihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]propyl]-2,6-dimethyl- piperazin-1-yl]acetic acid (100 mg, 124.22 µmol, 1 equiv.), 3-(7-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (32.08 mg, 124.22 µmol, 1 equiv.), HATU (47.23 mg, 124.22 µmol, 1 equiv.), DIEA (16.06 mg, 124.22 µmol, 21.64 µL, 1 equiv.) in DMF (2 mL), and then the mixture was stirred at 60 o C for 12 hours. H2O (20 mL) was then added dropwise into the reaction mixture, which was filtered to give compound tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(3S,5 R)-4-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol-7-yl]amino]-2-oxo-ethyl]-3,5-dim ethyl-piperazin-1-yl]propoxy]- 2-methyl-phenyl]pyridine-2-carboxylate (90 mg, 86.1 µmol, 69.3% yield) as a yellow solid. MS (ESI) m/z: 523.4 [M/2+H] + Step B. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[(3S,5R)-4-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol- 7-yl]amino]-2-oxo-ethyl]-3,5-dimethyl-piperazin-1-yl]propoxy ]-2-methyl- phenyl]pyridine-2-carboxylic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[(3S,5R)-4-[2-[[3-(2,6-dioxo-3-piperidyl)-1-meth yl-indazol-7-yl]amino]-2-oxo- ethyl]-3,5-dimethyl-piperazin-1-yl]propoxy]-2-methyl-phenyl] pyridine-2-carboxylate (90 mg, 86.10 µmol, 1 equiv.) in DCM (1 mL) and TFA (1 mL) was stirred at 40 o C for 4 hours. The reaction mixture was concentrated under reduced pressure to remove solvent to give the crude product. The crude product was purified by reverse-phase HPLC to give compound 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[3-[(3S,5R)-4-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-oxo-e thyl]-3,5-dimethyl-piperazin- 1-yl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid (20 mg, 17.8 µmol, 20.6% yield, 88% purity) as a white solid. MS (ESI) m/z: 989.3 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 10.96 (s, 1H), 9.87 (s, 1H), 8.21 (s, 1H), 8.09 (d, J = 8.0 Hz, 1H), 7.85 (d, J = 8.0 Hz, 1H), 7.68 (d, J = 8.4 Hz, 1H), 7.63 (d, J = 8.0 Hz, 1H), 7.58 - 7.34 (m, 7H), 7.20 - 7.09 (m, 2H), 7.02 (d, J = 8.8 Hz, 1H), 6.94 (d, J = 8.4 Hz, 1H), 6.70 (d, J = 7.6 Hz, 1H), 5.04 (s, 2H), 4.44 (dd, J = 5.2, 10.0 Hz, 1H), 4.18 (s, 3H), 4.06 ( t, J = 5.6 Hz, 2H), 3.98 ( t, J = 5.6 Hz, 2H), 3.65 (s, 1H), 3.09 ( t, J = 5.6 Hz, 2H), 2.87 ( d, J = 10. Hz, 2H), 2.81 - 2.69 (m, 4H), 2.53 - 2.36 (m, 4H), 2.22 (dd, J = 5.2, 13.2 Hz, 1H), 2.00 - 1.90 (m, 7H), 1.15 ( d, J = 6.0 Hz, 6H). Example 18. Preparation of Compound 116a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[(3S,5R)-4- [2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]- 2-oxo-ethyl]-3,5-dimethyl- piperazin-1-yl]propoxy]-2-methyl-phenyl]pyridine-2-carboxyli c acid Step A. Procedure for preparation of 1-bromo-3-(3-bromopropoxy)-2-methyl-benzene To a solution of 3-bromo-2-methyl-phenol (2 g, 10.69 mmol, 1 equiv.) in acetone (30 mL) were added 1,3-dibromopropane (5.18 g, 25.66 mmol, 2.62 mL, 2.4 equiv.) and K2CO3 (4.43 g, 32.08 mmol, 3 equiv.), the mixture was stirred at 60 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give the residue. The residue was purified by flash silica gel chromatography (eluent of 0~5% ethyl acetate/petroleum ether) to give the compound 1-bromo-3-(3-bromopropoxy)-2-methyl-benzene (1.6 g, 5.2 mmol, 48.6% yield) as a colorless oil. 1 H NMR (400 MHz, CDCl3) δ = 7.09 (d, J = 8.0 Hz, 1H), 6.93 (t, J = 8.0 Hz, 1H), 6.71 (d, J = 8.4 Hz, 1H), 4.02 (t, J = 5.6 Hz, 2H), 3.54 (t, J = 6.4 Hz, 2H), 2.27 (t, J = 6.0 Hz, 2H), 2.24 (s, 3H) Step B. Procedure for preparation of (3S,5R)-1-[3-(3-bromo-2-methyl-phenoxy)propyl]- 3,5-dimethyl-piperazine A mixture of 1-bromo-3-(3-bromopropoxy)-2-methyl-benzene (1.6 g, 5.19 mmol, 1 equiv.), (2S, 6R)-2,6-dimethylpiperazine (593.17 mg, 5.19 mmol, 1 equiv.), K 2 CO 3 (1.44 g, 10.39 mmol, 2 equiv.) in CH3CN (20 mL) was stirred at 60 o C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give compound (3S,5R)-1-[3-(3-bromo-2- methyl-phenoxy)propyl]-3,5-dimethyl-piperazine (1.75 g, 5.1 mmol, 98.7% yield) as a yellow oil. MS (ESI) m/z: 341.1 [M+H] + 1 H NMR (400 MHz, CDCl 3 ) δ = 7.06 (d, J = 7.6 Hz, 1H), 6.91 (t, J = 8.0 Hz, 1H), 6.70 (d, J = 8.0 Hz, 1H), 3.93 (t, J = 6.4 Hz, 2H), 2.86 - 2.84 (m, 2H), 2.74 (dd, J = 2.4, 11.6 Hz, 2H), 2.47 - 2.42 (m, 2H), 2.24 (s, 3H), 1.95 - 1.90 (m, 2H), 1.53 (t, J = 10.8 Hz, 2H), 0.99 (d, J = 6.4 Hz, 6H). Step C. Procedure for preparation of ethyl 2-[(2S,6R)-4-[3-(3-bromo-2-methyl- phenoxy)propyl]-2,6-dimethyl-piperazin-1-yl]acetate A mixture of (3S,5R)-1-[3-(3-bromo-2-methyl-phenoxy)propyl]-3,5-dimethyl- piperazine (1.75 g, 5.13 mmol, 1 equiv.), ethyl 2-bromoacetate (856.33 mg, 5.13 mmol, 567.10 µL, 1 equiv.), K 2 CO 3 (1.42 g, 10.26 mmol, 2.0 equiv.), KI (851.20 mg, 5.13 mmol, 1 equiv.) in CH 3 CN (20 mL) was stirred at 50 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (DCM: MeOH = 1/0 to 10/1) to give compound ethyl 2-[(2S,6R)-4-[3-(3-bromo-2-methyl-phenoxy)propyl]- 2,6-dimethyl-piperazin-1-yl]acetate (2.0 g, 4.7 mmol, 91.3% yield) as a yellow oil. MS (ESI) m/z: 429.0 [M+H] + Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(3S,5R)-4-(2-ethox y-2-oxo-ethyl)-3,5-dimethyl- piperazin-1-yl]propoxy]-2-methyl-phenyl]pyridine-2-carboxyla te Ethyl 2-[(2S,6R)-4-[3-(3-bromo-2-methyl-phenoxy)propyl]-2,6-dimeth yl-piperazin-1- yl]acetate (200 mg, 467.97 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3, 2-dioxaborolan-2-yl)pyridine-2- carboxylate (286.66 mg, 467.97 µmol, 1 equiv.), K2CO3 (129.36 mg, 935.95 µmol, 2.0 equiv.) and Ad 2 nBuP Pd G 3 (cataCXium® A Pd G 3 ) (34.08 mg, 46.80 µmol, 0.1 equiv.) were taken up into a microwave tube in dioxane (5 mL) and H2O (5 mL). The sealed tube was heated at 100 °C for 2 hours under microwave. The reaction mixture was concentrated under reduced pressure to remove solvent to give the crude product. The crude product was purified by reverse-phase HPLC to give compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(3S,5R)-4-(2-ethoxy-2- oxo-ethyl)-3,5-dimethyl- piperazin-1-yl]propoxy]-2-methyl-phenyl]pyridine-2-carboxyla te (172 mg, 206.4 µmol, 44.1% yield) as a yellow oil. MS (ESI) m/z: 833.5 [M+H] + Step E. Procedure for preparation of 2-[(2S,6R)-4-[3-[3-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-2- methyl-phenoxy]propyl]-2,6-dimethyl-piperazin-1-yl]acetic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[(3S,5R)-4-(2-ethoxy-2-oxo-ethyl)-3,5-dimethyl-p iperazin-1-yl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylate (270 mg, 324.11 µmol, 1 equiv.), LiOH•H 2 O (81.61 mg, 1.94 mmol, 6.0 equiv.) in THF (5 mL) and H2O (5 mL) was stirred at 20 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove THF. The mixture was adjusted pH to 2~3 and then extracted with DCM (10 mL × 3). The combined organic layers were concentrated under reduced pressure to give compound 2-[(2S,6R)-4-[3-[3-[6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -2-tert-butoxycarbonyl-3- pyridyl]-2-methyl-phenoxy]propyl]-2,6-dimethyl-piperazin-1-y l]acetic acid (260 mg, 322.9 µmol, 99.6% yield) as a yellow solid. MS (ESI) m/z: 805.4[M+H] + Step F. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(3S,5R)-4-[2-[[3-( 2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]amino]-2-oxo-ethyl]-3,5-dimethyl-piperaz in-1-yl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylate A mixture of 2-[(2S,6R)-4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3, 4-dihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]propyl]-2,6-dimethyl- piperazin-1-yl]acetic acid (100 mg, 124.22 µmol, 1 equiv.), 3-(6-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (32.08 mg, 124.22 µmol, 1 equiv.), HATU (47.23 mg, 124.22 µmol, 1 equiv.), DIEA (16.06 mg, 124.22 µmol, 21.64 µL, 1.0 equiv.) in DMF (3 mL) was stirred at 25 o C for 2 hours. H 2 O (20 mL) was added dropwise into the reaction mixture. The mixture was then filtered and concentrated under reduced pressure to give compound tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[3-[(3S,5R)-4-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-3,5-dimethyl-piperazin- 1-yl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate (110 mg, 105.2 µmol, 84.7% yield) as a pink solid. MS (ESI) m/z: 523.3.3[M/2+H] + Step G. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[(3S,5R)-4-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-3,5-dimethyl-piperazin-1-yl]propoxy ]-2-methyl- phenyl]pyridine-2-carboxylic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[(3S,5R)-4-[2-[[3-(2,6-dioxo-3-piperidyl)-1-meth yl-indazol-6-yl]amino]-2-oxo- ethyl]-3,5-dimethyl-piperazin-1-yl]propoxy]-2-methyl-phenyl] pyridine-2-carboxylate (110 mg, 105.24 µmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL), the mixture was stirred at 40 o C for 4 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give compound 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(3S,5 R)-4-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]-3,5-dim ethyl-piperazin-1-yl]propoxy]- 2-methyl-phenyl]pyridine-2-carboxylic acid (50.3 mg, 49.2 µmol, 46.8% yield, 96.8% purity) as a white solid. MS (ESI) m/z: 989.3 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.89 (s, 1H), 9.78 (s, 1H), 8.15 (s, 1H), 8.05 - 8.02 (m, 2H), 7.79 (d, J = 8.4 Hz, 1H), 7.67 - 7.60 (m, 2H), 7.48 - 7.34 (m, 5H), 7.24 (d, J = 8.8 Hz, 1H), 7.14 - 7.06 (m, 1H), 6.98 (d, J = 8.8 Hz, 1H), 6.89 ( d, J = 8.4 Hz, 1H), 6.65 (d, J = 7.6 Hz, 1H), 4.99 (s, 2H), 4.34 (dd, J = 5.2, 9.6 Hz, 1H), 4.01 (t, J = 5.2 Hz, 2H), 3.98 - 3.85 (m, 6H), 3.03 (t, J = 5.2 Hz, 2H), 2.85 - 2.75 (m, 4H), 2.70 - 2.56 (m, 3H), 2.40 - 2.29 (m, 1H), 2.21 - 2.13 (m, 1H), 2.42 - 2.12 (m, 1H), 1.95 - 1.87 (m, 7H), 1.02 (d, J = 5.6 Hz, 6H). Example 19. Preparation of Compound 119 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4-[3-[4-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]piperazin-1-yl]propoxy]- 2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 4-[3-(4-bromo-3-methyl-phenoxy) propyl] piperazine-1-carboxylate A mixture of 4-bromo-3-methyl-phenol (382.75 mg, 2.05 mmol, 1 equiv.), tert-butyl 4-(3- hydroxypropyl)piperazine-1-carboxylate (500 mg, 2.05 mmol, 1 equiv.), PPh 3 (697.77 mg, 2.66 mmol, 1.3 equiv.) in THF (9 mL) was degassed and purged with N2 three times, and then DIAD (496.56 mg, 2.46 mmol, 477.46 µL, 1.2 equiv.) was added dropwise at 0 °C. The mixture was stirred at 25 °C for 12 hours under N 2 atmosphere. The reaction mixture was diluted with H 2 O (5 mL × 3) and extracted with ethyl acetate (10 mL × 3). The combined organic layers were washed with brine (5 mL × 3), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~47% ethyl acetate/petroleum ether) to give compound tert-butyl 4-[3-(4-bromo-3- methyl-phenoxy) propyl] piperazine-1-carboxylate (450 mg, 870.9 µmol, 42.5% yield, 80% purity) as a yellow oil. MS (ESI) m/z: 414.9 [M+H] + . 1 H NMR (400 MHz, CD 3 OD) δ = 7.38 (d, J = 8.4 Hz, 1H), 6.85 (d, J = 3.2 Hz, 1H), 6.66 (dd, J = 3.2, 3.2 Hz, 1H), 4.00 (t, J = 6.4 Hz, 2H), 3.43 (d, J = 4.4 Hz, 4H), 2.55 (t, J = 7.2 Hz, 2H), 2.48 - 2.40 (m, 4H), 2.33 (s, 3H), 2.00 - 1.92 (m, 2H), 1.46 (s, 9H). Step B. Procedure for preparation of 1-[3-(4-bromo-3-methyl-phenoxy) propyl] piperazine To a solution of tert-butyl 4-[3-(4-bromo-3-methyl-phenoxy) propyl] piperazine-1-carboxylate (450 mg, 1.09 mmol, 1 equiv.) was added HCl/dioxane (4 M, 4.50 mL, 16.53 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give compound 1-[3-(4-bromo-3-methyl-phenoxy) propyl] piperazine (400 mg, crude) as a white solid. MS (ESI) m/z: 315.0 [M+H] + . Step C. Procedure for preparation of ethyl 2-[4-[3-(4-bromo-3-methyl- phenoxy)propyl]piperazin-1-yl]acetate To a solution of 1-[3-(4-bromo-3-methyl-phenoxy) propyl] piperazine (350 mg, 1.00 mmol, 1 eq, HCl) in CH 3 CN (3 mL) was added K 2 CO 3 (414.98 mg, 3.00 mmol, 3 equiv.) and ethyl 2- bromoacetate (167.15 mg, 1.00 mmol, 110.69 µL, 1 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give the crude product. The residue was purified by prep-TLC (SiO 2 , DCM: MeOH = 30: 1) to give compound ethyl 2-[4-[3-(4-bromo-3-methyl-phenoxy)propyl]piperazin-1-yl]acet ate (230 mg, 518.3 µmol, 51.7% yield, 90% purity) as a yellow oil. MS (ESI) m/z: 401.1 [M+H] + Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[3-[4-(2-ethoxy-2-oxo- ethyl)piperazin-1- yl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine- 2-carboxylate (260 mg, 424.46 µmol, 1 equiv.), ethyl 2-[4-[3-(4-bromo-3-methyl-phenoxy)propyl]piperazin-1-yl]acet ate (225.99 mg, 509.35 µmol, 90% purity, 1.2 equiv.) in dioxane (3 mL) was added KF (1.5 M, 848.92 µL, 3 equiv.) and Ad 2 nBuP Pd G 3 (cataCXium® A Pd G 3 ) (30.91 mg, 42.45 µmol, 0.1 equiv.). After addition, the mixture was degassed and purged with N2 three times, and then the mixture was stirred at 100 °C for 2 hours under N2 atmosphere. The reaction mixture was diluted with H 2 O 6 mL and extracted with ethyl acetate (5 mL × 4). The combined organic layers were washed with brine (5 mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~9% dichloromethane/methanol) to give compound tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[4-[3-[4-(2-ethoxy-2-oxo- ethyl)piperazin-1-yl]propoxy]-2-methyl-phenyl]pyridine-2-car boxylate (220 mg, 218.6 µmol, 51.5% yield, 80% purity) as a yellow oil. MS (ESI) m/z: 805.3 [M+H] + 1 H NMR (400 MHz, CDCl3) δ = 7.85 (d, J = 7.6 Hz, 1H), 7.56 (dd, J = 5.2, 4.0 Hz, 2H), 7.38 - 7.34 (m, 3H), 7.32 - 7.29 (m, 1H), 6.96 (d, J = 8.4 Hz, 1H), 6.88 (d, J = 8.8 Hz, 1H), 6.78 (d, J = 2.8 Hz, 1H), 6.70 (dd, J = 2.4, 2.4 Hz, 1H), 6.61 (dd, J = 3.2, 2.8 Hz, 1H), 4.11 - 4.07 (m, 2H), 4.03 - 3.98 (m, 2H), 3.23 (s, 3H), 3.06 (t, J = 5.6 Hz, 2H), 2.72 - 2.57 (m, 12H), 2.36 (s, 2H), 2.09 (s, 3H), 1.69 - 1.54 (m, 4H), 1.18 (s, 9H) Step E. Procedure for preparation of 2-[4-[3-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyr idyl]-3-methyl- phenoxy]propyl]piperazin-1-yl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[4-[3-[4-(2-ethoxy-2-oxo-ethyl)piperazin-1-yl]propox y]-2-methyl-phenyl]pyridine-2- carboxylate (220 mg, 218.64 µmol, 80% purity, 1 equiv.) in THF (2 mL) was added LiOH (15.71 mg, 655.91 µmol, 3 equiv.) and H2O (0.5 mL) at 25 °C. The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove THF. The aqueous phase was adjusted to pH = 4~5 with 1 M HCl. The reaction mixture was filtered. The solid was diluted in ethyl acetate. The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to give compound 2-[4-[3-[4-[6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -2-tert-butoxycarbonyl-3- pyridyl]-3-methyl-phenoxy]propyl]piperazin-1-yl]acetic acid (160 mg, crude) as a yellow oil. MS (ESI) m/z: 777.2 [M+H] + Step F. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[3-[4-[2-[[3-(2,6-diox o-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]piperazin-1-yl]propoxy]-2-me thyl-phenyl]pyridine-2- carboxylate To a solution of 2-[4-[3-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-3-methyl- phenoxy]propyl]piperazin-1- yl]acetic acid (80 mg, 102.97 µmol, 1 equiv.) in DMF (1 mL) was added HATU (46.98 mg, 123.56 µmol, 1.2 equiv.), DIPEA (39.92 mg, 308.90 µmol, 53.80 µL, 3 equiv.), and 3-(6- amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (31.91 mg, 123.56 µmol, 1.2 equiv.). The mixture was stirred at 25 °C for 3 hours. The reaction mixture was quenched by addition H2O (2mL). The reaction mixture was filtered and washed with water (5 mL). The solid was diluted in DCM (10 mL). The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[3-[4-[2-[[3-(2,6-diox o-3-piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]piperazin-1-yl]propoxy]-2-methyl-phe nyl]pyridine-2-carboxylate (90 mg, crude) as a yellow solid. MS (ESI) m/z: 1017.4 [M+H] + Step G. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[4-[3-[4-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]piperazin-1-yl]propoxy]-2-methyl-pheny l]pyridine-2-carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[4-[3-[4-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-ind azol-6-yl]amino]-2-oxo- ethyl]piperazin-1-yl]propoxy]-2-methyl-phenyl]pyridine-2-car boxylate (90 mg, 88.48 µmol, 1 equiv.) in DCM (0.5 mL) was added TFA (770.00 mg, 6.75 mmol, 500.00 µL, 76.32 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give compound 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4-[3-[4-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]piperazin-1-yl]propoxy]- 2-methyl-phenyl]pyridine-2-carboxylic acid (12.5 mg, 12.2 µmol, 13.8% yield, 94.3% purity) as a yellow solid. MS (ESI) m/z: 961.1 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.86 (s, 1H), 9.85 (s, 1H), 8.05 – 8.01 (m, 2H), 7.79 (d, J = 9.2 Hz, 1H), 7.63 (d, J = 8.8 Hz, 2H), 7.50 - 7.33 (m, 6H), 7.22 - 7.18 (m, 1H), 6.93 (d, J = 8.4 Hz, 2H), 6.78 (s, 1H), 6.73 - 6.68 (m, 1H), 4.97 (s, 2H), 4.36 - 4.29 (m, 1H), 3.98 (s, 2H), 3.92 - 3.91 (m, 5H), 3.61 - 3.56 (m, 2H), 3.16 (s, 3H), 3.05 - 2.99 (m, 4H), 2.97 - 2.94 (m, 1H), 2.61 (d, J = 6.4 Hz, 4H), 2.18 (s, 3H), 2.03 (s, 4H), 1.89 - 1.84 (m, 2H). Example 20. Preparation of Compound 120 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[4-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-1-piperidyl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of ethyl 2-[1-[3-(3-bromo-2-methyl-phenoxy)propyl]- 4-piperidyl]acetate A mixture of 1-bromo-3-(3-bromopropoxy)-2-methyl-benzene (800 mg, 2.60 mmol, 1 equiv.), ethyl 2-(4-piperidyl)acetate (444.76 mg, 2.60 mmol, 1 equiv.), K 2 CO 3 (717.93 mg, 5.19 mmol, 2.0 equiv.) in ACN (20 mL), and then the mixture was stirred at 50 °C for 4 hours. The reaction mixture was concentrated under reduced pressure to remove CH3CN to give a residue. The residue was purified by column chromatography (SiO 2 , DCM: MeOH =1/0 to 10/1) to give compound ethyl 2-[1-[3-(3-bromo-2-methyl-phenoxy)propyl]-4-piperidyl]acetat e (650 mg, 1.6 mmol, 62.8% yield) was obtained as a colorless oil. MS (ESI) m/z: 398.0 [M+H] + 1 H NMR (400 MHz, CDCl 3 ) δ = 7.16 (d, J = 8.0 Hz, 1H), 7.00 (t, J = 8.0 Hz, 1H), 6.79 (d, J = 8.4 Hz, 1H), 4.15 (q, J = 7.2 Hz, 2H), 4.01 (t, J = 6.0 Hz, 2H), 2.95 ( d, J = 11.6 Hz, 2H), 2.54 ( t, J = 7.2 Hz, 2H), 2.32 (s, 3H), 2.25 (d, J = 7.2 Hz, 2H), 2.05 - 1.99 (m, 4H), 1.87 - 1.80 (m, 1H), 1.75 ( d, J = 14.0 Hz, 2H), 1.41 - 1.32 (m, 2H), 1.28 (t, J = 7.2 Hz, 3H). Step B. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[4-(2-ethoxy-2-oxo- ethyl)-1- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate Ethyl 2-[1-[3-(3-bromo-2-methyl-phenoxy)propyl]-4-piperidyl]acetat e (400 mg, 1.00 mmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-2-carb oxylate (430.58 mg, 702.93 µmol, 0.7 equiv.), K 2 CO 3 (277.57 mg, 2.01 mmol, 2.0 equiv.) and Ad 2 nBuP Pd G3(cataCXium® A Pd G3) (73.13 mg, 100.42 µmol, 0.1 equiv.) were taken up into a microwave tube in dioxane (5 mL) and H2O (2 mL). The sealed tube was heated at 100 °C for 2 hours under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (neutral condition) to give compound tert- butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[4-(2- ethoxy-2-oxo-ethyl)-1-piperidyl]propoxy]-2-methyl-phenyl]pyr idine-2-carboxylate (300 mg, 373.1 µmol, 37.1% yield) as a yellow solid. MS (ESI) m/z: 804.7 [M+H] + Step C. Procedure for preparation of 2-[1-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyr idyl]-2-methyl- phenoxy]propyl]-4-piperidyl]acetic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[4-(2-ethoxy-2-oxo-ethyl)-1-piperidyl]propoxy]-2 -methyl-phenyl]pyridine-2- carboxylate (300 mg, 373.1 µmol, 1 equiv.), LiOH•H 2 O (93.94 mg, 2.24 mmol, 6 equiv.) in THF (6 mL) and H2O (3 mL) was stirred at 25 o C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove THF, and then adjusted pH to 2~3 with HCl (3 M). The mixture was then filtered and concentrated under reduced pressure to give compound 2-[1-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H-isoquinolin-2-yl]-2-tert- butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]propyl]-4-piperid yl]acetic acid (280 mg, 360.8 µmol, 96.7% yield) as a yellow solid. MS (ESI) m/z: 776.2 [M+H] + Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[4-[2-[[3-(2,6-diox o-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-1-piperidyl]propoxy]-2-meth yl-phenyl]pyridine-2- carboxylate A mixture of 2-[1-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H-isoquinolin- 2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]prop yl]-4-piperidyl]acetic acid (100 mg, 128.87 µmol, 1 equiv.), 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (33.28 mg, 128.87 µmol, 1 equiv.), HATU (49.00 mg, 128.87 µmol, 1 equiv.), DIEA (16.66 mg, 128.87 µmol, 22.45 µL, 1 equiv.) in DMF (5 mL) was stirred at 25 o C for 2 hours. The reaction mixture was diluted with water 10 mL, filtered to give a the crude product tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[4-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-1-piperidyl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylate (100 mg, 98.4 µmol, 76.4% yield) as a yellow solid. MS (ESI) m/z: 960.3 [M-56+H] + Step E. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[4-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-1-piperidyl]propoxy]-2-methyl-phenyl] pyridine-2-carboxylic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[4-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indaz ol-6-yl]amino]-2-oxo-ethyl]-1- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate (100 mg, 98.40 µmol, 1 equiv.) in DCM (1 mL) and TFA (1 mL) was stirred at 40 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a crude product. The crude product was purified by reverse-phase HPLC to give compound 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[4-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-1-piperidyl]propoxy]-2-methyl-phenyl] pyridine-2-carboxylic acid (35.1 mg, 34.8 µmol, 35.3% yield, 95% purity) as a white solid. MS (ESI) m/z: 960.3 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 10.86 (s, 1H), 10.08 (s, 1H), 8.18 (s, 1H), 8.09 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.67 - 7.59 (m, 2H), 7.46 - 7.34 (m, 5H), 7.14 - 7.05 (m, 2H), 6.94 (d, J = 8.4 Hz, 1H), 6.87 ( d, J = 8.4 Hz, 1H), 6.65 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.31 ( dd, J = 5.2, 9.6 Hz, 1H), 3.99 ( t, J = 5.6 Hz, 3H), 3.95 - 3.88 (m, 6H), 3.04 ( d, J = 5.6 Hz, 2H), 2.93 ( d, J = 8.4 Hz, 3H), 2.71 - 2.56 (m, 4H), 2.35 - 2.32 (m, 1H), 2.29 ( d, J = 6.4 Hz, 2H), 2.18 ( dd, J = 5.2, 13.6 Hz, 1H), 2.06 ( t, J = 11.2 Hz, 2H), 1.91 (s, 3H), 1.69 ( d, J = 12.4 Hz, 2H), 1.36 - 1.23 (m, 2H). Example 21. Preparation of Compound 127 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((1R,5S)-8- (2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)am ino)-2-oxoethyl)-3,8- diazabicyclo[3.2.1]octan-3-yl)propoxy)-2-methylphenyl)picoli nic acid Step A. Procedure for preparation of (1R, 5S)-tert-butyl 3-(3-(3-bromo-2- methylphenoxy)propyl)-3,8-diazabicyclo[3.2.1]octane-8-carbox ylate To a solution of 1-bromo-3-(3-bromopropoxy)-2-methylbenzene (1 g, 3.25 mmol, 1 equiv.) and (1R,5S)-tert-butyl 3,8-diazabicyclo[3.2.1]octane-8-carboxylate (1.38 g, 6.49 mmol, 2 equiv.) in CH3CN (10 mL) was added DIPEA (1.26 g, 9.74 mmol, 1.70 mL, 3 equiv.). The mixture was stirred at 60 °C for 12 hours. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~16% ethyl acetate/petroleum ether) to give compound (1R, 5S)-tert-butyl 3-(3-(3-bromo-2- methylphenoxy)propyl)-3,8-diazabicyclo[3.2.1]octane-8-carbox ylate (1.5 g, 3.4 mmol, 50.0% yield) as a colorless oil. MS (ESI) m/z: 441.1 [M+H] + . Step B. Procedure for preparation of (1R, 5S)-3-(3-(3-bromo-2-methylphenoxy)propyl)- 3,8-diazabicyclo[3.2.1]octane A mixture of (1R, 5S)-tert-butyl 3-(3-(3-bromo-2-methylphenoxy)propyl)-3,8- diazabicyclo[3.2.1]octane-8-carboxylate (1.5 g, 3.41 mmol, 1 equiv.), HCl/1,4-dioxane (4 M, 10 mL, 11.72 equiv.) in DCM (5 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 25 °C for 2 hours under N 2 atmosphere. The mixture was concentrated under reduced pressure to give compound (1R, 5S)-3-(3-(3-bromo-2- methylphenoxy)propyl)-3,8-diazabicyclo[3.2.1]octane (2 g, crude) as a white solid. MS (ESI) m/z: 340.8 [M+H] + . Step C. Procedure for preparation of ethyl 2-((1R, 5S)-3-(3-(3-bromo-2- methylphenoxy)propyl)-3,8-diazabicyclo[3.2.1]octan-8-yl)acet ate A mixture of (1R, 5S)-3-(3-(3-bromo-2-methylphenoxy)propyl)-3,8-diazabicyclo[3 .2.1]octane (1 g, 2.95 mmol, 1 equiv.), ethyl 2-bromoacetate (984.47 mg, 5.90 mmol, 651.97 µL, 2 equiv.) and DIEA (1.14 g, 8.84 mmol, 1.54 mL, 3 equiv.) in CH3CN (30 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 60 °C for 16 hours under N2 atmosphere. The reaction mixture was diluted with water 50 mL and extracted with EtOAc 50 mL × 3. The combined organic layers were washed with brine 50 mL × 3, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by reverse-phase HPLC (with 0.1% HCOOH) to give compound ethyl 2-((1R, 5S)-3- (3-(3-bromo-2-methylphenoxy)propyl)-3,8-diazabicyclo[3.2.1]o ctan-8-yl)acetate (800 mg, 1.84 mmol, 62.5% yield, 98% purity) as a yellow oil. MS (ESI) m/z: 426.9 [M+H] + . Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((1R,5S)-8-(2-ethoxy-2- oxoethyl)-3,8- diazabicyclo[3.2.1]octan-3-yl)propoxy)-2-methylphenyl)picoli nate Ethyl 2-((1R, 5S)-3-(3-(3-bromo-2-methylphenoxy)propyl)-3,8-diazabicyclo[3 .2.1]octan-8- yl)acetate (150 mg, 352.64 µmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3, 2-dioxaborolan-2-yl)picolinate (216.01 mg, 352.64 µmol, 1 equiv.), KF (1.5 M, 705.29 µL, 3 equiv.) and Ad 2 nBuP Pd G 3 (cataCXium® A Pd G 3 ) (25.68 mg, 35.26 µmol, 0.1 equiv.) were taken up into a microwave tube in 1,4-dioxane (3.5 mL).The sealed tube was heated at 100 °C for 60 minutes under microwave. The reaction mixture was diluted with water 20 mL and extracted with DCM 25 mL × 3. The combined organic layers were washed with brine 15 mL × 3, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH = 10: 1) to give compound tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-((1R,5S)-8-(2- ethoxy-2-oxoethyl)-3,8-diazabicyclo[3.2.1]octan-3-yl)propoxy )-2-methylphenyl)picolinate (190 mg, 225.84 µmol, 64.04% yield, 98.78% purity) as a yellow oil. MS (ESI) m/z: 831.7 [M+H] + . Step E. Procedure for preparation of 2-((1R,5S)-3-(3-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)propyl)-3,8-diazabicyclo[3.2.1]octan-8-yl)acet ic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-((1R,5S)-8-(2-ethoxy-2-oxoethyl)-3,8-diazabicycl o[3.2.1]octan-3-yl)propoxy)-2- methylphenyl)picolinate (190 mg, 228.63 µmol, 1 equiv.) and LiOH·H 2 O (28.78 mg, 685.89 µmol, 3 equiv.) in THF (1.5 mL) and H2O (0.5 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 1 hour under N2 atmosphere. The mixture was concentrated under reduced pressure to give compound 2-((1R,5S)-3-(3-(3-(6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)propyl)-3,8-dia zabicyclo[3.2.1]octan-8- yl)acetic acid (150 mg, 186.8 µmol, 81.7% yield) as a white solid . MS (ESI) m/z: 803.5 [M+H] + . Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((1R,5S)-8-(2-((3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)-3,8-diazabicyclo[3 .2.1]octan-3-yl)propoxy)- 2-methylphenyl)picolinate To a solution of 2-((1R,5S)-3-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4 - dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)propyl)-3,8-diazabicyclo[3.2.1]octan-8-yl)acet ic acid (100 mg, 124.54 µmol, 1 equiv.) in DMF (2 mL) was added DIEA (48.29 mg, 373.61 µmol, 65.07 uL, 3 equiv.) and HATU (56.82 mg, 149.44 µmol, 1.2 equiv.), 3-(6-amino-1-methyl-1H-indazol-3-yl)piperidine- 2,6-dione (35.38 mg, 136.99 µmol, 1.1 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction was diluted with water (30 mL) and extracted with DCM (20mL × 2). The combined organic layers were washed with brine (30 mL × 3), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH = 10: 1, Rf =0.5) to give compound tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((1R,5 S)-8-(2-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2-oxoet hyl)-3,8- diazabicyclo[3.2.1]octan-3-yl)propoxy)-2-methylphenyl)picoli nate (120 mg, 115.0 µmol, 92.3% yield) as a yellow solid. MS (ESI) m/z: 1043.1 [M+H] + . Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((1R,5S)-8-(2-((3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)-3,8-diazabicyclo[3 .2.1]octan-3-yl)propoxy)- 2-methylphenyl)picolinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-((1R,5S)-8-(2-((3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)amino)-2- oxoethyl)-3,8-diazabicyclo[3.2.1]octan-3-yl)propoxy)-2-methy lphenyl)picolinate (100 mg, 95.86 µmol, 1 equiv.) in TFA (0.5 mL) and DCM (0.5 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 1 hour under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give compound 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((1R,5S)-8-(2-((3-( 2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)-3,8-diazabicyclo[3 .2.1]octan-3-yl)propoxy)-2- methylphenyl)picolinic acid (28.6 mg, 28.9 µmol, 30.1% yield, 99.5% purity) as a white solid. MS (ESI) m/z: 494.3 [M/2+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.86 (s, 1H), 9.93 (s, 1H), 8.07 - 8.00 (m, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.67 - 7.59 (m, 2H), 7.49 - 7.42 (m, 3H), 7.39 - 7.32 (m, 2H), 7.20 (dd, J = 8.8, 2.0 Hz, 1H), 7.14 - 7.07 (m, 1H), 6.96 (d, J = 8.8 Hz, 1H), 6.89 (d, J = 8.0 Hz, 1H), 6.64 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.33 (q, J = 4.8 Hz, 1H), 4.02 (t, J = 6.0 Hz, 2H), 3.94 - 3.91 (m, 4H), 3.20 (s, 4H), 3.12 (s, 3H), 3.03 (t, J = 5.6 Hz, 2H), 2.68 - 2.61 (m, 4H), 2.42 - 2.23 (m, 4H), 2.21 - 2.14 (m, 1H), 1.90 (s, 3H), 1.87 (d, J = 6.4 Hz, 1H), 1.85 - 1.79 (m, 2H), 1.75 - 1.68 (m, 2H). Example 22. Preparation of Compound 131a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[(2R,6S)-4- [2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]- 2-oxo-ethyl]-2,6-dimethyl- piperazin-1-yl]propoxy]-2-methyl-phenyl]pyridine-2-carboxyli c acid Step A. Procedure for preparation of tert-butyl (3R,5S)-4-(3-iodopropyl)-3,5-dimethyl- piperazine-1-carboxylate To a solution of tert-butyl (3R,5S)-3,5-dimethylpiperazine-1-carboxylate (2.0 g, 9.33 mmol, 1 equiv.) in CH3CN (5 mL) was added 1,3-diiodopropane (2.76 g, 9.33 mmol, 1.08 mL, 1 equiv.) and K2CO3 (1.29 g, 9.33 mmol, 1 equiv.). The mixture was stirred at 60 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove solvent to give a residue. The residue was purified by column chromatography (SiO2, DCM: MeOH = 10:1) to give the compound tert-butyl (3R,5S)-4-(3-iodopropyl)-3,5-dimethyl-piperazine-1-carboxyla te (1.0 g, 2.62 mmol, 28.0% yield) as a yellow oil. 1 H NMR (400 MHz, CDCl3) δ = 5.23 – 5.07 (m, 1H), 3.96 – 3.58 (m, 2H), 3.39 ( s, 1H), 3.05 (t, J = 6.8 Hz, 1H), 2.81 – 2.71 (m, 1H), 2.64 – 2.28 (m, 4H), 1.88 – 1.78 (m, 1H), 1.52 ( d, J = 14.8 Hz, 1H), 1.39 (s, 9H), 1.02 ( d, J = 5.2 Hz, 6H). Step B. Procedure for preparation of tert-butyl (3R,5S)-4-[3-(3-bromo-2-methyl- phenoxy)propyl]-3,5-dimethyl-piperazine-1-carboxylate To a solution of tert-butyl (3R,5S)-4-(3-iodopropyl)-3,5-dimethyl-piperazine-1-carboxyla te (1 g, 2.62 mmol, 1 equiv.) in CH 3 CN (15 mL) was added K 2 CO 3 (1.08 g, 7.85 mmol, 3 equiv.) and 3-bromo-2-methyl-phenol (489.26 mg, 2.62 mmol, 1 equiv.). The mixture was stirred at 60 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , DCM: MeOH = 1/0 to 10/1) to give the compound tert-butyl (3R,5S)-4-[3-(3-bromo-2-methyl- phenoxy)propyl]-3,5-dimethyl-piperazine-1-carboxylate (540 mg, 1.22 mmol, 46.7% yield) as a yellow oil. MS (ESI) m/z: 443.3 [M+H] + Step C. Procedure for preparation of (2R,6S)-1-[3-(3-bromo-2-methyl-phenoxy)propyl]- 2,6-dimethyl-piperazine To a solution of tert-butyl (3R,5S)-4-[3-(3-bromo-2-methyl-phenoxy)propyl]-3,5-dimethyl- piperazine-1-carboxylate (500 mg, 1.13 mmol, 1 equiv.) in EtOAc (5 mL) was added HCl/EtOAc (3 M, 1.13 mL, 3 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove EtOAc to give the crude product (2R,6S)-1-[3-(3-bromo-2-methyl-phenoxy)propyl]-2,6-dimethyl- piperazine (420 mg, 1.11 mmol, 98.1% yield) as a white solid. MS (ESI) m/z: 342.8 [M+H] + Step D. Procedure for preparation of ethyl 2-[(3R,5S)-4-[3-(3-bromo-2-methyl- phenoxy)propyl]-3,5-dimethyl-piperazin-1-yl]acetate To a solution of (2R,6S)-1-[3-(3-bromo-2-methyl-phenoxy)propyl]-2,6-dimethyl- piperazine (420 mg, 1.23 mmol, 1 equiv.) in CH 3 CN (10 mL) was added K 2 CO 3 (510.25 mg, 3.69 mmol, 3 equiv.) and ethyl 2-bromoacetate (205.52 mg, 1.23 mmol, 136.10 µL, 1 equiv.). The mixture was stirred at 60 °C for 12 hours. The reaction mixture was filtered, washed by EtOAc (10 mL), and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2, DCM: MeOH = 1/0 to 10/1) to give the compound ethyl 2- [(3R,5S)-4-[3-(3-bromo-2-methyl-phenoxy)propyl]-3,5-dimethyl -piperazin-1-yl]acetate (350 mg, 818.9 µmol, 66.5% yield) as a pink solid. MS (ESI) m/z: 429.3 [M+H] + Step E. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(2R,6S)-4-(2-ethox y-2-oxo-ethyl)-2,6-dimethyl- piperazin-1-yl]propoxy]-2-methyl-phenyl]pyridine-2-carboxyla te To a solution of ethyl 2-[(3R,5S)-4-[3-(3-bromo-2-methyl-phenoxy)propyl]-3,5-dimeth yl- piperazin-1-yl]acetate (250 mg, 584.97 µmol, 1 equiv.) taken up into a microwave tube in dioxane (5 mL) and H2O (1 mL) was added tert-butyl 6-[8-(3H-indol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)pyridine-2- carboxylate (347.77 mg, 584.97 µmol, 1 equiv.), Ad2nBuP Pd G3(cataCXium® A Pd G3 ) (42.60 mg, 58.50 µmol, 0.1 equiv.), and K2CO3 (242.54 mg, 1.75 mmol, 3 equiv.). The reaction mixture was degassed and purged with N 2 three times and was then stirred at 100 °C for 1 hour under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give the crude product. The crude product was purified by reverse-phase HPLC to give the compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[(2R,6S)-4-(2-ethoxy-2-oxo-ethyl)-2,6-dimethyl-p iperazin-1-yl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylate (100 mg, 120.04 µmol, 20.52% yield) as a yellow oil. 1 H NMR (400 MHz, CDCl3) δ = 8.23 (s, 2H), 7.78 (d, J = 7.6 Hz, 1H), 7.62 - 7.56 (m, 2H), 7.41 - 7.24 (m, 6H), 7.21 - 7.16 (m, 2H), 7.04 (t, J = 8.0 Hz, 1H), 6.83 - 6.78 (m, 1H), 6.73 - 6.64 (m, 2H), 5.04 - 4.89 (m, 2H), 4.12 (q, J = 7.2 Hz, 2H), 4.06 - 3.90 (m, 4H), 3.40 - 3.20 (m, 4H), 3.19 - 3.16 (m, 2H), 3.03 - 2.96 (m, 2H), 2.87 (d, J = 11.2 Hz, 2H), 2.69 - 2.60 (m, 2H), 2.08 - 1.97 (m, 2H), 1.92 - 1.88 (m, 3H), 1.30 ( s, 6H), 1.23 - 1.18 (m, 3H), 1.09 (s, 9H). Step F. Procedure for preparation of 2-[(3R,5S)-4-[3-[3-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-2- methyl-phenoxy]propyl]-3,5-dimethyl-piperazin-1-yl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[3-[(2R,6S)-4-(2-ethoxy-2-oxo-ethyl)-2,6-dimethyl -piperazin-1-yl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylate (100 mg, 120.04 µmol, 1 equiv.) in THF (3 mL) and H2O (1 mL) was added LiOH.H2O (50.4 mg, 12.00 µmol, 10 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The resulting residue was diluted with H 2 O (10 mL) and adjusted to pH = 3~4. The mixture was then extracted with EtOAc 10 mL (10 mL × 3), and the combined organic layers were washed with brine (10 mL), filtered and concentrated under reduced pressure to give the crude product 2-[(3R,5S)-4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3, 4-dihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]propyl]-3,5-dimethyl- piperazin-1-yl]acetic acid (84 mg, 104.3 µmol, 86.9% yield, 100% purity) as a yellow solid. MS (ESI) m/z: 805.2 [M+H] + Step G. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(2R,6S)-4-[2-[[3-( 2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]amino]-2-oxo-ethyl]-2,6-dimethyl-piperaz in-1-yl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylate To a solution of 2-[(3R,5S)-4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3, 4-dihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]propyl]-3,5-dimethyl- piperazin-1-yl]acetic acid (84 mg, 104.35 µmol, 1 equiv.) in DMF (1 mL) was added 3-(6- amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (26.95 mg, 104.35 µmol, 1 equiv.), HATU (39.68 mg, 104.35 µmol, 1 equiv.) and DIEA (13.49 mg, 104.35 µmol, 18.18 µL, 1 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was diluted with water (10 mL) and extracted with EtOAc (10 mL). The combined extracts were filtered and concentrated under reduced pressure to give the crude product tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(2R,6 S)-4-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]-2,6-dim ethyl-piperazin-1-yl]propoxy]- 2-methyl-phenyl]pyridine-2-carboxylate (80 mg, 67.3 µmol, 64.5% yield, 88% purity) as a yellow solid. Step H. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[(2R,6S)-4-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-2,6-dimethyl-piperazin-1-yl]propoxy ]-2-methyl- phenyl]pyridine-2-carboxylic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[(2R,6S)-4-[2-[[3-(2,6-dioxo-3-piperidyl)-1-meth yl-indazol-6-yl]amino]-2-oxo- ethyl]-2,6-dimethyl-piperazin-1-yl]propoxy]-2-methyl-phenyl] pyridine-2-carboxylate (70 mg, 66.97 µmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL) was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give the crude product. The crude product was purified by reverse-phase HPLC to give the compound 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(2R,6 S)-4-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]-2,6-dim ethyl-piperazin-1-yl]propoxy]- 2-methyl-phenyl]pyridine-2-carboxylic acid (41.2 mg, 39.6 µmol, 59.2% yield, 94.9% purity) as a white solid. MS (ESI) m/z: 989.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.90 (s, 1H), 9.90 - 9.85 (m, 1H), 8.16 - 8.14 (m, 1H), 8.08 - 8.01 (m, 2H), 7.82 - 7.77 (m, 1H), 7.63 (d, J = 8.8 Hz, 2H), 7.50 - 7.42 (m, 3H), 7.41 - 7.33 (m, 2H), 7.22 - 7.17 (m, 1H), 7.14 - 7.07 (m, 1H), 7.00 - 6.94 (m, 1H), 6.92 - 6.84 (m, 1H), 6.64 (d, J = 7.6 Hz, 1H), 5.02 - 4.95 (m, 2H), 4.36 - 4.30 (m, 1H), 4.03 - 3.96 (m, 2H), 3.95 - 3.89 (m, 5H), 3.12 (s, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.96 - 2.88 (m, 2H), 2.78 (d, J = 9.2 Hz, 4H), 2.64 (dt, J = 5.6, 10.4 Hz, 2H), 2.41 - 2.30 (m, 1H), 2.22 - 2.13 (m, 1H), 2.07 - 1.97 (m, 2H), 1.94 - 1.90 (m, 3H), 1.90 - 1.80 (m, 2H), 1.07 - 0.99 (m, 6H). Example 23. Preparation of Compound 135 N-(1,3-benzothiazol-2-yl)-2-[5-(1-benzyl-5-methyl-pyrazol-4- yl)-6-[[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]carbamoy l]-2-pyridyl]-3,4-dihydro-1H- isoquinoline-8-carboxamide Step A. Procedure for preparation of 1-benzyl-4-iodo-pyrazole To a solution of bromomethylbenzene (13.6 g, 79.6 mmol, 9.45 mL, 1.05 equiv.) and 4-iodo- 1H-pyrazole (14.7 g, 75.8 mmol, 1 equiv.) in acetone (200 mL) was added K 2 CO 3 (26.2 g, 189 mmol, 2.5 equiv.). The mixture was stirred at 70 °C for 4 h. After completion, the reaction was filtered and concentrated under reduced pressure to give the crude product. The crude product was purified by flash silica gel chromatography (eluent of 0~10% ethyl acetate/petroleum ether).1-Benzyl-4-iodo-pyrazole (20.4 g, 68.2 mmol, 90.0% yield, 95% purity) was obtained as an off-white solid. 1 H NMR (400 MHz, CD 3 Cl) δ = 7.46 (s, 1 H), 7.31 (s, 1 H), 7.22 - 7.28 (m, 3 H), 7.14 (dd, J = 8.0, 1.75 Hz, 2 H), 5.21 (s, 2 H) Step B. Procedure for preparation of 1-benzyl-4-iodo-5-methyl-pyrazole To a solution of 1-benzyl-4-iodo-pyrazole (10.0 g, 35.2 mmol, 1 equiv.) in THF (100 mL) was added LDA (2 M, 21.1 mL, 1.2 equiv.) at -70 °C, and then the mixture was stirred at -70 °C for 30 min. MeI (5.50 g, 38.7 mmol, 2.41 mL, 1.1 equiv.) was added dropwise at -70 °C. Then the mixture was stirred at 25 °C for 5 h. After completion, the reaction was quenched with a saturated solution of NH4Cl (50 ml) and extracted with ethyl acetate (70 ml × 3). The combined organic layers were dried over Na2SO4, filtered, and concentrated under reduced pressure to give 1-benzyl-4-iodo-5-methyl-pyrazole (9.6 g, crude) as a white solid. MS (ESI) m/z: 298.7 [M+H] + . Step C. Procedure for preparation of 1-benzyl-5-methyl-4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyrazole To a solution of 1-benzyl-4-iodo-5-methyl-pyrazole (8.50 g, 28.5 mmol, 1 equiv.) and 4,4,5,5- tetramethyl-1,3,2-dioxaborolane (14.6 g, 114 mmol, 16.6 mL, 4 equiv.) in CH3CN (100 mL) was added TEA (11.5 g, 114 mmol, 15.9 mL, 4 equiv.) and cyclopentyl(diphenyl)phosphane;dichloromethane;dichloropalla dium;iron (2.33 g, 2.85 mmol, 0.1 equiv.). The mixture was stirred at 80 °C for 4 h under N2 atmosphere. The reaction mixture was partitioned between water (100 mL) and ethyl acetate (200 mL). The organic phase was separated, washed with ethyl acetate (200 mL × 2), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0 ~ 20% ethyl acetate / petroleum ether) to give 1-benzyl-5- methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazo le (7.00 g, 21.1 mmol, 74.1% yield, 90% purity) as a white solid. 1 H NMR (400 MHz, CDCl3) δ = 7.67 (s, 1 H), 7.13 - 7.27 (m, 3 H), 7.02 (d, J =7.2 Hz, 2 H), 5.22 (s, 2 H), 2.30 (s, 3 H), 1.15 - 1.23 (m, 12 H) Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-(1-benzyl-5-methyl-pyrazo l-4-yl)pyridine-2- carboxylate To a solution of 1-benzyl-5-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyrazole (571 mg, 1.92 mmol, 1.3 equiv.) and tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-bromo-pyridine-2-carboxylate (833 mg, 1.47 mmol, 1 equiv.) in dioxane (10 mL) and H 2 O (3 mL) was added [2-(2- aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-butyl-phos phane;methanesulfonate (215 mg, 295 µmol, 0.2 equiv.) and K2CO3 (611 mg, 4.42 mmol, 3 equiv.). The mixture was stirred at 100 °C for 1 h under N 2 atmosphere. The reaction mixture was partitioned between water (30 mL) and ethyl acetate (30 mL). The organic phase was separated, washed with ethyl acetate (30 mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~10% methanol/dichloromethane ether). tert-Butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-(1-benzyl-5-methyl-pyrazol-4- yl)pyridine-2-carboxylate (0.9 g, 1.23 mmol, 83.7% yield, 90% purity) was obtained as a yellow solid which was used in the next step directly. MS (ESI) m/z: 657.4 [M+H] + . Step E. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-(1-benzyl-5-methyl-pyrazol-4-yl)pyrid ine-2-carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-(1-benzyl-5-methyl-pyrazol-4-yl)pyridine-2-carboxyla te (0.9 g, 1.37 mmol, 1 equiv.) in DCM (10 mL) was added TFA (7.70 g, 67.5 mmol, 5 mL, 49.3 equiv.). The mixture was stirred at 25 °C for 16 h. The reaction mixture was concentrated under reduced pressure to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-(1-benzyl-5- methyl-pyrazol-4-yl)pyridine-2-carboxylic acid (867 mg, crude) as a yellow oil which was used in next step directly. Step F. Procedure for preparation of methyl 2-[[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-(1-benzyl-5-methyl-pyrazo l-4-yl)pyridine-2- carbonyl]amino]acetate To a solution of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- (1-benzyl-5-methyl-pyrazol-4-yl)pyridine-2-carboxylic acid (260 mg, 433 µmol, 1 equiv.) in DMF (5 mL) was added HATU (197 mg, 519 µmol, 1.2 equiv.) and DIEA (168 mg, 1.30 mmol, 226 µL, 3 equiv.). The mixture was stirred at 25 °C for 30 min. Methyl 2-aminoacetate (65.2 mg, 519 µmol, 1.2 equiv., HCl) was added into the mixture and stirred at 25 °C for 1 h. To the reaction mixture was added water (50 mL), then the mixture was filtered to give a crude product. Methyl 2-[[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-iso quinolin-2- yl]-3-(1-benzyl-5-methyl-pyrazol-4-yl)pyridine-2-carbonyl]am ino]acetate (0.2 g, crude) was obtained as a yellow solid which was used in the next step directly. MS (ESI) m/z: 672.5 [M+H] + . Step G. Procedure for preparation of 2-[[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-(1-benzyl-5-methyl-pyrazol-4- yl)pyridine-2- carbonyl]amino]acetic acid To a solution of methyl 2-[[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(1-benzyl-5-methyl-pyrazol-4-yl)pyridine -2-carbonyl]amino]acetate (180 mg, 268 µmol, 1 equiv.) in THF (3 mL) was added LiOH.H2O (0.5 M, 1 mL, 1.87 equiv.). The mixture was stirred at 25 °C for 1 h. The reaction mixture was acidified with aqueous HCl (1 M) till pH = 3, and then the mixture was filtered to give 2-[[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-(1-benzyl-5- methyl-pyrazol-4-yl)pyridine- 2-carbonyl]amino]acetic acid (100 mg, crude) as a white solid which was used in the next step directly. MS (ESI) m/z: 658.1 [M+H] + . Step H. Procedure for preparation of N-(1,3-benzothiazol-2-yl)-2-[5-(1-benzyl-5-methyl- pyrazol-4-yl)-6-[[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-ind azol-6-yl]amino]-2-oxo- ethyl]carbamoyl]-2-pyridyl]-3,4-dihydro-1H-isoquinoline-8-ca rboxamide To a solution of 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (47.1 mg, 182 µmol, 1.2 equiv.) in DMF (3 mL) was added HATU (63.6 mg, 167.24 µmol, 1.1 equiv.) and DIEA (58.9 mg, 456 µmol, 79.4 µL, 3 equiv.), and then the mixture was stirred at 25 °C for 30 min. 2-[[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-iso quinolin-2-yl]-3-(1-benzyl-5- methyl-pyrazol-4-yl)pyridine-2-carbonyl]amino]acetic acid (100 mg, 152 µmol, 1 was added into the mixture and stirred at 25 °C for 30 min. The reaction mixture was filtered. The filtrate was purified by prep-HPLC. N-(1,3-benzothiazol-2-yl)-2-[5-(1-benzyl-5-methyl- pyrazol-4-yl)-6-[[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-ind azol-6-yl]amino]-2-oxo- ethyl]carbamoyl]-2-pyridyl]-3,4-dihydro-1H-isoquinoline-8-ca rboxamide (10.6 mg, 10.9 µmol, 7.20% yield, 93% purity) was obtained as a yellow solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ ppm = 10.88 (s, 1 H), 10.16 (s, 1 H), 8.71 (t, J = 5.6 Hz, 1 H), 7.95 (s, 1 H), 7.78 (d, J = 7.2 Hz, 1 H), 7.69 (m, 1 H), 7.57 - 7.64 (m, 2 H), 7.51 (d, J = 8.8 Hz, 1 H), 7.24 - 7.41 (m, 7 H), 7.17 - 7.24 (m, 2 H), 7.02 - 7.13 (m, 4 H), 5.27 (s, 2 H), 5.05 (s, 2 H), 4.32 (m, 1 H), 4.07 (m, 2 H), 3.98 (s, 2 H), 3.87 (s, 3 H), 2.96 - 3.07 (m, 3 H), 2.62 - 2.67 (m, 1 H), 2.27 - 2.35 (m, 1 H), 2.16 (s, 1 H), 2.03 (s, 3 H) Example 24. Preparation of Compound 137 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-((7-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin-1-yl )heptyl)oxy)-2- methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 4-[3-(2,6-dibenzyloxy-3-pyridyl)-1- methyl-indazol-6-yl]piperazine-1-carboxylate To a solution of 6-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (5 g, 9.99 mmol, 1.00 equiv.) in dioxane (60 mL) was added RuPhos (932.55 mg, 2.00 mmol, 0.20 equiv.), Cs2CO3 (9.77 g, 29.98 mmol, 3.00 equiv.), tert-butyl piperazine-1-carboxylate (5.58 g, 29.98 mmol, 3.00 equiv.) and Pd(dba) 2 (574.56 mg, 999.23 µmol, 0.10 equiv.). The mixture was stirred at 100 °C for 3 hours. The reaction mixture was concentrated under reduced pressure to remove solvent and then diluted with H2O (100 mL) and extracted with ethyl acetate (100 mL × 3). The combined organic layers were washed with H 2 O (100 mL × 3), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=100/1 to 3/1) to give tert- butyl 4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-yl]piper azine-1-carboxylate (5.6 g, 9.25 mmol, 92.52% yield) as a yellow oil. MS (ESI) m/z: 606.5 [M+H] + . Step B. Procedure for preparation of tert-butyl 4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazine-1-carboxylate To a solution of tert-butyl 4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-yl]piper azine- 1-carboxylate (5.6 g, 9.25 mmol, 1.00 equiv.) in THF (40 mL) and EtOH (30 mL) was added Pd(OH) 2 (2.86 g, 4.07 mmol, 20% purity, 0.30 equiv.) and Pd/C (2.85 g, 2.72 mmol, 10% purity, 0.20 equiv.), AcOH (2.45 g, 40.73 mmol, 2.33 mL, 3.00 equiv.) under N2 atmosphere. The suspension was degassed and purged with H 2 three times. The mixture was stirred under H 2 (50 Psi) at 50 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give tert-butyl 4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)piperazine-1-carboxylate (2.6 g, 6.08 mmol) as a white solid. MS (ESI) m/z: 428.5 [M+H] + . Step C. Procedure for preparation of 3-(1-methyl-6-piperazin-1-yl-indazol-3- yl)piperidine-2,6-dione To a solution of tert-butyl 4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)piperazine-1-carboxylate (2.4 g, 5.61 mmol, 1.00 equiv.) in DCM (25 mL) was added HCl/dioxane (4 M, 1.40 mL, 1.00 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give 3-(1-methyl-6-piperazin-1- yl-indazol-3-yl)piperidine-2,6-dione (1 g, 2.75 mmol, 48.9% yield) as a white solid. 1 H NMR (400 MHz, DMSO) δ = 10.87 (s, 1H), 9.20 (s, 2H), 7.57 (d, J = 8.8 Hz, 1H), 7.00 - 6.90 (m, 2H), 4.32 - 4.24 (m, 1H), 3.94 - 3.89 (m, 3H), 3.50 - 3.42 (m, 4H), 3.26 (s, 4H), 2.68 - 2.59 (m, 2H), 2.32 - 2.13 (m, 2H) Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-((7-(4-(3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)heptyl)oxy)-2-methylphenyl)picol inate To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2-methyl-3-((7-oxoheptyl)oxy)phenyl)picolinate (100 mg, 141.87 µmol, 1.00 equiv.) in DCM (1.5 mL) was added NMM (14.35 mg, 141.87 µmol, 15.60 µL, 1.00 equiv.) and 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (56.78 mg, 156.06 µmol, 1.10 equiv., HCl) at 25 °C for 4 hours. NaBH3CN (26.75 mg, 425.61 µmol, 3.00 equiv.) was added to the mixture and stirred at 25 °C for 1 hour. The reaction was diluted with water (20 mL) and extracted with DCM 30 mL (30 mL × 3). The combined organic layers were washed with brine 20 mL (20 mL × 1), dried over Na2SO4, filtered and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-((7-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1- yl)heptyl)oxy)-2-methylphenyl)picolinate (100 mg, 95.6 µmol, 67.4% yield, 97.2% purity) was obtained as a yellow oil. MS (ESI) m/z: 960.7 [M+H] + . Step E. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-((7-(4-(3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)heptyl)oxy)-2-methylphenyl)picol inic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-((7-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1- yl)heptyl)oxy)-2-methylphenyl)picolinate (100 mg, 98.40 µmol, 1.00 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 137.26 equiv.). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-((7-(4-(3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H-indazol- 6-yl)piperazin-1-yl)heptyl)oxy)-2-methylphenyl)picolinic acid (35.9 mg, 34.6 µmol, 35.2% yield, 97.1% purity, FA) as a white solid. MS (ESI) m/z: 960.1 [M+H] + . 1 H NMR (400 MHz, DMSO) δ = 13.05 - 12.70 (m, 1H), 10.86 (s, 1H), 8.14 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 7.2 Hz, 1H), 7.57 - 7.40 (m, 5H), 7.40 - 7.33 (m, 2H), 7.10 (t, J = 7.6 Hz, 1H), 6.97 - 6.87 (m, 4H), 6.64 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.31 - 4.24 (m, 1H), 3.98 (t, J = 6.0 Hz, 2H), 3.92 (s, 2H), 3.90 (s, 3H), 3.59 - 3.53 (m, 2H), 3.01 (t, J = 5.6 Hz, 2H), 2.88 - 2.75 (m, 4H), 2.65 - 2.57 (m, 6H), 2.32 - 2.26 (m, 1H), 2.20 - 2.14 (m, 1H), 1.91 (s, 3H), 1.78 - 1.72 (m, 2H), 1.55 (s, 2H), 1.49 - 1.44 (m, 2H), 1.41 - 1.32 (m, 4H) Example 25. Preparation of Compound 138b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4s)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1-yl)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo -3-piperidyl)-1-methyl- indazol-6-yl]piperazin-1-yl]-3-oxo-propyl]cyclohexoxy]-2-met hyl-phenyl]pyridine-2- carboxylate To a solution of 3-[4-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1 H-isoquinolin- 2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]cycl ohexyl]propanoic acid (150 mg, 200.83 µmol, 1 equiv.), 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (87.68 mg, 240.99 µmol, 1.2 equiv., HCl) in DMF (2 mL) was added HATU (91.63 mg, 240.99 µmol, 1.2 equiv.) and DIEA (77.87 mg, 602.48 µmol, 104.94 µL, 3 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was diluted with water (10 mL) and filtered to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- [3-[4-[3-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl ]piperazin-1-yl]-3-oxo- propyl]cyclohexoxy]-2-methyl-phenyl]pyridine-2-carboxylate (170 mg, crude) as a yellow solid. MS (ESI) m/z: 529.0 [1/2M+H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4s)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1-yl)-3-oxopropyl)cyclohexyl)oxy)- 2-methylphenyl)picolinic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-inda zol-6-yl]piperazin-1-yl]-3-oxo- propyl]cyclohexoxy]-2-methyl-phenyl]pyridine-2-carboxylate (170 mg, 160.94 µmol, 1 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 83.92 equiv.)The mixture was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1s,4s)-4-(3-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin -1-yl)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (21.1 mg, 20.1 µmol, 12.5% yield, 95.1% purity) as a white solid. MS (ESI) m/z: 500.9 [1/2M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 13.12 - 12.29 (m, 2H), 10.85 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 6.8 Hz, 1H), 7.52 (d, J = 9.2 Hz, 1H), 7.49 - 7.42 (m, 3H), 7.40 - 7.32 (m, 2H), 7.10 - 7.04 (m, 1H), 6.98 - 6.91 (m, 2H), 6.91 - 6.86 (m, 2H), 6.60 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.61 (s, 1H), 4.26 (dd, J = 5.0, 9.2 Hz, 1H), 3.93 - 3.90 (m, 2H), 3.89 (s, 3H), 3.61 (d, J = 4.8 Hz, 4H), 3.24 - 3.16 (m, 4H), 3.02 (t, J = 5.6 Hz, 2H), 2.65 - 2.56 (m, 2H), 2.41 - 2.36 (m, 2H), 2.33 - 2.27 (m, 1H), 2.19 - 2.12 (m, 1H), 1.93 (s, 3H), 1.91 - 1.86 (m, 1H), 1.55 (d, J = 1.6 Hz, 4H), 1.50 - 1.44 (m, 2H), 1.41 - 1.20 (m, 4H) Example 26. Preparation of Compound 141 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(4-(2-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin -1-yl)-2-oxoethyl)piperazin-1- yl)ethoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-(2-(4-(3-(2,6-dioxop iperidin-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)-2-oxoethyl)piperazin-1-yl)ethox y)-2- methylphenyl)picolinate To a solution of 2-(4-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)ethyl)piperazin-1-yl)acetic acid (100 mg, 131.08 µmol, 1 equiv.) in DMF (1 mL) was added HATU (64.79 mg, 170.40 µmol, 1.3 equiv.), DIEA (50.82 mg, 393.23 µmol, 68.49 µL, 3 equiv.) and 3-(1-methyl-6- piperazin-1-yl-indazol-3-yl)piperidine-2,6-dione (52.46 mg, 144.18 µmol, 1.1 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by prep-TLC (SiO 2 , DCM: MeOH = 10:1). The compound tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(4-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)piperazin- 1-yl)-2-oxoethyl)piperazin-1-yl)ethoxy)-2-methylphenyl)picol inate (70 mg, 65.28 µmol, 49.80% yield) was obtained as a white solid. MS (ESI) m/z: 1072.6 [M+H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-(2-(4-(3-(2,6-dioxop iperidin-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)-2-oxoethyl)piperazin-1-yl)ethox y)-2-methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(4-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)piperazin- 1-yl)-2-oxoethyl)piperazin-1-yl)ethoxy)-2-methylphenyl)picol inate (70 mg, 65.28 µmol, 1 equiv.) in DCM (0.5 mL) was added TFA (770.00 mg, 6.75 mmol, 0.5 mL, 103.44 equiv.). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by prep-HPLC. The compound 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(2-(4-(2-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin-1-yl )-2-oxoethyl)piperazin-1- yl)ethoxy)-2-methylphenyl)picolinic acid (18.5 mg, 16.8 µmol, 25.7% yield, 96.5% purity) was obtained as a yellow solid. MS (ESI) m/z: 1016.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.86 (s, 1H), 8.17 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 7.6 Hz, 1H), 7.56 - 7.32 (m, 7H), 7.09 (t, J = 8.0 Hz, 1H), 6.95 (d, J = 8.8 Hz, 2H), 6.92 - 6.87 (m, 2H), 6.64 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.30 - 4.25 (m, 1H), 4.08 - 4.04 (m, 2H), 3.92 (s, 2H), 3.90 (s, 3H), 3.74 (s, 2H), 3.62 (d, J = 2.0 Hz, 2H), 3.28 - 3.25 (m, 4H), 3.19 (s, 4H), 3.04 (d, J = 5.2 Hz, 2H), 2.72 (t, J = 5.6 Hz, 2H), 2.63 - 2.56 (m, 6H), 2.46 (s, 2H), 2.32 - 2.25 (m, 1H), 2.19 - 2.12 (m, 1H), 1.89 (s, 3H) Example 27. Preparation of Compound 144 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[1-[2-[4-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]piperazin-1-yl ]-2-oxo-ethyl]-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[4-[3-(2,6-di oxo-3-piperidyl)-1-methyl- indazol-7-yl]piperazin-1-yl]-2-oxo-ethyl]-4-piperidyl]propox y]-2-methyl- phenyl]pyridine-2-carboxylate To a solution of 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]propyl]-1- piperidyl]acetic acid (70 mg, 90.21 µmol, 1 equiv.) in DMF (1 mL) was added 3-(1-methyl-7- piperazin-1-yl-indazol-3-yl)piperidine-2,6-dione (29.53 mg, 90.21 µmol, 1 equiv.), HATU (34.30 mg, 90.21 µmol, 1 equiv.) and DIEA (11.66 mg, 90.21 µmol, 15.71 µL, 1 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give the crude product tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[4-[3-(2,6-dioxo- 3-piperidyl)-1-methyl-indazol- 7-yl]piperazin-1-yl]-2-oxo-ethyl]-4-piperidyl]propoxy]-2-met hyl-phenyl]pyridine-2- carboxylate (70 mg, 38.05 µmol, 42.18% yield, 59% purity) as a yellow oil. MS (ESI) m/z: 543.4 [M/2+H] + Step B. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[4-[3-(2,6-dioxo-3-piperi dyl)-1-methyl-indazol-7- yl]piperazin-1-yl]-2-oxo-ethyl]-4-piperidyl]propoxy]-2-methy l-phenyl]pyridine-2- carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[3-[1-[2-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-i ndazol-7-yl]piperazin-1-yl]-2- oxo-ethyl]-4-piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-c arboxylate (70 mg, 64.50 µmol, 1 equiv.) in TFA (1 mL) and DCM (2 mL). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove solvent to give the crude product. The crude product was purified by reverse-phase HPLC to give compound 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[1-[2-[4-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]piperazin-1-yl ]-2-oxo-ethyl]-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid (15.9 mg, 14.6 µmol, 22.6% yield, 94.1% purity) as a white solid. MS (ESI) m/z: 1029.1 [M+H] + . 1 H NMR (400 MHz, CDCl3) δ = 7.84 - 7.65 (m, 2H), 7.56 ( s, 1H), 7.41 - 7.28 (m, 4H), 7.24 ( t, J = 7.6 Hz, 2H), 7.06 (t, J = 8.0 Hz, 1H), 6.99 - 6.84 (m, 3H), 6.73 ( d, J = 8.4 Hz, 1H), 6.62 (d, J = 7.6 Hz, 1H), 5.15 ( d, J = 5.2 Hz, 2H), 4.59 ( d, J = 10.8 Hz, 1H), 4.23 (s, 3H), 3.97 - 3.86 (m, 3H), 3.83 - 3.72 (m, 1H), 3.37 - 3.21 (m, 2H), 3.13 ( s, 3H), 3.03 ( d, J = 4.4 Hz, 2H), 2.87 - 2.72 (m, 4H), 2.64 - 2.55 (m, 1H), 2.48 - 2.24 (m, 4H), 2.15 - 1.98 (m, 4H), 1.84 ( s, 3H), 1.75 - 1.62 (m, 5H), 1.38 - 1.31 (m, 2H), 1.18 (s, 2H), 0.93 - 0.63 (m, 1H). Example 28. Preparation of Compound 147b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4s)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pip erazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of methyl 4-(3-bromo-2-methyl-phenoxy) cyclohexanecarboxylate To a solution of 3-bromo-2-methyl-phenol (5.91 g, 31.61 mmol, 1 equiv.) in toluene (50 mL) was added methyl 4-hydroxycyclohexanecarboxylate (5 g, 31.61 mmol, 1 equiv.) and 2- (tributyl-λ 5 -phosphanylidene) acetonitrile (9.15 g, 37.93 mmol, 1.2 equiv.) at 25 °C. The reaction mixture was degassed and purged with N 2 three times, and then the mixture was stirred at 120 °C for 16 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 20/1 to 10/1). The compound methyl 4-(3-bromo-2- methyl-phenoxy) cyclohexanecarboxylate (22.8 g, 43.9 mmol, 69.6% yield, 62.9% purity) was obtained as a colorless oil. Step B. Procedure for preparation of [4-(3-bromo-2-methyl-phenoxy) cyclohexyl]methanol To a solution of methyl 4-(3-bromo-2-methyl-phenoxy) cyclohexanecarboxylate (7.61 g, 14.65 mmol, 62.98% purity, 1 equiv.) in THF (70 mL) was added LiAlH 4 (555.86 mg, 14.65 mmol, 1 equiv.) dropwise at 0 °C under N 2 . The reaction mixture was stirred under N 2 at 0 °C for 2 hours. The reaction mixture was quenched with Na2SO4•10 H2O (100 mg) and slowly warmed to 20 °C. The mixture was filtered and washed with DCM (50 mL). Then the organic layers were concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 50/1 to 20/1). The compound [4-(3-bromo-2-methyl-phenoxy) cyclohexyl]methanol (8.4 g, 27.7 mmol, 63.0% yield, 98.7% purity) was obtained as a yellow oil. 1 H NMR (400 MHz, CDCl3-d) δ = 7.12 (d, J = 8.0 Hz, 1H), 6.98 (t, J = 8.0 Hz, 1H), 6.78 (d, J = 8.4 Hz, 1H), 4.61 - 4.54 (m, 1H), 3.52 (d, J = 6.0 Hz, 2H), 2.34 (s, 3H), 2.11 - 2.05 (m, 2H), 1.66 - 1.61 (m, 3H), 1.59 - 1.53 (m, 2H), 1.49 - 1.40 (m, 2H) Step C. Procedure for preparation of 4-(3-bromo-2-methyl-phenoxy) cyclohexanecarbaldehyde To a solution of DMSO (4.39 g, 56.15 mmol, 4.39 mL, 4 equiv.) in DCM (80 mL) was added dropwise to a solution of oxalyl dichloride (3.56 g, 28.07 mmol, 2.46 mL, 2 equiv.) in DCM (5 mL) at -70 °C under N 2 atmosphere. The mixture was stirred at -70 °C for 30 minutes. After which time [4-(3-bromo-2-methyl-phenoxy) cyclohexyl]methanol (4.2 g, 14.04 mmol, 1 equiv.) in DCM (5 mL) was added dropwise at -70 °C. The solution was stirred for 30 minutes at - 70 °C. Then TEA (8.52 g, 84.22 mmol, 11.72 mL, 6 equiv.) was added into the solution. The solution was stirred at 25 °C for 1 hour under N 2 atmosphere. The reaction mixture was diluted with water 20 mL and extracted with DCM 60 mL (20 mL × 3). The combined organic layers were washed with brine (10 mL × 2), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The compound 4-(3-bromo-2-methyl-phenoxy) cyclohexanecarbaldehyde (9.33 g, crude) was obtained as a yellow oil. Step D. Procedure for preparation of ethyl (E)-3-[4-(3-bromo-2-methyl-phenoxy) cyclohexyl]prop-2-enoate To a solution of NaH (282.62 mg, 7.07 mmol, 60% purity, 2.1 equiv.) in THF (10 mL) was added ethyl 2-diethoxyphosphorylacetate (1.51 g, 6.73 mmol, 1.34 mL, 2 equiv.) under 0 °C. The mixture was stirred at 0 °C for 4 hours. Then 4-(3-bromo-2-methyl-phenoxy) cyclohexanecarbaldehyde (1 g, 3.36 mmol, 1 equiv.) was added and the mixture was stirred at 25 °C for 12 hours. The reaction mixture was quenched by addition sat. NH4Cl (10 mL) at 0 °C, and then extracted with ethyl acetate 60 mL (20 mL × 3). The combined organic layers were washed with brine (20 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 – 10% ethyl acetate/petroleum ether). The compound ethyl (E)-3-[4-(3-bromo-2-methyl- phenoxy) cyclohexyl]prop-2-enoate (1.2 g, 2.3 mmol, 68.8% yield, 74% purity) was obtained as a yellow oil. 1 H NMR (400 MHz, CDCl3-d) δ = 7.14 (d, J = 8.0 Hz, 1H), 7.01 - 6.92 (m, 2H), 6.78 (d, J = 8.4 Hz, 1H), 5.86 - 5.79 (m, 1H), 4.60 - 4.52 (m, 1H), 4.24 - 4.19 (m, 2H), 2.37 - 2.32 (m, 3H), 2.31 - 2.22 (m, 1H), 2.14 - 2.05 (m, 2H), 1.66 - 1.62 (m, 4H), 1.36 - 1.28 (m, 5H) Step E. Procedure for preparation of ethyl 3-[4-(3-bromo-2-methyl-phenoxy) cyclohexyl]propanoate To a solution of ethyl (E)-3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]prop-2-enoate (1.14 g, 3.11 mmol, 1 equiv.) in EtOH (10 mL) was added PtO2 (141.43 mg, 622.81 μmol, 0.2 equiv.) under N 2 atmosphere. The suspension was degassed and purged with H 2 three times. The mixture was stirred under H2 (15 Psi) at 25 °C for 40 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The compound ethyl 3-[4-(3- bromo-2-methyl-phenoxy) cyclohexyl]propanoate (890 mg, crude) was obtained as a yellow oil. 1 H NMR (400 MHz, CDCl3-d) δ = 7.13 (d, J = 8.0 Hz, 1H), 6.98 (t, J = 8.0 Hz, 1H), 6.78 (d, J = 8.0 Hz, 1H), 4.59 - 4.53 (m, 1H), 4.16 (m, J = 7.2 Hz, 2H), 2.39 - 2.32 (m, 5H), 2.08 - 1.99 (m, 2H), 1.65 - 1.54 (m, 5H), 1.44 - 1.33 (m, 4H), 1.28 (t, J = 7.2 Hz, 3H) SFC (EC2592-447-P1): Column: Chiralpak AD - 350 x 4.6 mm I.D., 3um Mobile phase: Phase A for CO2, and Phase B for MeOH (0.05% DEA); Gradient elution: MeOH (0.05% DEA) in CO 2 from 5% to 40% Flow rate: 3 mL/min; Detector: PDA; Column Temp: 35 C; Back Pressure: 100 Bar" Step F. Procedure for preparation of 3-[4-(3-bromo-2-methyl-phenoxy) cyclohexyl]propan-1-ol To a solution of ethyl 3-[4-(3-bromo-2-methyl-phenoxy) cyclohexyl]propanoate (280 mg, 758.21 μmol, 1 equiv.) in THF (3 mL) was added LiAlH4 (34.53 mg, 909.85 μmol, 1.2 equiv.) dropwise at 0 °C under N 2 . The reaction mixture was stirred under N 2 at 0 °C for 2 hours. The reaction mixture was quenched with Na 2 SO 4 •10 H 2 O (10 mg) and slowly warmed to 20 °C. The mixture was filtered and washed with DCM (10 mL). Then the organic layers were concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate = 50/1 to 20/1). The compound 3-[4-(3- bromo-2-methyl-phenoxy) cyclohexyl]propan-1-ol (160 mg, 433.67 μmol, 57.20% yield, 88.7% purity) was obtained as a colorless oil. Step G. Procedure for preparation of 3-[4-(3-bromo-2-methyl-phenoxy) cyclohexyl]propanal A solution of DMSO (135.53 mg, 1.73 mmol, 135.53 μL, 4 equiv.) in DCM (2 mL) was added dropwise to a solution of oxalyl dichloride (110.09 mg, 867.33 μmol, 75.92 μL, 2 equiv.) in DCM (1 mL) at -70 °C under N2 atmosphere. The mixture was stirred at -70 °C for 30 minutes. After which time 3-[4-(3-bromo-2-methyl-phenoxy) cyclohexyl]propan-1-ol (160 mg, 433.67 μmol, 88.7% purity, 1 equiv.) in DCM (1 mL) was added dropwise at -70 °C. The solution was stirred for 30 minutes at -70 °C. Then TEA (263.30 mg, 2.60 mmol, 362.17 μL, 6 equiv.) was added into the solution. The solution was stirred at 25 °C for 1 hour under N2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with DCM (10 mL × 3). The combined organic layers were washed with brine (10 mL × 1), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The compound 3-[4-(3-bromo-2- methyl-phenoxy) cyclohexyl]propanal (215 mg, crude) was obtained as a yellow oil. Step H. Procedure for tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H- isoquinolin-2-yl]-3-[2-methyl-3-[4-(3-oxopropyl) cyclohexoxy]phenyl]pyridine-2- carboxylate tert-Butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-3-(4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridine-2-carboxylate (327.33 mg, 534.37 μmol, 1.1 equiv.), 3-[4-(3-bromo-2-methyl-phenoxy) cyclohexyl]propanal (200 mg, 485.80 μmol, 79% purity, 1 equiv.), KF (84.67 mg, 1.46 mmol, 34.14 μL, 3 equiv.) and [2-(2-aminophenyl) phenyl]palladium (1+) ;bis (1-adamantyl)-butyl-phosphane;methanesulfonate (70.76 mg, 97.16 μmol, 0.2 equiv.) were taken up into a microwave tube in dioxane (4 mL) and H2O (0.4 mL). The sealed tube was heated at 100 °C for 60 min under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0 ~ 30% ethyl acetate/petroleum ether). The compound tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H- isoquinolin-2-yl]-3-[2-methyl-3-[4-(3-oxopropyl) cyclohexoxy]phenyl]pyridine-2- carboxylate (240 mg, 292.9 μmol, 60.3% yield, 89.2% purity) was obtained as a yellow solid. MS (ESI) m/z: 731.5 [M+H] + . Step I. Procedure for tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H- isoquinolin-2-yl]-3-[3-[4-[3-[4-[3-(2, 6-dioxo-3-piperidyl) -1-methyl-indazol-7- yl]piperazin-1-yl]propyl]cyclohexoxy]-2-methyl-phenyl]pyridi ne-2-carboxylate To a solution of tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H- isoquinolin-2-yl]-3-[2-methyl-3-[4-(3-oxopropyl) cyclohexoxy]phenyl]pyridine-2- carboxylate (140 mg, 191.54 μmol, 1 equiv.) in DCM (2 mL) was added 3-(1-methyl-7- piperazin-1-yl-indazol-3-yl) piperidine-2, 6-dione (69.69 mg, 191.54 μmol, 1 equiv., HCl) at 25 °C. The mixture was stirred at 25 °C for 16 hours. NaBH(OAc) 3 (121.79 mg, 574.62 μmol, 3 equiv.) was added to the mixture at 25 °C. The reaction mixture was stirred at 25 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , DCM: MeOH = 20:1). The compound tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4- [3-[4-[3-(2, 6-dioxo-3-piperidyl) -1-methyl-indazol-7-yl]piperazin-1-yl]propyl]cyclohexoxy]- 2-methyl-phenyl]pyridine-2-carboxylate (240 mg, 148.06 μmol, 77.3% yield, 64.3% purity) was obtained as a yellow solid. MS (ESI) m/z: 1042.5 [M+H] + . Step J. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4s)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)propyl)cyclohexyl)oxy) -2-methylphenyl)picolinic acid To a solution of tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H- isoquinolin-2-yl]-3-[3-[4-[3-[4-[3-(2, 6-dioxo-3-piperidyl) -1-methyl-indazol-7-yl]piperazin- 1-yl]propyl]cyclohexoxy]-2-methyl-phenyl]pyridine-2-carboxyl ate (240 mg, 148.06 μmol, 64.3% purity, 1 equiv.) in DCM (1.5 mL) and TFA (1.5 mL) was stirred at 25 °C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC. The compound 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4s) -4-(3-(4-(3-(2,6-dioxopiperidin- 3-yl)-1-methyl-1H-indazol-7-yl)piperazin-1-yl)propyl)cyclohe xyl)oxy)-2- methylphenyl)picolinic acid (28.9 mg, 28.5 μmol, 19.2% yield, 96.9% purity) was obtained as a yellow solid. MS (ESI) m/z: 986.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.88 (s, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.49 - 7.42 (m, 3H), 7.40 - 7.32 (m, 3H), 7.06 (d, J = 8.0 Hz, 1H), 7.03 - 7.00 (m, 2H), 6.96 - 6.84 (m, 2H), 6.61 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.60 (s, 1H), 4.33 (dd, J = 5.2, 9.6 Hz, 1H), 4.22 (s, 3H), 3.91 (t, J = 5.6 Hz, 2H), 3.02 (t, J = 5.6 Hz, 2H), 2.95 - 2.85 (m, 3H), 2.69 - 2.58 (m, 3H), 2.43 - 2.25 (m, 5H), 2.18 (d, J = 5.6 Hz, 1H), 1.97 - 1.86 (m, 5H), 1.62 - 1.46 (m, 6H), 1.43 - 1.10 (m, 6H). Example 29. Preparation of Compound 149 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(4-(3-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin -1-yl)propyl)phenoxy)-2- methylphenyl)picolinic acid Step A. Procedure for preparation of methyl 3-(4-(3-bromo-2- methylphenoxy)phenyl)propanoate A mixture of methyl 3-(4-hydroxyphenyl)propanoate (4.64 g, 25.74 mmol, 1.0 equiv.), 1,3- dibromo-2-methyl-benzene (12.87 g, 51.49 mmol, 2.0 equiv.), Cs 2 CO 3 (12.58 g, 38.61 mmol, 1.5 equiv.) and 2,2,6,6-tetramethylheptane-3,5-dione (1.19 g, 6.44 mmol, 1.33 mL, 0.25 equiv.) in NMP (50 mL) was degassed and purged with N 2 three times, and then the mixture was added CuI (980.56 mg, 5.15 mmol, 0.2 equiv.) at 120 °C for 12 hours under N 2 atmosphere. The reaction mixture was quenched by addition of water 30 mL at 25°C and extracted with ethyl acetate (50 mL). The combined organic layers were washed with NaHCO3 (50 mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~5% ethyl acetate/petroleum ether). The compound methyl 3-(4-(3-bromo-2-methylphenoxy)phenyl)propanoate (8.5 g, 24.3 mmol, 94.5% yield) was obtained as a yellow oil. 1 H NMR (400 MHz, CDCl 3 ) δ = 7.24 (d, J = 8.0 Hz, 1H), 7.05 (d, J = 8.4 Hz, 2H), 6.91 (t, J = 8.0 Hz, 1H), 6.78 - 6.70 (m, 3H), 3.58 (s, 3H), 2.83 (t, J = 7.6 Hz, 2H), 2.53 (t, J = 7.6 Hz, 2H), 2.24 (s, 3H) Step B. Procedure for preparation of 3-(4-(3-bromo-2-methylphenoxy)phenyl)propan-1- ol A mixture of methyl 3-[4-(3-bromo-2-methyl-phenoxy)phenyl]propanoate (4.00 g, 11.45 mmol, 1 equiv.) in THF (50 mL) was slowly added LiAlH 4 (521.62 mg, 13.74 mmol, 1.2 equiv.) at 0 °C, and then the mixture was stirred at 25 °C for 2 hours. The mixture was poured into ice- water (60 mL) slowly, acidified to pH = 4-5, extracted with ethyl acetate 120 mL (40 mL × 3), washed with brine 40 mL (20 mL × 2), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The compound 3-(4-(3-bromo-2-methylphenoxy)phenyl)propan-1-ol (3.3 g, 10.3 mmol, 89.7% yield) was obtained as a yellow oil. Step C. Procedure for preparation of 3-(4-(3-bromo-2-methylphenoxy)phenyl)propanal To a solution of oxalyl dichloride (395.16 mg, 3.11 mmol, 272.53 µL, 2.0 equiv.) in DCM (10 mL) was added the mixture of DMSO (486.48 mg, 6.23 mmol, 486.48 µL, 4.0 equiv.) under - 70°C and stirred for 0.5 hours. 3-[4-(3-bromo-2-methyl-phenoxy)phenyl]propan-1-ol (0.5 g, 1.56 mmol, 1.0 equiv.) in DCM (10 mL) was added into the mixture and stirred for 0.5 hours. TEA (945.09 mg, 9.34 mmol, 1.30 mL, 6 equiv.) was added into the mixture and stirred at -70 °C for 1 hour. The reaction mixture was diluted with citric acid 20 mL and extracted with DCM 45 mL (15 mL × 3). The combined organic layers were washed with NaHCO 3 (10 mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~15% ethyl acetate/petroleum ether). The compound 3-(4-(3-bromo-2-methylphenoxy)phenyl)propanal (250 mg, 783.2 µmol, 50.3% yield) was obtained as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 9.71 (s, 1H), 7.41 (d, J = 8.0 Hz, 1H), 7.24 - 7.18 (m, 2H), 7.15 (t, J = 8.0 Hz, 1H), 6.88 - 6.81 (m, 3H), 2.82 (d, J = 6.4 Hz, 2H), 2.79 - 2.71 (m, 2H), 2.26 (s, 3H) Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(2-methyl-3-(4-(3-oxopropyl)p henoxy)phenyl)picolinate A mixture of 3-(4-(3-bromo-2-methylphenoxy)phenyl)propanal (250 mg, 783.23 µmol, 1.0 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)picolinate (479.76 mg, 783.23 µmol, 1.0 equiv.), K2CO3 (1.5 M, 1.57 mL, 3.0 equiv.), and Ad2nBuP Pd G3(cataCXium® A Pd G3 ) (114.08 mg, 156.65 µmol, 0.2 equiv.) in 1,4-dioxane (3 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 80 °C for 1 hour under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0-30% ethyl acetate/petroleum ether). The compound tert- butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2-methyl-3- (4-(3-oxopropyl)phenoxy)phenyl)picolinate (200 mg, 219.1 µmol, 27.9% yield, 79.4% purity) was obtained as a yellow oil. Step E. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(3-(4-(3-(2,6-dioxopipe ridin-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)propyl)phenoxy)-2-methyl phenyl)picolinate A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(2-methyl-3-(4-(3-oxopropyl)phenoxy)phenyl)picolinate (100 mg, 108.99 µmol, 79.4% purity, 1.0 equiv.), 3-(1-methyl-6-(piperazin-1-yl)-1H-indazol-3-yl)piperidine-2, 6-dione (42.82 mg, 130.78 µmol, 1.2 equiv.), HCOOH (5.24 mg, 108.99 µmol, 1 equiv.), and NaBH(OAc) 3 (34.65 mg, 163.48 µmol, 1.5 equiv.) in DCM (2 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 16 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The compound tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)- 3-(3-(4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indaz ol-6-yl)piperazin-1- yl)propyl)phenoxy)-2-methyl phenyl)picolinate (150 mg, crude) was obtained as a yellow oil. MS (ESI) m/z: 1036.7 [M+H] + . Step F. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(3-(4-(3-(2,6-dioxopipe ridin-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)propyl)phenoxy)-2-methylphenyl)p icolinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-6-yl)piperazin-1- yl)propyl)phenoxy)-2-methyl phenyl)picolinate (150 mg, 144.75 µmol, 1.0 equiv.), TFA (16.50 mg, 144.75 µmol, 10.72 µL, 1.0 equiv.) in DCM (2 mL) was stirred at 25 °C for 21 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(3-(4-(3-(2,6-dioxopipe ridin-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)propyl)phenoxy)-2-methylphenyl)p icolinic acid (20.9 mg, 20.1 µmol, 13.9% yield, 94.4% purity) as a white solid. MS (ESI) m/z: 981.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.05 - 12.66 (m, 1H), 10.85 (s, 1H), 8.13 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.59 - 7.50 (m, 2H), 7.50 - 7.42 (m, 3H), 7.40 - 7.33 (m, 2H), 7.22 - 7.15 (m, 3H), 7.01 (d, J = 8.8 Hz, 1H), 6.90 (dd, J = 8.4, 15.2 Hz, 2H), 6.86 - 6.80 (m, 4H), 4.99 (s, 2H), 4.25 (dd, J = 4.8, 8.8 Hz, 1H), 3.93 (s, 2H), 3.89 (s, 3H), 3.03 (t, J = 4.8 Hz, 2H), 2.67 (s, 2H), 2.64 - 2.56 (m, 10H), 2.33 (s, 2H), 2.17 (d, J = 5.2 Hz, 2H), 1.90 (s, 3H), 1.83 - 1.74 (m, 2H) Example 30. Preparation of Compound 155 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[9-[4-[3-(2,6- dioxo-3-piperidyl)-1-methyl-indazol-6-yl]piperazin-1-yl]nono xy]-2-methyl-phenyl]pyridine- 2-carboxylic acid Step A. Procedure for preparation of ethyl 9-(3-bromo-2-methyl-phenoxy)nonanoate To a solution of 3-bromo-2-methyl-phenol (2.0 g, 10.69 mmol, 1 equiv.) in CH3CN (50 mL) was added K 2 CO 3 (4.43 g, 32.07 mmol, 3 equiv.) and ethyl 9-bromononanoate (2.83 g, 10.69 mmol, 1 equiv.). The mixture was stirred at 60 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate =1/0 to 10/1) to give compound ethyl 9- (3-bromo-2-methyl-phenoxy)nonanoate (3.2 g, 8.6 mmol, 80.6% yield) as a yellow oil. 1 H NMR (400 MHz, CDCl3) δ = 7.15 (d, J = 8.0 Hz, 1H), 7.00 (t, J = 8.0 Hz, 1H), 6.78 (d, J = 8.0 Hz, 1H), 4.15 (q, J = 7.2 Hz, 2H), 3.96 (t, J = 6.4 Hz, 2H), 3.43 (t, J = 6.8 Hz, 1H), 2.36 - 2.28 (m, 5H), 1.85 - 1.77 (m, 2H), 1.65 ( t, J = 7.2 Hz, 2H), 1.53 - 1.44 (m, 2H), 1.44 - 1.31 (m, 8H), 1.28 (t, J = 7.2 Hz, 3H). Step B. Procedure for preparation of 9-(3-bromo-2-methyl-phenoxy)nonan-1-ol A mixture of LiAlH 4 (204.44 mg, 5.39 mmol, 1 equiv.) in THF (25 mL) was degassed and purged with N2 three times, and ethyl 9-(3-bromo-2-methyl-phenoxy)nonanoate (2.0 g, 5.39 mmol, 1 equiv.) was added at 0 °C, and then the mixture was stirred at 0 °C for 2 hours under N 2 atmosphere. The mixture was quenched by water (0.6 mL) at 0 °C, and then 15% sodium hydroxide aqueous solution (0.6 mL) and water (0.6 mL), anhydrous sodium sulfate (2.0 g) was added. The mixture was stirred at 25 °C for 30 minutes, filtered, and dissolved in dichloromethane/methanol (4/1, 10 mL). The mixture was stirred at 25 °C for 1 hour, filtered, and concentrated. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate =1/0 to 4/1) to give the compound 9-(3-bromo-2-methyl-phenoxy)nonan-1- ol (1.5 g, 4.5 mmol, 84.6% yield) as a yellow oil. 1 H NMR (400 MHz, CDCl 3 ) δ = 7.06 (d, J = 8.0 Hz, 1H), 6.91 (t, J = 8.0 Hz, 1H), 6.68 (d, J = 8.0 Hz, 1H), 3.87 (t, J = 6.4 Hz, 2H), 3.57 (t, J = 6.8 Hz, 2H), 2.24 (s, 3H), 1.75 - 1.68 (m, 2H), 1.52 - 1.47 (m, 2H), 1.42 - 1.34 (m, 4H), 1.27 ( s, 6H). Step C. Procedure for preparation of 9-(3-bromo-2-methyl-phenoxy)nonanal To a solution of DMSO (759.33 mg, 9.72 mmol, 759.33 µL, 4 equiv.) in DCM (20 mL) was added dropwise (COCl)2 (616.76 mg, 4.86 mmol, 425.35 µL, 2 equiv.) at -70 °C over 10 min. After addition, the mixture was stirred at this temperature for 1 hour, and then 9-(3-bromo-2- methyl-phenoxy)nonan-1-ol (800 mg, 2.43 mmol, 1 equiv.) was added dropwise at 70 °C for 2 hours under N2 atmosphere. TEA (1.48 g, 14.58 mmol, 2.03 mL, 6 equiv.) was added dropwise at 70 °C for 0.5 hour under N 2 . The mixture was stirred at -70 °C for 1 hour. The reaction mixture was extracted with DCM (10 ml) and concentrated under reduced pressure to remove solvent to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=1/0 to 5/1) to give the compound 9-(3-bromo-2-methyl- phenoxy)nonanal (360 mg, 1.1 mmol, 45.3% yield) as a colorless oil. 1 H NMR (400 MHz, CDCl3) δ = 9.70 (t, J = 1.6 Hz, 1H), 7.06 (d, J = 8.0 Hz, 1H), 6.91 (t, J = 8.0 Hz, 1H), 6.68 (d, J = 8.0 Hz, 1H), 3.90 - 3.84 (m, 2H), 2.39 - 2.32 (m, 2H), 2.24 (s, 3H), 1.78 - 1.67 (m, 2H), 1.57 (m, J = 7.2 Hz, 2H), 1.42 - 1.36 (m, 2H), 1.28 ( s, 7H), 0.85 - 0.74 (m, 1H) Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(2-methyl-3-((9-oxononyl)oxy) phenyl)picolinate 9-(3-bromo-2-methyl-phenoxy)nonanal (300 mg, 916.71 µmol, 1 equiv.), tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyridine-2-carboxylate (561.53 mg, 916.71 µmol, 1 equiv.), K 2 CO 3 (380.09 mg, 2.75 mmol, 3 equiv.), and Ad2nBuP Pd G3(cataCXium® A Pd G3 ) (66.76 mg, 91.67 µmol, 0.1 equiv.) were taken up into a microwave tube in 1,4-dioxane (5 mL) and H2O (1 mL).The sealed tube was heated at 100 °C for 1 hour under microwave. The reaction mixture was concentrated under reduced pressure to remove solution to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate =1/0 to 2/1) to give the compound tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(2-methyl-3-((9-oxononyl)oxy)phenyl)picolinate (200 mg, 272.9 µmol, 29.8% yield) as a yellow solid. MS (ESI) m/z: 733.5 [M+H] + Step E. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[9-[4-[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol- 6-yl]piperazin-1-yl]nonoxy]-2-methyl-phenyl]pyridine-2-carbo xylate To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2-methyl-3-((9-oxononyl)oxy)phenyl)picolinate (100 mg, 136.44 µmol, 1 equiv.) in EtOH (2 mL) were added 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (44.67 mg, 136.44 µmol, 1 equiv.), TFA (15.56 mg, 136.44 µmol, 10.10 µL, 1 equiv.). The mixture was stirred at 25 °C for 0.5 hour, and then NaBH(OAc)3 (86.75 mg, 409.32 µmol, 3 equiv.) was added to the mixture, and the final mixture was stirred at 25 o C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2,DCM: MeOH = 1/0 to 20/1) to give the compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[9- [4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]piperazi n-1-yl]nonoxy]-2-methyl- phenyl]pyridine-2-carboxylate (15 mg, 14.4 µmol, 10.5% yield) as a yellow solid. MS (ESI) m/z: 523.0 [M/2+H] + Step F. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[9-[4-[3-(2,6-dioxo-3-piperidyl)-1 -methyl-indazol-6- yl]piperazin-1-yl]nonoxy]-2-methyl-phenyl]pyridine-2-carboxy lic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[9-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6 -yl]piperazin-1-yl]nonoxy]-2- methyl-phenyl]pyridine-2-carboxylate (80 mg, 76.61 µmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL) was stirred at 40 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by reverse-phase HPLC to give the compound 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[9- [4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]piperazi n-1-yl]nonoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid (50.2 mg, 47.0 µmol, 61.3% yield, 92.4% purity) as a white solid. MS (ESI) m/z : 988.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.29 - 12.43 (m, 1H), 10.86 (s, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.52 (d, J = 8.8 Hz, 1H), 7.49 - 7.40 (m, 3H), 7.39 - 7.32 (m, 2H), 7.12 - 7.06 (m, 1H), 6.98 - 6.84 (m, 4H), 6.63 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.26 (dd, J = 4.8, 9.2 Hz, 1H), 3.96 ( t, J = 6.4 Hz, 2H), 3.89 (s, 5H), 3.65 - 3.50 (m, 4H), 3.00 ( t, J = 5.6 Hz, 2H), 2.97 - 2.81 (m, 4H), 2.72 - 2.58 (m, 4H), 2.34 - 2.26 (m, 1H), 2.20 - 2.10 (m, 1H), 1.90 (s, 3H), 1.79 - 1.68 (m, 2H), 1.55 (d, J = 2.0 Hz, 2H), 1.45 (d, J = 6.8 Hz, 2H), 1.31 (s, 8H). Example 31. Preparation of Compound 160a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[(3R)-1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-o xo-ethyl]pyrrolidin-3- yl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl (3S)-3-[(E)-3-ethoxy-3-oxo-prop-1- enyl]pyrrolidine-1-carboxylate To a solution of tert-butyl (3R)-3-formylpyrrolidine-1-carboxylate (4 g, 20.08 mmol, 1 equiv.) in THF (60 mL) was added NaH (1.20 g, 30.11 mmol, 60% purity, 1.5 equiv.) stirred at 0 °C for 10 min under N2 atmosphere, then added ethyl 2-diethoxyphosphorylacetate (9.00 g, 40.15 mmol, 7.97 mL, 2 equiv.) the mixture was stirred at 0 °C for 2 hours under N 2 atmosphere. The reaction mixture was quenched by slowly adding water (30 mL) at 0 °C. The resulting mixture was then extracted with ethyl acetate (30 mL × 3). The combined organic layers were washed with water (30 mL × 3), filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 28% ethyl acetate / petroleum ether) to give tert-butyl (3S)-3-[(E)-3-ethoxy-3-oxo-prop-1-enyl]pyrrolidine-1- carboxylate (4.5 g, 16.7 mmol, 83.2% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 6.88 - 6.82 (m, 1H), 5.95 - 5.91 (m, 1H), 5.77 - 5.74 (m, 1H), 4.14 - 4.09 (m, 2H), 3.48 - 3.43 (m, 1H), 3.38 - 3.29 (m, 2H), 3.25 - 3.16 (m, 1H), 3.08 - 2.94 (m, 2H), 1.80 - 1.70 (m, 1H), 1.39 (s, 9H), 1.22 - 1.19 (t, J = 7.1 Hz, 3H). Step B. Procedure for preparation of tert-butyl (3R)-3-(3-ethoxy-3-oxo-propyl) pyrrolidine-1-carboxylate To a solution of tert-butyl (3S)-3-[(E)-3-ethoxy-3-oxo-prop-1-enyl]pyrrolidine-1-carboxy late (4.5 g, 16.71 mmol, 1 equiv.) in EtOH (60 mL) was added PtO2 (1.14 g, 5.01 mmol, 0.3 equiv.) in EtOH 5 mL under H 2 atmosphere (15 Psi). The mixture was stirred at 25 °C for 12 hours. The mixture was filtered and washed with EtOH 15 mL (5 mL × 3), concentrated under reduced pressure to give a residue. The residue used in the next step without purification. The compound tert-butyl (3R)-3-(3-ethoxy-3-oxo-propyl) pyrrolidine-1-carboxylate (3.8 g, crude) was obtained as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ = 4.08 - 4.03 (m, 2H), 3.47 - 3.38 (m, 2H), 3.30 - 3.29 (m, 1H), 3.18 - 3.08 (m, 1H), 2.80 - 2.74 (m, 1H), 2.34 - 2.30 (m, 2H), 2.11 - 2.00 (m, 1H), 1.95 - 1.90 (m, 1H), 1.66 - 1.53 (m, 2H), 1.39 (s, 9H), 1.24 - 1.16 (m, 3H). Step C. Procedure for preparation of tert-butyl (3R)-3-(3-hydroxypropyl)pyrrolidine-1- carboxylate To a solution of tert-butyl (3R)-3-(3-ethoxy-3-oxo-propyl) pyrrolidine-1-carboxylate (3.5 g, 12.90 mmol, 1 equiv.) in THF (40 mL) was added LiAlH4 (489.55 mg, 12.90 mmol, 1 equiv.). The mixture was stirred at 0 °C for 2 hours. The reaction mixture was quenched by addition water (0.49 mL) and 15% NaOH (0.49 mL) at 0°C, and then diluted with water (5 mL) and extracted with ethyl acetate (15 mL × 3). The combined organic layers were washed with water (15 mL × 3), concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 40% ethyl acetate / petroleum ether) to give tert-butyl (3R)-3-(3-hydroxypropyl)pyrrolidine-1-carboxylate (2.6 g, 11.34 mmol, 87.9% yield) as a white solid. MS (ESI) m/z: 130.5 [M+H-100] + Step D. Procedure for preparation of tert-butyl (3R)-3-[3-(3-bromo-2-methyl- phenoxy)propyl]pyrrolidine-1-carboxylate A mixture of tert-butyl (3R)-3-(3-hydroxypropyl)pyrrolidine-1-carboxylate (2.6 g, 11.34 mmol, 1 equiv.), 3-bromo-2-methyl-phenol (2.54 g, 13.61 mmol, 1.2 equiv.), 2-(tributyl-λ5- phosphanylidene)acetonitrile (3.28 g, 13.61 mmol, 1.2 equiv.) in toluene (30 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 120 °C for 12 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 17% ethyl acetate / petroleum ether) to give tert-butyl (3R)-3-[3-(3-bromo-2-methyl- phenoxy)propyl]pyrrolidine-1-carboxylate (3.4 g, 8.0 mmol, 70.7% yield, 94% purity) as a white solid. MS (ESI) m/z: 300.2 [M+H-100] + Step E. Procedure for preparation of (3R)-3-[3-(3-bromo-2-methyl-phenoxy) propyl] pyrrolidine To a solution of tert-butyl (3R)-3-[3-(3-bromo-2-methyl-phenoxy)propyl]pyrrolidine-1- carboxylate (3.4 g, 8.54 mmol, 1 equiv.) in HCl/dioxane (4 M) (10 mL). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was used in the next step without purification. The compound (3R)- 3-[3-(3-bromo-2-methyl-phenoxy) propyl] pyrrolidine (2.8 g, crude) was obtained as a white solid. Step F. Procedure for preparation of ethyl 2-[(3R)-3-[3-(3-bromo-2-methyl- phenoxy)propyl]pyrrolidin-1-yl]acetate A mixture of (3R)-3-[3-(3-bromo-2-methyl-phenoxy)propyl]pyrrolidine (2.8 g, 8.37 mmol, 1 equiv., HCl), ethyl 2-bromoacetate (1.12 g, 6.69 mmol, 740.22 µL, 0.8 equiv.), K 2 CO 3 (3.47 g, 25.10 mmol, 3 equiv.) in CH 3 CN (40 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 40 °C for 1 hour under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~17% ethyl acetate/petroleum ether) to give a crude. The crude was purified by prep-SFC to give ethyl 2-[(3R)-3-[3-(3-bromo-2-methyl- phenoxy)propyl]pyrrolidin-1-yl]acetate (2 g, 4.8 mmol, 57.8% yield, 93% purity) as a white solid. MS (ESI) m/z: 384.2 [M+H] + Step G. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(3R)-1-(2-ethoxy-2 -oxo-ethyl)pyrrolidin-3- yl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate Ethyl 2-[(3R)-3-[3-(3-bromo-2-methyl-phenoxy)propyl]pyrrolidin-1-y l]acetate (300 mg, 780.63 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (525.99 mg, 858.69 µmol, 1.1 equiv.), Ad2nBuP Pd G3(cataCXium® A Pd G3 ) (113.70 mg, 156.13 µmol, 0.2 equiv.) and K 2 CO 3 (1.5 M, 780.63 µL, 1.5 equiv.) were taken up into a microwave tube in 1,4-dioxane (4 mL) .The sealed tube was heated at 100 °C for 1 hour under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 55% ethyl acetate / petroleum ether) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[3-[(3R)-1-(2-ethoxy-2-oxo-ethyl)pyrr olidin-3-yl]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (450 mg, 569.6 µmol, 72.9% yield) as a white solid. MS (ESI) m/z: 790.4 [M+H] + Step H. Procedure for preparation of 2-[(3R)-3-[3-[3-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-2- methyl-phenoxy] propyl] pyrrolidin-1-yl] acetic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[(3R)-1-(2-ethoxy-2-oxo-ethyl)pyrrolidin-3-yl]pr opoxy]-2-methyl- phenyl]pyridine-2-carboxylate (450 mg, 569.64 µmol, 1 equiv.) , LiOH•H 2 O (71.71 mg, 1.71 mmol, 3 equiv.) in THF (8 mL) and H 2 O (2 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 25 °C for 3 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to remove THF, then added HCl (1 mol/L) and adjusted pH to 5. The mixture was filtered and used in the next step without purification. The compound 2-[(3R)-3-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy] propyl] pyrrolidin-1- yl] acetic acid (360 mg, crude) was obtained as a white solid. MS (ESI) m/z: 762.8 [M+H] + Step I. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(3R)-1-[2-[[3-(2,6-dio xo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]pyrrolidin-3-yl]propoxy]-2-m ethyl-phenyl]pyridine-2- carboxylate A mixture of 2-[(3R)-3-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy] propyl] pyrrolidin-1- yl] acetic acid (120 mg, 157.50 µmol, 1 equiv.), 3-(6-amino-1-methyl-indazol-3-yl)piperidine- 2,6-dione (48.81 mg, 188.99 µmol, 1.2 equiv.), DIPEA (61.07 mg, 472.49 µmol, 82.30 µL, 3 equiv.), HATU (59.88 mg, 157.50 µmol, 1.5 equiv.) in DMF (3 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 3 hours under N2 atmosphere. The reaction mixture was quenched by slow addition of water 30 mL at 0 °C, and then extracted with ethyl acetate (30 mL × 3). The combined organic layers were washed with water (30 mL × 3), filtered, and concentrated under reduced pressure to give a residue. The residue was used in the next step without purification. The compound tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(3R)- 1-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]pyrrolid in-3-yl]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (150 mg, crude) was obtained as a white solid. MS (ESI) m/z: 1002.6 [M+H] + Step J. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[(3R)-1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]pyrrolidin-3-yl]propoxy]-2-methyl-phen yl]pyridine-2-carboxylic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[(3R)-1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo- ethyl]pyrrolidin-3-yl]propoxy]-2-methyl-phenyl]pyridine-2-ca rboxylate (150 mg, 149.67 µmol, 1 equiv.) in TFA (3 mL) and DCM (3 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 40 °C for 2 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep- HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3- [3-[(3R)-1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6 -yl]amino]-2-oxo- ethyl]pyrrolidin-3-yl]propoxy]-2-methyl-phenyl]pyridine-2-ca rboxylic acid (54.2 mg, 56.2 µmol, 37.5% yield, 98.0% purity) as a white solid. MS (ESI) m/z: 946.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 13.36 - 12.29 (m, 1H), 10.88 (s, 1H), 10.00 (s, 1H), 8.14 (s, 1H), 8.06 - 8.00 (m, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 8.4 Hz, 2H), 7.49 - 7.42 (m, 3H), 7.39 - 7.32 (m, 2H), 7.19 (d, J = 9.2 Hz, 1H), 7.12 - 7.05 (m, 1H), 6.95 (d, J = 9.2 Hz, 1H), 6.88 (d, J = 8.4 Hz, 1H), 6.62 (d, J = 7.2 Hz, 1H), 4.99 - 4.96 (m, 2H), 4.33 - 4.30 (m, 9.8 Hz, 1H), 4.02 - 3.93 (m, 3H), 3.92 -3.90 (m, 4H), 3.05 - 2.95 (m, 5H), 2.84 - 2.79 (m, 2H), 2.66 - 2.62 (m, 2H), 2.35 - 2.32 (m, 2H), 2.24 - 2.15 (m, 2H), 2.05 - 1.98 (m, 1H), 1.90 (s, 3H), 1.77 - 1.69 (m, 2H), 1.57 - 1.51 (m, 2H), 1.47 - 1.40 (m, 1H). Example 32. Preparation of Compound 160b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((3S)-1-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino )-2-oxoethyl)pyrrolidin-3- yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of (R,E)-tert-butyl 3-(3-ethoxy-3-oxoprop-1-en-1- yl)pyrrolidine-1-carboxylate A mixture of NaH (1.69 g, 42.16 mmol, 60% purity, 2.1 equiv.) in THF (40 mL) was purged with N2 for three times. To the mixture was slowly added ethyl 2-(diethoxyphosphoryl)acetate (9.00 g, 40.15 mmol, 7.97 mL, 2 equiv.) at 0 °C, and then the mixture was stirred for 0.5 hours under N2 atmosphere. To the resulting solution was added (S)-tert-butyl 3-formylpyrrolidine- 1-carboxylate (4 g, 20.08 mmol, 1 equiv.) at 0 °C. The solution was warm to 25 °C and stirred for 16 hours under N 2 atmosphere. The reaction mixture was quenched by addition saturated NH 4 Cl (40 mL) under 0°C. The mixture was extracted with ethyl acetate (20 mL × 3). The combined organic layers were washed with brine (20 mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give (R,E)-tert-butyl 3-(3-ethoxy-3-oxoprop-1-en-1- yl)pyrrolidine-1-carboxylate (2.4 g, 9.0 mmol, 45.1% yield) as a white oil. 1 H NMR (400 MHz, DMSO-d6) δ = 6.85 (dd, J = 7.6, 15.6 Hz, 1H), 5.93 (dd, J = 0.8, 15.6 Hz, 1H), 4.11 (q, J = 7.2 Hz, 2H), 3.45 (dd, J = 7.2, 10.0 Hz, 1H), 3.39 - 3.31 (m, 3H), 3.26 - 3.15 (m, 1H), 2.07 - 1.96 (m, 1H), 1.82 - 1.67 (m, 1H), 1.39 (s, 9H), 1.24 - 1.17 (m, 3H) Step B. Procedure for preparation of (S)-tert-butyl 3-(3-ethoxy-3-oxopropyl)pyrrolidine- 1-carboxylate A mixture of (R,E)-tert-butyl 3-(3-ethoxy-3-oxoprop-1-en-1-yl)pyrrolidine-1-carboxylate (2.44 g, 9.06 mmol, 1 equiv.) PtO2 (1.44 g, 6.34 mmol, 0.7 equiv.) in EtOH (25 mL) was degassed and purged with H2 three times, and then the mixture was stirred at 25 °C for 15 hours under H 2 (15 Psi) atmosphere. The mixture was filtered by EtOH (30 mL), and the filtrate was concentrated under reduced pressure to give (S)-tert-butyl 3-(3-ethoxy-3- oxopropyl)pyrrolidine-1-carboxylate (1.41 g, crude) as a black oil (The filter cake was immersed in EtOH). 1 H NMR (400 MHz, DMSO-d 6 ) δ = 4.05 (q, J = 7.2 Hz, 2H), 3.44 - 3.28 (m, 4H), 2.34 - 2.27 (m, 2H), 2.11 - 1.99 (m, 1H), 1.97 - 1.86 (m, 1H), 1.59 (dq, J = 3.2, 7.6 Hz, 2H), 1.49 - 1.41 (m, 1H), 1.38 (s, 9H), 1.18 (t, J = 7.2 Hz, 3H) Step C. Procedure for preparation of (S)-tert-butyl 3-(3-hydroxypropyl)pyrrolidine-1- carboxylate A mixture of (S)-tert-butyl 3-(3-ethoxy-3-oxopropyl)pyrrolidine-1-carboxylate (1.41 g, 5.20 mmol, 1 equiv.) in THF (25 mL) was slowly added LiAlH 4 (197.20 mg, 5.20 mmol, 1 equiv.) at 0 °C, then the mixture was stirred at 25 ℃ for 5 hours. The mixture was quenched by 6.5 g Na 2 SO 4 .10 H 2 O. The solution was poured onto ice-water (30 mL) slowly and acidified to pH = 4-5 with 1 M HCl. The resulting mixture was extracted with EtOAc 60 mL (20 mL × 3), washed with brine 40 mL (20 mL × 2), dried by sodium sulfate, filtered, and concentrated under reduced pressure to give (S)-tert-butyl 3-(3-hydroxypropyl)pyrrolidine-1-carboxylate (1.08 g, 4.07 mmol, 78.31% yield, 86.4% purity) as a white oil. Step D. Procedure for preparation of (S)-tert-butyl 3-(3-(3-bromo-2- methylphenoxy)propyl)pyrrolidine-1-carboxylate A mixture of (S)-tert-butyl 3-(3-hydroxypropyl)pyrrolidine-1-carboxylate (1.08 g, 4.71 mmol, 1 equiv.), 3-bromo-2-methylphenol (1.06 g, 5.65 mmol, 1.2 equiv.), 2-(tributyl-λ 5 - phosphanylidene)acetonitrile (1.36 g, 5.65 mmol, 1.2 equiv.) in toluene (10 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 120 °C for 18 hours under N2 atmosphere. The mixture concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~7% ethyl acetate / petroleum ether) to give (S)-tert-butyl 3-(3-(3-bromo-2-methylphenoxy)propyl)pyrrolidine-1-carboxyla te (1.05 g, 2.48 mmol, 52.61% yield, 94.0% purity) as a yellow oil. Step E. Procedure for preparation of (S)-3-(3-(3-bromo-2- methylphenoxy)propyl)pyrrolidine A mixture of (S)-tert-butyl 3-(3-(3-bromo-2-methylphenoxy)propyl)pyrrolidine-1-carboxyla te (905 mg, 2.27 mmol, 1 equiv.) in HCl/EtOAc (10 mL) was stirred at 25 °C for 16 hours. The mixture was concentrated under the reduced pressure to give (S)-3-(3-(3-bromo-2- methylphenoxy)propyl)pyrrolidine (685 mg, 2.2 mmol, 99.2% yield, 98.2% purity) a colorless oil. MS (ESI) m/z: 300.1 [M+H] + Step F. Procedure for preparation of (S)-ethyl 2-(3-(3-(3-bromo-2- methylphenoxy)propyl)pyrrolidin-1-yl)acetate A mixture of (S)-3-(3-(3-bromo-2-methylphenoxy)propyl)pyrrolidine (685 mg, 2.30 mmol, 1 equiv.), ethyl 2-bromoacetate (383.60 mg, 2.30 mmol, 254.04 µL, 1 equiv.), K 2 CO 3 (952.40 mg, 6.89 mmol, 3 equiv.) in CH3CN (1 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 60 °C for 6 hours under N 2 atmosphere. The mixture concentrated under the reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~75% ethyl acetate / petroleum ether) to give (S)-ethyl 2-(3-(3-(3-bromo-2-methylphenoxy)propyl)pyrrolidin-1-yl)acet ate (903 mg, 2.30 mmol, 99.94% yield, 97.7% purity) as a blue oil. MS (ESI) m/z: 384.1 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 7.16 - 7.12 (m, 1H), 7.11 - 7.06 (m, 1H), 6.95 (d, J = 8.0 Hz, 1H), 4.10 (q, J = 7.2 Hz, 2H), 3.96 (t, J = 6.4 Hz, 2H), 3.47 (d, J = 4.4 Hz, 2H), 2.96 (t, J = 8.4 Hz, 1H), 2.81 (q, J = 7.6 Hz, 1H), 2.73 - 2.66 (m, 1H), 2.40 - 2.31 (m, 1H), 2.23 (s, 3H), 2.20 - 2.11 (m, 1H), 2.01 - 1.91 (m, 1H), 1.77 - 1.65 (m, 2H), 1.54 - 1.45 (m, 2H), 1.44 - 1.34 (m, 1H), 1.19 (t, J = 7.2 Hz, 3H) Step H. Procedure for preparation of (S)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-methoxy-2-oxo ethyl)pyrrolidin-3- yl)propoxy)-2-methylphenyl)picolinate (S)-ethyl 2-(3-(3-(3-bromo-2-methylphenoxy)propyl)pyrrolidin-1-yl)acet ate (314 mg, 817.06 µmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pic olinate (550.53 mg, 898.76 µmol, 1.1 equiv.), Ad 2 nBuP Pd G 3 (119.01 mg, 163.41 µmol, 0.2 equiv.), KF (1.5 M, 817.06 µL, 1.5 equiv.) in dioxane (10 mL) and were taken up into a microwave tube in dioxane (10 mL), The sealed tube was heated at 100 °C for 2 hours under microwave. The reaction mixture was partitioned between H 2 O (15 mL) and ethyl acetate 60 mL (20 mL × 3). The organic phase was separated, washed with aqueous NaCl 45 mL (15 mL × 3), dried over (Na 2 SO 4 ), filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~74% ethyl acetate / petroleum ether) to give (S)-tert- butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2- methoxy-2-oxoethyl)pyrrolidin-3-yl)propoxy)-2-methylphenyl)p icolinate (368 mg, 465.83 µmol, 57.01% yield) as a yellow solid. MS (ESI) m/z: 790.3 [M+H] + Step I. Procedure for preparation of (S)-2-(3-(3-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)propyl)pyrrolidin-1-yl)acetic acid A mixture of (S)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-methoxy-2-oxoethyl)pyrrolidin-3-yl)p ropoxy)-2- methylphenyl)picolinate (368 mg, 465.83 µmol, 1 equiv.), LiOH.H 2 O (1 M, 1.40 mL, 3 equiv.) in THF (6 mL)was stirred at 25 °C for 6 hours. The mixture was concentrated and diluted with H2O (2 mL), then the pH of the mixture was adjusted to 3 with 1 M HCl. Then the mixture was filtered and concentrated to give (S)-2-(3-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)propyl)pyrrolidin-1-yl)acetic acid (313 mg, 390.67 µmol, 83.86% yield, 95.1% purity) as a white solid. MS (ESI) m/z: 762.6 [M+H] + Step J. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3S)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)pyrrolidin-3-yl)propoxy)-2 -methylphenyl)picolinate A mixture of (S)-2-(3-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)pyrrolidin-1- yl)acetic acid (93 mg, 122.06 µmol, 1 equiv.), 3-(6-amino-1-methyl-1H-indazol-3- yl)piperidine-2,6-dione (37.83 mg, 146.47 µmol, 1.2 equiv.) in pyridine (2 mL) was added EDCI (35.10 mg, 183.09 µmol, 1.5 equiv.). The resulting mixture was stirred at 25 °C for 15 hours. The reaction mixture was partitioned between H 2 O (10 mL) and DCM (10 mL × 3). The organic phase was separated, washed with aqueous NaCl (10 mL × 2) , dried over (Na 2 SO 4 ), filtered, and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3S)- 1-(2-((3-(2,6-dioxopiperidin-3- yl)-1-methyl-1H-indazol-6-yl)amino)-2-oxoethyl)pyrrolidin-3- yl)propoxy)-2- methylphenyl)picolinate (113 mg, crude) as a red solid. MS (ESI) m/z: 1002.4 [M+H] + Step K. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3S)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)pyrrolidin-3-yl)propoxy)-2 -methylphenyl)picolinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-((3S)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-6-yl)amino)-2- oxoethyl)pyrrolidin-3-yl)propoxy)-2-methylphenyl)picolinate (113 mg, 112.75 µmol, 1 equiv.) in DCM (3 mL) and TFA (1 mL) was stirred at 40 °C for 69 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((3S)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1 -methyl-1H-indazol-6-yl)amino)- 2-oxoethyl)pyrrolidin-3-yl)propoxy)-2-methylphenyl)picolinic acid (57.2 mg, 55.8 µmol, 49.5% yield, 92.4% purity) as a yellow solid MS (ESI) m/z: 946.7 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 12.92 - 12.81 (m, 1H), 10.89 (s, 1H), 10.12 (d, J = 8.0 Hz, 1H), 8.05 - 8.01 (m, 1H), 7.98 (s, 1H), 7.79 (d, J = 7.6 Hz, 1H), 7.69 (d, J = 8.8 Hz, 1H), 7.63 (d, J = 6.4 Hz, 1H), 7.49 - 7.42 (m, 3H), 7.40 - 7.33 (m, 2H), 7.14 - 7.08 (m, 2H), 6.97 (d, J = 8.8 Hz, 1H), 6.89 (d, J = 8.4 Hz, 1H), 6.64 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.33 (dd, J = 5.2, 10.0 Hz, 1H), 4.27 (dd, J = 3.2, 4.4 Hz, 2H), 3.98 ( t, J = 6.0 Hz, 2H), 3.93 (s, 3H), 3.92 - 3.88 (m, 2H), 3.76 - 3.57 (m, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.74 - 2.59 (m, 4H), 2.36 - 2.30 (m, 2H), 2.27 - 2.00 (m, 3H), 1.91 (s, 3H), 1.81 - 1.74 (m, 2H), 1.66 - 1.59 (m, 2H) Example 33. Preparation of Compound 162a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[(1S)-3-[1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-o xo-ethyl]-4-piperidyl]-1- methyl-propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 4-[3-[methoxy(methyl)amino]-3-oxo- propyl]piperidine-1-carboxylate To a solution of 3-(1-tert-butoxycarbonyl-4-piperidyl)propanoic acid (4 g, 15.54 mmol, 1 equiv.) and N-methoxymethanamine (2.27 g, 23.32 mmol, 1.5 equiv., HCl) in DMF (10 mL) was added DIEA (6.03 g, 46.63 mmol, 8.12 mL, 3 equiv.) and HATU (8.87 g, 23.32 mmol, 1.5 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was quenched by addition H 2 O (30 mL), and then extracted with ethyl acetate (30 mL × 3). The combined organic layers were washed with brine (30 mL × 3), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=10/1 to 1/1) to give compound tert-butyl 4-[3-[methoxy(methyl)amino]-3-oxo-propyl]piperidine-1-carbox ylate (4.5 g, 14.9 mmol, 96.3% yield) as a yellow oil. 1 H NMR (400 MHz, CDCl 3 ,) δ = 3.69 (s, 3H), 3.18 (s, 3H), 2.68 (t, J = 12.0 Hz, 2H), 2.45 (t, J = 8.0 Hz, 2H), 1.69 (s, 1H), 1.66 (s, 2H), 1.63 - 1.56 (m, 2H), 1.45 (s, 11H), 1.18 - 1.05 (m, 2H). Step B. Procedure for preparation of tert-butyl 4-(3-oxobutyl)piperidine-1-carboxylate A mixture of tert-butyl 4-[3-[methoxy(methyl)amino]-3-oxo-propyl]piperidine-1-carbox ylate (4.5 g, 14.98 mmol, 1 equiv.) in THF (50 mL) was degassed and purged with N2 three times, and then MeMgBr (3 M, 29.96 mL, 6 equiv.) was added dropwise to the mixture and stirred at 0 °C for 4 hours under N2 atmosphere. The reaction mixture was quenched by addition saturated ammonium chloride (50 mL) at 0 °C, and then diluted with water (50 mL) and extracted with CH 2 Cl 2 (50 mL × 3). The combined organic layers were washed with brine (50 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give compound tert-butyl 4-(3-oxobutyl)piperidine-1-carboxylate (3.23 g, 12.65 mmol, 84.44% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ,) δ = 3.90 (d, J = 11.2 Hz, 2H), 2.64 - 2.63 (m, 2H), 2.44 (t, J = 7.2 Hz, 2H), 2.07 (s, 3H), 1.59 (d, J = 12.4 Hz, 2H), 1.41 - 1.35 (m, 12H), 0.98 - 0.86 (m, 2H). Step C. Procedure for preparation of tert-butyl 4-(3-hydroxybutyl)piperidine-1- carboxylate To a solution of tert-butyl 4-(3-oxobutyl)piperidine-1-carboxylate (2 g, 7.83 mmol, 1 equiv.) in EtOH (20 mL) was added NaBH 4 (296.32 mg, 7.83 mmol, 1 equiv.). The mixture was stirred at 0 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to remove EtOH, and then diluted with EtOAc 10 mL and saturated ammonium chloride solution 10 ml and extracted with EtOAc (10 mL × 3). The combined organic layers were washed with brine (10 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give compound tert-butyl 4-(3-hydroxybutyl)piperidine-1-carboxylate (1.8 g, 6.99 mmol, 89.30% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ,) δ = 4.31 (d, J = 4.4 Hz, 1H), 3.90 (d, J = 12.4 Hz, 2H), 3.58 - 3.48 (m, 1H), 2.73 - 2.61 (m, 2H), 1.61 (d, J = 12.4 Hz, 2H), 1.38 (s, 12H), 1.33 - 1.22 (m, 4H), 1.02 (d, J = 6.0 Hz, 3H). Step D. Procedure for preparation of tert-butyl 4-[(3R)-3-hydroxybutyl]piperidine-1- carboxylate This page was used for purification. This tert-butyl 4-(3-hydroxybutyl)piperidine-1- carboxylate (3 g, 11.66 mmol, 1 equiv.) was separated by SFC to give tert-butyl 4-[(3S)-3- hydroxybutyl]piperidine-1-carboxylate (1.2 g, 4.66 mmol, 40.00% yield) as a colorless oil and tert-butyl 4-[(3R)-3-hydroxybutyl]piperidine-1-carboxylate (1.08 g, 4.20 mmol, 36.00% yield) as a pink oil. 1 H NMR (400 MHz, DMSO-d6,) δ = 4.29 (d, J = 4.4 Hz, 1H), 3.90 (d, J = 12.4 Hz, 2H), 3.61 - 3.46 (m, 1H), 2.66 - 2.65 (m, 2H), 1.61 (d, J = 12.8 Hz, 2H), 1.38 (s, 9H), 1.35 - 1.23 (m, 4H), 1.21 - 1.11 (m, 1H), 1.02 (d, J = 6.0 Hz, 3H), 0.99 - 0.85 (m, 2H). Step E. Procedure for preparation of tert-butyl 4-[(3S)-3-(3-bromo-2-methyl- phenoxy)butyl]piperidine-1-carboxylate To a solution of tert-butyl 4-[(3R)-3-hydroxybutyl]piperidine-1-carboxylate (1.1 g, 4.27 mmol, 1 equiv.) and 3-bromo-2-methyl-phenol (959.27 mg, 5.13 mmol, 1.2 equiv.) in toluene (10 mL) was added 2-(tributyl-λ5-phosphanylidene)acetonitrile (1.55 g, 6.41 mmol, 1.5 equiv.). The mixture was stirred at 120 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=0/1 to 10/1) to give compound tert-butyl 4-[(3S)-3-(3-bromo-2- methyl-phenoxy)butyl]piperidine-1-carboxylate (1.86 g, crude) as a colorless oil. 1 H NMR (400 MHz, CDCl 3 ,) δ = 7.13 (d, J = 8.0 Hz, 1H), 6.98 (t, J = 8.0 Hz, 1H), 6.76 (d, J = 8.4 Hz, 1H), 4.08 (d, J = 12.4 Hz, 2H), 2.68 (t, J = 11.2 Hz, 2H), 2.30 (s, 3H), 1.70 - 1.58 (m, 4H), 1.46 (s, 9H), 1.29 (d, J = 6.4 Hz, 5H), 1.15 - 1.07 (m, 2H), 0.94 - 0.81 (m, 2H). Step F. Procedure for preparation of 4-[(3S)-3-(3-bromo-2-methyl- phenoxy)butyl]piperidine A solution of tert-butyl 4-[(3S)-3-(3-bromo-2-methyl-phenoxy)butyl]piperidine-1-carbo xylate (1.86 g, 4.36 mmol, 1 equiv.) in HCl/EtOAc (4 M, 10 mL, 9.17 equiv.) was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=1/1 to DCM: MeOH = 10:1) to give compound 4-[(3S)-3-(3-bromo-2-methyl- phenoxy)butyl]piperidine (1.55 g, crude) as a light yellow oil. Step G. Procedure for preparation of ethyl 2-[4-[(3S)-3-(3-bromo-2-methyl- phenoxy)butyl]-1-piperidyl]acetate To a solution of 4-[(3S)-3-(3-bromo-2-methyl-phenoxy)butyl]piperidine (0.8 g, 2.45 mmol, 1 equiv.) and ethyl 2-bromoacetate (409.48 mg, 2.45 mmol, 271.18 µL, 1 equiv.) in CH3CN (10 mL) was added K 2 CO 3 (1.02 g, 7.36 mmol, 3 equiv.). The mixture was stirred at 60 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=0/1 to DCM: MeOH = 10/1) to give compound ethyl 2-[4-[(3S)-3-(3-bromo-2-methyl- phenoxy)butyl]-1-piperidyl]acetate (575 mg, 1.3 mmol, 56.8% yield) as a yellow oil. MS (ESI) m/z: 414.1 [M+H] + 1 H NMR (400 MHz, CDCl3,) δ = 7.13 (d, J = 8.0 Hz, 1H), 6.98 (t, J = 8.0 Hz, 1H), 6.76 (d, J = 8.0 Hz, 1H), 4.35 - 4.27 (m, 1H), 4.24 - 4.15 (m, 2H), 3.21 (s, 2H), 2.94 (d, J = 10.8 Hz, 2H), 2.30 (s, 3H), 2.18 - 2.15 (m, 2H), 1.75 - 1.62 (m, 4H), 1.40 - 1.25 (m, 11H). Step H. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[(1S)-3-[1-(2-ethoxy-2 -oxo-ethyl)-4-piperidyl]-1- methyl-propoxy]-2-methyl-phenyl]pyridine-2-carboxylate Ethyl 2-[4-[(3S)-3-(3-bromo-2-methyl-phenoxy)butyl]-1-piperidyl]ac etate (575 mg, 1.39 mmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (854.14 mg, 1.39 mmol, 1 equiv.), [2-(2-aminophenyl)phenyl]palladium(1+);bis(1- adamantyl)-butyl-phosphane;methanesulfonate (203.10 mg, 278.88 µmol, 0.2 equiv.), and KF (1.5 M, 2.79 mL, 3 equiv.) were taken up into a microwave tube in dioxane (5 mL). The sealed tube was heated at 100 °C for 60 minutes under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=0/1 to 1/1) to give compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[(1S)-3-[1-(2- ethoxy-2-oxo-ethyl)-4-piperidyl]-1-methyl-propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (0.616 g, 753.03 µmol, 54.00% yield) as a yellow oil. MS (ESI) m/z: 818.6 [M+H] + 1 H NMR (400 MHz, DMSO-d6,) δ = 12.86 (s, 1H), 8.02 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.60 (d, J = 7.6 Hz, 1H), 7.49 - 7.42 (m, 3H), 7.41 - 7.31 (m, 2H), 7.13 - 7.04 (m, 1H), 6.97 - 6.88 (m, 2H), 6.54 (d, J = 7.2 Hz, 1H), 4.97 (s, 2H), 4.42 - 4.41 (m, 1H), 4.07 - 4.01 (m, 4H), 3.87 (t, J = 5.2 Hz, 2H), 3.30 (s, 4H), 3.13 (s, 2H), 3.03 (t, J = 6.0 Hz, 3H), 2.77 (d, J = 9.2 Hz, 2H), 2.12 - 2.04 (m, 2H), 1.99 (s, 4H), 1.85 (s, 3H), 1.65 - 1.53 (m, 4H), 1.00 (s, 9H). Step I. Procedure for preparation of 2-[4-[(3S)-3-[3-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-2- methyl-phenoxy]butyl]-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[(1S)-3-[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]-1- methyl-propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (0.616 g, 753.03 µmol, 1 equiv.) in THF (3 mL) and H 2 O (3 mL) was added LiOH•H 2 O (158.00 mg, 3.77 mmol, 5 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was quenched by addition water 10 mL, and then filtered and concentrated under reduced pressure to give compound 2-[4-[(3S)-3-[3-[6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -2-tert-butoxycarbonyl-3- pyridyl]-2-methyl-phenoxy]butyl]-1-piperidyl]acetic acid (640 mg, crude) as a yellow solid. MS (ESI) m/z: 790.4 [M+H] + Step J. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[(1S)-3-[1-[2-[[3-(2,6 -dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-4-piperidyl]-1-methyl-propo xy]-2-methyl- phenyl]pyridine-2-carboxylate To a solution of 2-[4-[(3S)-3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]butyl]-1- piperidyl]acetic acid (60 mg, 75.95 µmol, 1 equiv.) and 3-(6-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (23.54 mg, 91.14 µmol, 1.2 equiv.) in DMF (1.0 mL) was added HATU (43.32 mg, 113.93 µmol, 1.5 equiv.) and DIEA (49.08 mg, 379.76 µmol, 66.15 µL, 5 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was quenched by addition of water (10 mL), filtered and concentrated under reduced pressure to give tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[(1S)-3-[1-[2-[[3-(2,6- dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl] -4-piperidyl]-1-methyl- propoxy]-2-methyl-phenyl]pyridine-2-carboxylate (120 mg, crude) as a pink solid. MS (ESI) m/z: 1030.9 [M+H] + Step K. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[(1S)-3-[1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]-1-methyl-propoxy]-2-meth yl-phenyl]pyridine-2- carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[(1S)-3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methy l-indazol-6-yl]amino]-2-oxo- ethyl]-4-piperidyl]-1-methyl-propoxy]-2-methyl-phenyl]pyridi ne-2-carboxylate (120 mg, 116.48 µmol, 1 equiv.) in DCM (1 mL) was added TFA (13.28 mg, 116.48 µmol, 8.62 µL, 1 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give compound 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3- [(1S)-3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6 -yl]amino]-2-oxo-ethyl]-4- piperidyl]-1-methyl-propoxy]-2-methyl-phenyl]pyridine-2-carb oxylic acid (24.5 mg, 24.0 µmol, 20.6% yield, 95.2% purity) as a white solid. MS (ESI) m/z: 974.4 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ,) δ = 10.89 (s, 1H), 9.84 (s, 1H), 8.06 - 7.99 (m, 2H), 7.78 (d, J = 7.6 Hz, 1H), 7.62 (d, J = 8.4 Hz, 2H), 7.49 - 7.41 (m, 3H), 7.40 - 7.31 (m, 2H), 7.23 - 7.18 (m, 1H), 7.11 - 7.04 (m, 1H), 6.97 - 6.85 (m, 2H), 6.61 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.46 - 4.37 (m, 1H), 4.35 - 4.28 (m, 1H), 3.91 (s, 5H), 3.12 (s, 2H), 3.02 (br t, J = 5.6 Hz, 2H), 2.86 (d, J = 11.2 Hz, 2H), 2.69 - 2.59 (m, 2H), 2.38 - 2.27 (m, 2H), 2.19 - 2.12 (m, 2H), 1.88 (s, 3H), 1.72 - 1.59 (m, 4H), 1.26 - 1.23 (m, 8H). Example 34. Preparation of Compound 163 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(4-(4-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin -1-yl)butyl)phenoxy)-2- methylphenyl)picolinic acid Step A. Procedure for preparation of ethyl 4-(4-hydroxyphenyl)butanoate A mixture of 4-(4-hydroxyphenyl)butanoic acid (900 mg, 4.99 mmol, 1 equiv.), SOCl2 (891.28 mg, 7.49 mmol, 543.47 µL, 1.5 equiv.), in EtOH (10 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 90 °C for 10 h under N 2 atmosphere. The mixture was concentrated under reduced pressure to give the crude compound ethyl 4-(4- hydroxyphenyl)butanoate (1.24 g, crude) as a yellow oil. The residue was used for next step without other purification. 1 H NMR (400 MHz, DMSO-d6) δ = 9.15 (s, 1H), 6.96 (d, J = 8.4 Hz, 2H), 6.67 (d, J = 8.4 Hz, 2H), 4.04 (q, J = 7.2 Hz, 2H), 2.46 (t, J = 7.6 Hz, 2H), 2.25 (t, J = 7.6 Hz, 2H), 1.76 (quin, J = 7.6 Hz, 2H), 1.17 (t, J = 7.2 Hz, 3H) Step B. Procedure for preparation of ethyl 4-(4-(3-bromo-2- methylphenoxy)phenyl)butanoate A mixture of 1,3-dibromo-2-methyl-benzene (4.18 g, 16.71 mmol, 3 equiv.), ethyl 4-(4- hydroxyphenyl)butanoate (1.16 g, 5.57 mmol, 1 equiv.), CuI (530.42 mg, 2.79 mmol, 0.5 equiv.), Cs 2 CO 3 (2.18 g, 6.68 mmol, 1.2 equiv.) and 2,2,6,6-tetramethylheptane-3,5-dione (256.62 mg, 1.39 mmol, 286.72 µL, 0.25 equiv.) in NMP (12.4 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 120 °C for 16 h under N2 atmosphere. The mixture was diluted with H2O (10 mL) and extracted with ethyl acetate (5 mL × 3). The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 5% ethyl acetate / petroleum ether) to give ethyl 4-(4-(3-bromo-2-methylphenoxy)phenyl)butanoate (1.6 g, 4.2 mmol, 76.1% yield) as a colorless oil 1 H NMR (400 MHz, DMSO-d6) δ = 7.41 (d, J = 8.0 Hz, 1H), 7.20 - 7.12 (m, 3H), 6.89 - 6.83 (m, 3H), 4.04 (q, J = 7.2 Hz, 2H), 2.56 (t, J = 7.6 Hz, 2H), 2.31 - 2.27 (m, 2H), 2.26 (s, 3H), 1.80 (m, J = 7.6 Hz, 2H), 1.17 (t, J = 7.2 Hz, 3H) Step C. Procedure for preparation of 4-(4-(3-bromo-2-methylphenoxy)phenyl)butan-1-ol To a solution of ethyl 4-[4-(3-bromo-2-methylphenoxy)phenyl]butanoate (600 mg, 1.59 mmol, 1 equiv.) in THF (6 mL) was added LiAlH 4 (54.33 mg, 1.43 mmol, 0.9 equiv.) under 0 °C at N2 atmosphere, The mixture was stirred at 0 °C for 2 h. The reaction mixture was quenched by addition Na2SO4•10H2O (1 g) under 0 °C and N2 atmosphere, and then filtered and concentrated under reduced pressure to give a residue. The residue was used for next step without other purification. The compound 4-(4-(3-bromo-2-methylphenoxy)phenyl)butan-1-ol (861 mg, crude) was obtained as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.43 (d, J = 7.8 Hz, 1H), 7.21 - 7.15 (m, 3H), 6.89 - 6.86 (m, 3H), 4.40 (t, J = 5.2 Hz, 1H), 3.45 - 3.40 (m, 2H), 2.57 (t, J = 7.6 Hz, 2H), 2.29 (s, 3H), 1.63 - 1.56 (m, 2H), 1.48 - 1.41 (m, 2H) Step D. Procedure for preparation of 4-(4-(3-bromo-2-methylphenoxy)phenyl)butanal To a solution of DMSO (802.70 mg, 10.27 mmol, 802.70 µL, 4 equiv.) in DCM (8 mL) was added dropwise to a solution of (COCl)2 (651.98 mg, 5.14 mmol, 449.64 µL, 2 equiv.) in DCM (4 mL) at -70 °C under N 2 atmosphere. The mixture was stirred at -70 °C for 1 h. After which time 4-(4-(3-bromo-2-methylphenoxy)phenyl)butan-1-ol (861 mg, 2.57 mmol, 1 equiv.) in DCM (2 mL) was added dropwise at -70 °C. The solution was stirred for 1 h at -70 °C. Then TEA (1.56 g, 15.41 mmol, 2.14 mL, 6 equiv.) was added into the solution. The solution was stirred at -70 °C for 0.5 h under N 2 atmosphere. The mixture was diluted with H 2 O (10 mL) and extracted with DCM (5 mL × 3). The combined organic layers were filtered and concentrated under reduced pressure to give the crude compound 4-(4-(3-bromo-2- methylphenoxy)phenyl)butanal (624 mg, 1.8 mmol, 72.9% yield) was obtained as a yellow oil, and it was used for next step without other purification. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 9.68 (s, 1H), 7.42 (d, J = 8.0 Hz, 1H), 7.22 - 7.15 (m, 3H), 6.90 - 6.84 (m, 3H), 2.59 - 2.54 (m, 2H), 2.44 (t, J = 7.2 Hz, 2H), 2.27 (s, 3H), 1.82 (m, J = 7.5 Hz, 2H) Step E. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(2-methyl-3-(4-(4-oxobutyl)ph enoxy)phenyl)picolinate A mixture of 4-[4-(3-bromo-2-methyl-phenoxy)phenyl]butanal (400 mg, 1.20 mmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-2-carb oxylate (1.03 g, 1.68 mmol, 1.4 equiv.), [2-(2-aminophenyl)phenyl]palladium(1 + );bis(1-adamantyl)-butyl- phosphane;methanesulfonate (174.84 mg, 240.08 µmol, 0.2 equiv.), K 2 CO 3 (1.5 M, 1.20 mL, 1.5 equiv.) in dioxane (4 mL) and H2O (1 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 100 °C for 1 hour at N2 atmosphere under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 10 ~ 36% ethyl acetate / petroleum ether) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- (2-methyl-3-(4-(4-oxobutyl)phenoxy)phenyl)picolinate (500 mg, 676.6 µmol, 56.3% yield) as a yellow solid. MS (ESI) m/z: 739.5 [M+H] + Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(4-(4-(3-(2,6-dioxopipe ridin-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)butyl)phenoxy)-2-methylphenyl)pi colinate A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(2-methyl-3-(4-(4-oxobutyl)phenoxy)phenyl)picolinate (75 mg, 101.50 µmol, 1 equiv.), 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (39.88 mg, 121.80 µmol, 1.2 equiv.), 4A MS (10 mg, 101.50 µmol, 1.00 equiv.) and HCOOH (97.53 µg, 2.03 µmol, 0.02 equiv.) in EtOH (2.4 mL) and THF (2.4 mL) was stirred at 25 °C for 2 h. NaBH 3 CN (19.14 mg, 304.51 µmol, 3 equiv.) was then added to the mixture, and the ensuing mixture was stirred at 25 °C for 14 h. The mixture was diluted with H2O (2 mL × 2) and extracted with ethyl acetate (2 mL × 3). The combined organic layers were filtered and concentrated under reduced pressure to give the crude compound tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(4-(4-(3-(2,6-dioxopipe ridin-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)butyl)phenoxy)-2-methylphenyl)pi colinate (100 mg, crude) as a yellow oil ,it was used for next step without other purification. MS (ESI) m/z: 1050.3 [M+H] + Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(4-(4-(3-(2,6-dioxopipe ridin-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)butyl)phenoxy)-2-methylphenyl)pi colinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-6-yl)piperazin-1- yl)butyl)phenoxy)-2-methylphenyl)picolinate (80 mg, 76.17 µmol, 1 equiv.), in TFA (0.8 mL) and DCM (0.8 mL) was stirred at 40 °C for 2 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep – HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin-1-yl )butyl)phenoxy)-2- methylphenyl)picolinic acid (27.5 mg, 26.9 µmol, 35.3% yield, 97.0% purity) as a white solid MS (ESI) m/z: 994.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.38 - 12.52 (m, 1H), 10.86 (s, 1H), 8.14 (s, 1H), 8.04 (d, J = 7.6 Hz, 1H), 7.80 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 7.2 Hz, 1H), 7.56 - 7.50 (m, 2H), 7.50 - 7.45 (m, 2H), 7.41 - 7.34 (m, 2H), 7.21 - 7.16 (m, 3H), 7.01 (d, J = 8.8 Hz, 1H), 6.95 - 6.82 (m, 6H), 5.00 (s, 2H), 4.31 - 4.23 (m, 1H), 3.95 - 3.88 (m, 5H), 3.62 - 3.47 (m, 4H), 3.24 - 3.15 (m, 4H), 3.03 (t, J = 5.6 Hz, 2H), 2.70 - 2.58 (m, 6H), 2.33 - 2.26 (m, 1H), 2.21 - 2.12 (m, 1H), 1.91 (s, 3H), 1.66 - 1.50 (m, 4H) Example 35. Preparation of Compound 164b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-((2R)-1- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino )-1-oxopropan-2-yl)piperidin- 4-yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 4-(3-(3-bromo-2- methylphenoxy)propyl)piperidine-1-carboxylate A mixture of 3-bromo-2-methylphenol (1 g, 5.35 mmol, 1 equiv.), tert-butyl 4-(3- hydroxypropyl)piperidine-1-carboxylate (1.30 g, 5.35 mmol, 1 equiv.) and 2-(tributyl-λ5- phosphanylidene)acetonitrile (1.55 g, 6.42 mmol, 1.2 equiv.) in toluene (20 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 120 °C for 2 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~6% ethyl acetate/petroleum ether) to give tert-butyl 4-(3-(3-bromo-2- methylphenoxy)propyl)piperidine-1-carboxylate (2.2 g, 4.8 mmol, 89.8% yield, 90% purity) as a white solid. 1 H NMR (400 MHz, DMSO-d6,) δ = 7.18 - 7.12 (m, 1H), 7.12 - 7.06 (m, 1H), 6.96 (d, J = 8.0 Hz, 1H), 4.02 - 3.86 (m, 4H), 2.67 (s, 2H), 2.23 (s, 3H), 1.81 - 1.71 (m, 2H), 1.65 (d, J = 12.4 Hz, 2H), 1.49 - 1.32 (m, 11H), 1.05 - 0.89 (m, 2H). Step B. Procedure for preparation of 4-(3-(3-bromo-2-methylphenoxy)propyl)piperidine To a solution of tert-butyl 4-(3-(3-bromo-2-methylphenoxy)propyl)piperidine-1-carboxylat e (1.7 g, 3.71 mmol, 90% purity, 1 equiv.) was added HCl/dioxane (4 M, 14.17 mL, 15.27 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give 4-(3-(3-bromo-2-methylphenoxy)propyl)piperidine (1.5 g, crude, HCl) as a white solid. 1 H NMR (400 MHz, CDCl3,) δ = 9.94 - 9.02 (m, 2H), 7.14 (d, J = 8.0 Hz, 1H), 6.99 (t, J = 8.0 Hz, 1H), 6.74 (d, J = 8.0 Hz, 1H), 3.93 (t, J = 6.4 Hz, 2H), 3.50 (d, J = 12.4 Hz, 2H), 2.87 (q, J = 11.6 Hz, 2H), 2.30 (s, 3H), 1.94 (d, J = 13.6 Hz, 2H), 1.86 - 1.78 (m, 2H), 1.75 - 1.62 (m, 2H), 1.56 - 1.49 (m, 2H). Step C. Procedure for preparation of ethyl 2-(4-(3-(3-bromo-2- methylphenoxy)propyl)piperidin-1-yl)propanoate To a solution of 4-(3-(3-bromo-2-methylphenoxy)propyl)piperidine (1.3 g, 3.73 mmol, 1 eq, HCl) in CH 3 CN (25 mL) was added K 2 CO 3 (1.55 g, 11.18 mmol, 3 equiv.) and ethyl 2- bromopropanoate (1.01 g, 5.59 mmol, 728.29 µL, 1.5 equiv.). The mixture was stirred at 80 °C for 5 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~25 % ethyl acetate/petroleum ether) to give ethyl 2-(4-(3-(3-bromo-2-methylphenoxy)propyl)piperidin-1- yl)propanoate (1.2 g, 2.8 mmol, 76.4% yield, 97.8% purity) as a yellow oil. MS (ESI) m/z: 413.9 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ,) δ = 7.14 (d, J = 8.0 Hz, 1H), 6.99 (t, J = 8.0 Hz, 1H), 6.75 (d, J = 8.0 Hz, 1H), 4.21 - 4.15 (m, 2H), 3.93 (t, J = 6.4 Hz, 2H), 3.28 (q, J = 6.8 Hz, 1H), 2.93 (d, J = 10.8 Hz, 2H), 2.34 - 2.17 (m, 5H), 1.86 - 1.77 (m, 2H), 1.73 (d, J = 9.2 Hz, 2H), 1.46 - 1.38 (m, 2H), 1.35 - 1.26 (m, 9H). Step D. Procedure for preparation of (R)-ethyl 2-(4-(3-(3-bromo-2- methylphenoxy)propyl)piperidin-1-yl)propanoate The ethyl 2-(4-(3-(3-bromo-2-methylphenoxy)propyl)piperidin-1-yl)propa noate residue was further separated by SFC to give (R)-ethyl 2-(4-(3-(3-bromo-2- methylphenoxy)propyl)piperidin-1-yl)propanoate (250 mg, 597.78 µmol, 20.54% yield, 98.6% purity) as a yellow oil and (S)-ethyl 2-(4-(3-(3-bromo-2-methylphenoxy)propyl)piperidin-1- yl)propanoate (250 mg, 555.0 µmol, 19.0% yield, 91.5% purity) as a yellow oil. 1 H NMR (400 MHz, CDCl3,) δ = 7.14 (d, J = 8.0 Hz, 1H), 6.99 (t, J = 8.0 Hz, 1H), 6.75 (d, J = 8.4 Hz, 1H), 4.31 - 4.10 (m, 2H), 3.93 (t, J = 6.4 Hz, 2H), 3.33 - 3.32 (m, 1H), 3.04 - 2.95 (m, 2H), 2.39 - 2.15 (m, 5H), 1.87 - 1.79 (m, 2H), 1.74 (d, J = 9.2 Hz, 2H), 1.45 - 1.39 (m, 2H), 1.37 - 1.26 (m, 9H). Step E. Procedure for preparation of (R)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(1-ethoxy-1-oxop ropan-2-yl)piperidin-4- yl)propoxy)-2-methylphenyl)picolinate To a mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pic olinate (408.50 mg, 666.89 µmol, 1.1 equiv.), and (R)-ethyl 2-(4-(3-(3-bromo-2-methylphenoxy)propyl)piperidin-1- yl)propanoate (250 mg, 606.27 µmol, 1 equiv.) in dioxane (5 mL) was added Ad 2 nBuP Pd G3(cataCXium® A Pd G3 ) (44.15 mg, 60.63 µmol, 0.1 equiv.) and KF (1.5 M, 1.21 mL, 3 equiv.). After addition, the mixture was degassed and purged with N2 three times, and then the mixture was stirred at 100 °C for 2 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~60% ethyl acetate/petroleum ether) to give (R)- tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1- (1-ethoxy-1-oxopropan-2-yl)piperidin-4-yl)propoxy)-2-methylp henyl)picolinate (320 mg, 361.1 µmol, 59.5% yield, 92.3% purity) as a yellow solid. MS (ESI) m/z: 818.8 [M+H] + . Step F. Procedure for preparation of (R)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)propyl)piperidin-1-yl)propanoic acid To a solution of (R)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(1-ethoxy-1-oxopropan-2-yl)piperidin-4- yl)propoxy)-2- methylphenyl)picolinate (320 mg, 391.18 µmol, 1 equiv.) in THF (3 mL) was added LiOH•H 2 O (49.25 mg, 1.17 mmol, 3 equiv.) and H 2 O (0.6 mL). The mixture was stirred at 40 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to remove THF. The aqueous phase was adjusted to pH=4~5 with 1M HCl. The reaction mixture was filtered and diluted in ethyl acetate. The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give (R)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)propyl)piperidin-1-yl)propanoic acid (350 mg, crude) as a yellow solid. MS (ESI) m/z: 790.7 [M+H] + Step G. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-((2R)-1-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-1-oxopropan-2-yl)piperidin-4-yl)propo xy)-2- methylphenyl)picolinate To a solution of (R)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)piperidin-1- yl)propanoic acid (100 mg, 112.93 µmol, 89.21% purity, 1 equiv.), and 3-(6-amino-1-methyl- 1H-indazol-3-yl)piperidine-2,6-dione (35.00 mg, 135.52 µmol, 1.2 equiv.) in pyridine (1 mL) was added EDCI (32.47 mg, 169.39 µmol, 1.5 equiv.). The mixture was stirred at 25 °C for 3 hours. The reaction mixture was quenched by addition H2O (2mL). The reaction mixture was filtered, washed with 5 mL of water, and diluted in DCM (10 mL). The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give tert- butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1- ((2R)-1-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)amino)-1-oxopropan-2- yl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinate (100 mg, crude) as a red solid. MS (ESI) m/z: 1030.8 [M+H] + . Step H. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-((2R)-1-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-1-oxopropan-2-yl)piperidin-4-yl)propo xy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-((2R)-1-((3-(2,6-dioxopiperidin-3-yl)-1 -methyl-1H-indazol-6-yl)amino)- 1-oxopropan-2-yl)piperidin-4-yl)propoxy)-2-methylphenyl)pico linate (100 mg, 97.06 µmol, 1 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 139.14 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-(1-((2R)-1-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-1- oxopropan-2-yl)piperidin-4- yl)propoxy)-2-methylphenyl)picolinic acid (63.5 mg, 64.6 µmol, 66.5% yield, 99.15% purity) as a white solid. MS (ESI) m/z: 975.0 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6, ) δ = 10.88 (s, 1H), 9.91 (s, 1H), 8.15 (s, 1H), 8.07 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 8.8 Hz, 2H), 7.51 - 7.40 (m, 3H), 7.40 - 7.30 (m, 2H), 7.19 (dd, J = 1.2, 7.6 Hz, 1H), 7.13 - 7.03 (m, 1H), 6.95 (d, J = 8.8 Hz, 1H), 6.87 (d, J = 8.4 Hz, 1H), 6.62 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.33 - 4.29 (m, 1H), 3.98 - 3.88 (m, 7H), 3.02 (t, J = 5.6 Hz, 2H), 2.88 - 2.77 (m, 2H), 2.68 - 2.59 (m, 2H), 2.40 - 2.26 (m, 2H), 2.22 - 2.10 (m, 2H), 1.89 (s, 3H), 1.82 - 1.63 (m, 4H), 1.40 - 1.30 (m, 2H), 1.34 - 1.09 (m, 7H). Example 36. Preparation of Compound 173a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-((3S)-3-(1-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino )-2-oxoethyl)piperidin-4- yl)butoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of (S)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-ethoxy-2-oxoe thyl)piperidin-4-yl)butoxy)- 2-methylphenyl)picolinate (S)-ethyl 2-(4-(4-(3-bromo-2-methylphenoxy)butan-2-yl)piperidin-1-yl)a cetate (90 mg, 218.26 µmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)picolinate (133.69 mg, 218.26 µmol, 1 equiv.), KF (38.04 mg, 654.77 µmol, 15.34 µL, 3 equiv.) and Ad2nBuP Pd G3 (15.89 mg, 21.83 µmol, 0.1 equiv.) were taken up into a microwave tube in dioxane (1 mL) and H2O (0.1 mL). The sealed tube was heated at 100 °C for 1 hour under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , DCM/MeOH = 20/1) to give (S)-tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-(1-(2-ethoxy-2- oxoethyl)piperidin-4-yl)butoxy)-2-methylphenyl)picolinate (96 mg, 117.3 µmol, 53.8% yield) as a yellow solid. MS (ESI) m/z: 818.4 [M+H] + . Step B. Procedure for preparation of (S)-2-(4-(4-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)butan-2-yl)piperidin-1-yl)acetic acid To a solution of (S)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-ethoxy-2-oxoethyl)piperidin-4-yl)but oxy)-2-methylphenyl)picolinate (90.00 mg, 110.02 µmol, 1 equiv.) in THF (1 mL) and H2O (0.5 mL) was added LiOH•H2O (1 M, 330.06 µL, 3 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was partitioned between DCM 15 (5 mL × 3) mL and H 2 O (6 mL). The organic phase was separated, washed with brine 9 mL (3 mL × 3), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give (S)-2-(4-(4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2-methylphenoxy)butan- 2-yl)piperidin-1-yl)acetic acid (80.00 mg, crude) as a yellow solid. MS (ESI) m/z: 818.4 [M+H] + . Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-((3S)-3-(1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-2-oxoethyl)piperidin-4-yl)butoxy)-2-m ethylphenyl)picolinate To a solution of (S)-2-(4-(4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)butan-2-yl)piperidin-1- yl)acetic acid (80 mg, 101.27 µmol, 1 equiv.) in pyridine (2 mL) was added dropwise EDCI (19.41 mg, 101.27 µmol, 1 equiv.) at 25°C. After addition, the mixture was stirred at this temperature for 15 minutes, and then 3-(7-amino-1-methyl-1H-indazol-3-yl)piperidine-2,6- dione (31.39 mg, 121.52 µmol, 1.2 equiv.) was added. The resulting mixture was stirred at 40 °C for 12 hours. The reaction mixture was partitioned between DCM (8 mL × 3) and H 2 O (15 mL). The organic phase was separated, washed with brine (5 mL × 3), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-((3S)-3-( 1-(2-((3-(2,6-dioxopiperidin-3- yl)-1-methyl-1H-indazol-7-yl)amino)-2-oxoethyl)piperidin-4-y l)butoxy)-2- methylphenyl)picolinate (90 mg, 87.4 µmol, 86.3% yield) as a yellow solid. MS (ESI) m/z: 804.7 [M+H] + Step D. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-((3S)-3-(1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-2-oxoethyl)piperidin-4-yl)butoxy)-2-m ethylphenyl)picolinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-((3S)-3-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-7-yl)amino)-2- oxoethyl)piperidin-4-yl)butoxy)-2-methylphenyl)picolinate (80 mg, 77.65 µmol, 1 equiv.) in DCM (0.6 mL) and TFA (0.5 mL) was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-((3S)-3- (1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl )amino)-2-oxoethyl)piperidin-4- yl)butoxy)-2-methylphenyl)picolinic acid (52.8 mg, 53.2 µmol, 68.6% yield, 98.2% purity) as a white solid. MS (ESI) m/z: 974.5 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.82 (s, 1H), 12.71 - 12.39 (m, 1H), 10.98 - 10.86 (m, 1H), 10.57 - 10.47 (m, 1H), 8.06 - 8.00 (m, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.68 (d, J = 8.4 Hz, 1H), 7.63 (d, J = 7.6 Hz, 1H), 7.50 - 7.42 (m, 3H), 7.40 - 7.33 (m, 2H), 7.22 - 7.18 (m, 1H), 7.16 - 7.07 (m, 2H), 6.97 (d, J = 8.8 Hz, 1H), 6.91 (d, J = 8.4 Hz, 1H), 6.66 - 6.61 (m, 1H), 5.02 - 4.94 (m, 2H), 4.46 - 4.35 (m, 1H), 4.31 - 4.17 (m, 2H), 4.09 - 4.02 (m, 3H), 3.92 (t, J = 6.0 Hz, 2H), 3.77 (s, 1H), 3.63 - 3.52 (m, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.73 - 2.61 (m, 2H), 2.33 (d, J = 2.0 Hz, 2H), 2.25 - 2.11 (m, 2H), 1.89 (d, J = 2.4 Hz, 3H), 1.85 - 1.79 (m, 2H), 1.70 - 1.49 (m, 6H), 1.26 - 1.20 (m, 1H), 0.96 - 0.88 (m, 3H) Example 37. Preparation of Compound 174b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(((1R,3s,5S)- 8-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl) amino)-2-oxoethyl)-8- azabicyclo[3.2.1]octan-3-yl)oxy)ethoxy)-2-methylphenyl)picol inic acid Step A. Procedure for preparation of (1R,3s,5S)-tert-butyl 3-(2-ethoxy-2-oxoethoxy)-8- azabicyclo[3.2.1]octane-8-carboxylate To a solution of (1R,3s,5S)-tert-butyl 3-hydroxy-8-azabicyclo[3.2.1]octane-8-carboxylate (1 g, 4.40 mmol, 1 equiv.) in DCM (20 mL) was added rhodium(II) acetate dimer (97.23 mg, 219.97 µmol, 0.05 equiv.). Ethyl 2-diazoacetate (652.59 mg, 5.72 mmol, 598.71 µL, 1.3 equiv.) was added at 0 °C. The mixture was stirred at 25 °C for 3 hours, and then ethyl 2-diazoacetate (251.00 mg, 2.20 mmol, 230.27 µL, 0.5 equiv.) was added at 0 °C. The mixture was stirred at 25 °C for 3 hours. DCM (50 mL) and water (50 mL) were added, and layers were separated. The aqueous phase was extracted with DCM (30 mL × 2). The combined organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum. The residue was purified by flash silica gel chromatography (Eluent of 0~22% ethyl acetate/petroleum ether) to give compound (1R,3s,5S)-tert-butyl 3-(2-ethoxy-2-oxoethoxy)-8-azabicyclo[3.2.1]octane-8- carboxylate (830 mg, 2.6 mmol, 60.2% yield) as a yellow oil. MS (ESI) m/z: 314.0 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 4.16 - 4.00 (m, 6H), 3.90 - 3.78 (m, 1H), 2.02 - 1.77 (m, 4H), 1.58 (d, J = 5.2 Hz, 2H), 1.46 - 1.37 (m, 9H), 1.35 (s, 2H), 1.23 - 1.16 (m, 3H) Step B. Procedure for preparation of (1R,3s,5S)-tert-butyl 3-(2-hydroxyethoxy)-8- azabicyclo[3.2.1]octane-8-carboxylate To a solution of LiAlH4 (80.42 mg, 2.12 mmol, 0.8 equiv.) in THF (20 mL) was added (1R,3s,5S)-tert-butyl 3-(2-ethoxy-2-oxoethoxy)-8-azabicyclo[3.2.1]octane-8-carboxy late (830 mg, 2.65 mmol, 1 equiv.) in THF (5 mL) at 0 °C. The mixture was stirred at 0 °C for 2 hours. The reaction mixture was quenched by addition water (0.1 mL) and 15% NaOH (0.1 mL) and water (0.3 mL). The combined mixture was dried with Na2SO4, filtered, and concentrated under reduced pressure to give compound (1R,3s,5S)-tert-butyl 3-(2-hydroxyethoxy)-8- azabicyclo[3.2.1]octane-8-carboxylate (600 mg, 2.21 mmol, 83.5% yield) as a colorless oil. MS (ESI) m/z: 294.2 [M+23] + 1 H NMR (400 MHz, DMSO-d6) δ = 4.53 (s, 1H), 4.06 (s, 2H), 3.82 - 3.70 (m, 1H), 3.48 - 3.37 (m, 4H), 2.03 - 1.75 (m, 5H), 1.68 - 1.55 (m, 2H), 1.41 (s, 9H), 1.34 (s, 1H) Step C. Procedure for preparation of (1R,3s,5S)-tert-butyl 3-(2-(3-bromo-2- methylphenoxy)ethoxy)-8-azabicyclo[3.2.1]octane-8-carboxylat e To a solution of (1R,3s,5S)-tert-butyl 3-(2-hydroxyethoxy)-8-azabicyclo[3.2.1]octane-8- carboxylate (500 mg, 1.84 mmol, 1 equiv.) in toluene (10 mL) was added 2-(tributyl-λ5- phosphanylidene)acetonitrile (667.09 mg, 2.76 mmol, 1.5 equiv.) and 3-bromo-2-methylphenol (689.27 mg, 3.69 mmol, 2 equiv.). The mixture was stirred at 120 °C for 12 hours. DCM (50 mL) and water (50 mL) were added and layers were separated. The aqueous phase was extracted with DCM (30 mL × 2). The combined organic phase was dried over anhydrous sodium sulfate, filtered and concentrated under vacuum. The residue was purified by flash silica gel chromatography (Eluent of 0~22% ethyl acetate/petroleum ether) to give compound (1R,3s,5S)-tert-butyl 3-(2-(3-bromo-2-methylphenoxy)ethoxy)-8-azabicyclo[3.2.1]oct ane-8- carboxylate (620 mg, 1.4 mmol, 76.4% yield) as a yellow oil. MS (ESI) m/z: 385.8 [M+H] + 1 H NMR (400 MHz, DMSO-d6,) δ = 7.18 - 7.14 (m, 1H), 7.09 (t, J = 8.0 Hz, 1H), 6.98 (d, J = 7.6 Hz, 1H), 4.13 - 4.00 (m, 5H), 3.91 - 3.81 (m, 1H), 3.76 - 3.71 (m, 2H), 2.23 (s, 3H), 2.06 - 1.71 (m, 5H), 1.62 (d, J = 5.2 Hz, 2H), 1.40 (s, 9H) Step D. Procedure for preparation of (1R,3s,5S)-3-(2-(3-bromo-2- methylphenoxy)ethoxy)-8-azabicyclo[3.2.1]octane To a solution of (1R,3s,5S)-tert-butyl 3-(2-(3-bromo-2-methylphenoxy)ethoxy)-8- azabicyclo[3.2.1]octane-8-carboxylate (320 mg, 726.66 µmol, 1 equiv.) in DCM (4 mL) was added HCl/dioxane (4 M, 1.60 mL, 8.81 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to remove solvent to give compound (1R,3S,5S)-3-(2-(3-bromo-2-methylphenoxy)ethoxy)-8-azabicycl o[3.2.1]octane (260 mg, 690.2 µmol, 94.9% yield) as a pink solid. 1 H NMR (400 MHz, DMSO-d6) δ = 9.11 - 8.71 (m, 2H), 7.19 - 7.15 (m, 1H), 7.10 (t, J = 8.0 Hz, 1H), 6.98 (d, J = 8.4 Hz, 1H), 4.08 (t, J = 4.4 Hz, 2H), 3.98 (s, 2H), 3.57 (s, 2H), 2.24 (s, 3H), 2.09 (d, J = 13.2 Hz, 2H), 1.98 - 1.81 (m, 4H), 1.72 - 1.56 (m, 2H) Step E. Procedure for preparation of ethyl 2-((1R,3S,5S)-3-(2-(3-bromo-2- methylphenoxy)ethoxy)-8-azabicyclo[3.2.1]octan-8-yl)acetate To a solution of (1R,3s,5S)-3-(2-(3-bromo-2-methylphenoxy)ethoxy)-8- azabicyclo[3.2.1]octane (300 mg, 796.36 µmol, 1 equiv., HCl) in CH 3 CN (3 mL) was added K 2 CO 3 (550.32 mg, 3.98 mmol, 5 equiv.) and ethyl 2-bromoacetate (159.59 mg, 955.63 µmol, 105.69 µL, 1.2 equiv.). The mixture was stirred at 60 °C for 2 hours. DCM (30 mL) and water (30 mL) were added, and layers were separated. The aqueous phase was extracted with DCM (15 mL × 2). The combined organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=10/1 to 0/1) to give compound ethyl 2-((1R,3s,5S)-3-(2-(3- bromo-2-methylphenoxy)ethoxy)-8-azabicyclo[3.2.1]octan-8-yl) acetate (320 mg, 750.5 µmol, 94.2% yield) as a yellow oil. MS (ESI) m/z: 428.1 [M+H] + Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(((1R,3S,5S)-8-(2-ethox y-2-oxoethyl)-8- azabicyclo[3.2.1]octan-3-yl)oxy)ethoxy)-2-methylphenyl)picol inate To a solution of ethyl 2-((1R,3s,5S)-3-(2-(3-bromo-2-methylphenoxy)ethoxy)-8- azabicyclo[3.2.1]octan-8-yl)acetate (320 mg, 750.57 µmol, 1 equiv.) in dioxane (4 mL) was added tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)picolinate (459.76 mg, 750.57 µmol, 1 equiv.), K 3 PO 4 (477.96 mg, 2.25 mmol, 3 equiv.), H 2 O (0.4 mL), and Ad 2 nBup-Pd-G 3 (54.66 mg, 75.06 µmol, 0.1 equiv.). The mixture was stirred at 100 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by flash silica gel chromatography (Eluent of 0~76% ethyl acetate/petroleum ether) to give compound tert- butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2- (((1R,3S,5S)-8-(2-ethoxy-2-oxoethyl)-8-azabicyclo[3.2.1]octa n-3-yl)oxy)ethoxy)-2- methylphenyl)picolinate (400 mg, 480.8 µmol, 64.0% yield) as a yellow solid. MS (ESI) m/z: 832.5 [M+H] + Step G. Procedure for preparation of 2-((1R,3s,5S)-3-(2-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)ethoxy)-8-azabicyclo[3.2.1]octan-8-yl)acetic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(((1R,3s,5S)-8-(2-ethoxy-2-oxoethyl)-8-aza bicyclo[3.2.1]octan-3- yl)oxy)ethoxy)-2-methylphenyl)picolinate (350 mg, 420.67 µmol, 1 equiv.) in THF (4 mL) and H2O (1 mL) was added LiOH‧H2O (52.96 mg, 1.26 mmol, 3 equiv.). The mixture was stirred at 25 °C for 3 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. To the residue was added 5 mL H 2 O. The pH of mixture was adjusted to 4 with 10% citric acid. Then the reaction mixture was filtered and concentrated under reduced pressure to give compound 2-((1R,3sS,5S)-3-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl) -3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)ethoxy)-8-azabicyclo[3.2.1]octan-8-yl)acetic acid (320 mg, 398.0 µmol, 94.6% yield) as a yellow solid. MS (ESI) m/z: 804.3 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.02 - 12.73 (m, 1H), 8.01 (d, J = 7.6 Hz, 1H), 7.77 (d, J = 8.0 Hz, 1H), 7.60 (d, J = 7.6 Hz, 1H), 7.47 - 7.41 (m, 3H), 7.39 - 7.31 (m, 2H), 7.13 - 7.06 (m, 1H), 6.97 - 6.91 (m, 2H), 6.59 (d, J = 7.6 Hz, 1H), 5.75 (s, 1H), 4.97 (s, 2H), 3.87 (t, J = 5.6 Hz, 2H), 3.17 (s, 2H), 3.03 (t, J = 5.6 Hz, 3H), 2.24 (s, 1H), 2.06 - 1.96 (m, 6H), 1.88 (s, 3H), 1.78 - 1.68 (m, 6H), 1.01 (s, 9H) Step H. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(((1R,3s,5S)-8-(2-((3-( 2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)-8-azabicyclo[3.2.1 ]octan-3-yl)oxy)ethoxy)-2- methylphenyl)picolinate To a solution of 2-((1R,3s,5S)-3-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)ethoxy)-8-azabicyclo[3.2.1]octan-8-yl)acetic acid (110 mg, 136.82 µmol, 1 equiv.) in DMF (1 mL) was added HATU (62.43 mg, 164.19 µmol, 1.2 equiv.) and DIEA (53.05 mg, 410.47 µmol, 71.50 µL, 3 equiv.). Then 3-(6-amino-1-methyl-1H-indazol-3- yl)piperidine-2,6-dione (38.87 mg, 150.50 µmol, 1.1 equiv.) was added into the mixture and stirred at 25 °C for 12 hours. DCM (20 mL) and water (20 mL) were added and layers were separated. The aqueous phase was extracted with DCM (10 mL × 2). The combined organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum to give compound tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(((1R,3s,5S)-8-(2-((3-(2,6-dioxopiperidin- 3-yl)-1-methyl-1H-indazol-6- yl)amino)-2-oxoethyl)-8-azabicyclo[3.2.1]octan-3-yl)oxy)etho xy)-2-methylphenyl)picolinate (120 mg, 114.9 µmol, 83.9% yield) as a brown solid. MS (ESI) m/z: 1044.5 [M+H] + Step I. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(((1R,3s,5S)-8-(2-((3-( 2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)-8-azabicyclo[3.2.1 ]octan-3-yl)oxy)ethoxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(((1R,3s,5S)-8-(2-((3-(2,6-dioxopiperidin- 3-yl)-1-methyl-1H-indazol-6- yl)amino)-2-oxoethyl)-8-azabicyclo[3.2.1]octan-3-yl)oxy)etho xy)-2-methylphenyl)picolinate (120 mg, 114.92 µmol, 1 equiv.) in DCM (2 mL) was added TFA (3.08 g, 27.01 mmol, 2 mL, 235.06 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by prep- HPLC to give compound 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(((1R,3s,5S)-8-(2-((3-(2,6-dioxopiperidin- 3-yl)-1-methyl-1H-indazol-6- yl)amino)-2-oxoethyl)-8-azabicyclo[3.2.1]octan-3-yl)oxy)etho xy)-2-methylphenyl)picolinic acid (35.7 mg, 33.2 µmol, 28.9% yield, 96.1% purity) as an off-white solid. MS (ESI) m/z: 494.9 [(M+2)/2] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.87 (s, 1H), 9.91 (s, 1H), 8.15 (s, 1H), 8.05 - 8.00 (m, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.65 - 7.60 (m, 2H), 7.49 - 7.41 (m, 3H), 7.39 - 7.31 (m, 2H), 7.26 - 7.21 (m, 1H), 7.14 - 7.05 (m, 1H), 6.98 - 6.87 (m, 2H), 6.65 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.37 - 4.28 (m, 1H), 4.07 (t, J = 4.4 Hz, 2H), 3.94 - 3.88 (m, 5H), 3.77 - 3.74 (m, 2H), 3.73 - 3.65 (m, 2H), 3.17 (s, 2H), 3.02 (t, J = 5.6 Hz, 2H), 2.68 - 2.62 (m, 2H), 2.38 - 2.29 (m, 2H), 2.20 - 2.14 (m, 1H), 1.91 (s, 7H), 1.66 - 1.56 (m, 4H) Example 38. Preparation of Compound 178 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(4-(4-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin -1-yl)-4-oxobutyl)phenoxy)-2- methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(4-ethoxy-4-oxobutyl)ph enoxy)-2- methylphenyl)picolinate Tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)picolinate (779.34 mg, 1.27 mmol, 1.2 equiv.), ethyl 4-(4-(3-bromo-2-methylphenoxy)phenyl)butanoate (400 mg, 1.06 mmol, 1 equiv.), K2CO3 (1.5 M, 1.06 mL, 3 equiv.), and [2-(2-aminophenyl)phenyl]palladium(1 + ); bis(1- adamantyl)-butylphosphane;methanesulfonate (154.43 mg, 212.05 µmol, 0.2 equiv.) were taken up into a microwave tube in dioxane (4 mL) and H 2 O (0.5 mL). The sealed tube was heated at 100 °C for 60 min under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 50% Ethylacetate / petroleum ether) to give tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(4-eth oxy-4-oxobutyl)phenoxy)-2- methylphenyl)picolinate (730 mg, crude) as a yellow oil. MS (ESI) m/z: 783.4 [M+H] + . Step B. Procedure for preparation of 4-(4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)phenyl)butanoic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(4-(4-ethoxy-4-oxobutyl)phenoxy)-2-methylphenyl)pic olinate (243 mg, 310.37 µmol, 1 equiv.), LiOH·H2O (78.14 mg, 1.86 mmol, 5 equiv.) in THF (1.8 mL) and H2O (0.6 mL) was stirred at 25 °C for 16 hours. The mixture was concentrated to remove the THF. It was then acidified to pH = 2~3 with 1 M HCl (5 mL). A white solid precipitated from the mixture which was collected by filtration to give 4-(4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)phenyl)butanoic acid (212 mg, crude) as a white solid. MS (ESI) m/z: 755.4 [M+H] + . Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(4-(4-(3-(2,6-dioxopipe ridin-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)-4-oxobutyl)phenoxy)-2-methylphe nyl)picolinate To a solution of 4-(4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroiso quinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)phe nyl)butanoic acid (150 mg, 198.70 µmol, 1 equiv.) in DMF (1.5 mL) was added HATU (113.33 mg, 298.06 µmol, 1.5 equiv.) and DIEA (77.04 mg, 596.11 µmol, 103.83 µL, 3 equiv.).The mixture was stirred at 25 °C for 1 hour. Then 3-(1-methyl-6-(piperazin-1-yl)-1H-indazol-3-yl)piperidine-2, 6-dione (78.06 mg, 238.45 µmol, 1.2 equiv.) was added. The mixture was stirred at 25 °C for 15 hours. The mixture was diluted with saturated Na2CO3 solution (10 mL). The solution was filtered and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(4-(4- (3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-4-oxobutyl)phenoxy)-2 -methylphenyl)picolinate (209 mg crude) as a white solid. MS (ESI) m/z: 1064.5 [M+H] + Step D. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(4-(4-(3-(2,6-dioxopipe ridin-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)-4-oxobutyl)phenoxy)-2-methylphe nyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-6-yl)piperazin-1- yl)-4-oxobutyl)phenoxy)-2-methylphenyl)picolinate (200 mg, 187.92 µmol, 1 equiv.) in DCM (1.5 mL) was added TFA (1.5 mL). The mixture was stirred at 25°C for 48 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give the compound 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(4-(4-(3-(2,6-dioxopipe ridin-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)-4-oxobutyl)phenoxy)-2-methylphe nyl)picolinic acid (26.3 mg, 24.6 µmol,13.1 % yield, 94.4 % purity) as a white solid. MS (ESI) m/z: 1008.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.02 - 12.55 (m, 2H), 10.86 (s, 1H), 8.06 - 8.02 (m, 1H), 7.83 - 7.76 (m, 1H), 7.67 - 7.59 (m, 1H), 7.55 - 7.50 (m, 2H), 7.50 - 7.43 (m, 2H), 7.40 - 7.33 (m, 2H), 7.21 - 7.14 (m, 3H), 7.00 (d, J = 8.8 Hz, 1H), 6.94 (d, J = 9.2 Hz, 1H), 6.90 - 6.80 (m, 5H), 4.99 (s, 2H), 4.22 (s, 1H), 3.95 - 3.88 (m, 5H), 3.61 (d, J = 15.6 Hz, 4H), 3.19 (d, J = 15.6 Hz, 4H), 3.03 (t, J = 5.6 Hz, 2H), 2.62 - 2.56 (m, 4H), 2.38 (t, J = 7.2 Hz, 2H), 2.33 - 2.27 (m, 1H), 2.19 - 2.11 (m, 1H), 1.90 (s, 3H), 1.81 (m, J = 7.2 Hz, 2H). Example 39. Preparation of Compound 180a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-((3S)-1-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino )-2-oxoethyl)piperidin-3- yl)ethoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of (S)-tert-butyl 3-(2-(3-bromo-2- methylphenoxy)ethyl)piperidine-1-carboxylate To a solution of 3-bromo-2-methylphenol (815.62 mg, 4.36 mmol, 1 equiv.) in toluene (10 mL) was added 2-(tributyl-λ5-phosphanylidene)acetonitrile (1.58 g, 6.54 mmol, 1.5 equiv.) and (S)- tert-butyl 3-(2-hydroxyethyl)piperidine-1-carboxylate (1 g, 4.36 mmol, 1 equiv.). The mixture was stirred at 120 °C for 2 hours under N2. The reaction mixture was concentrated under reduced pressure to a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~10% ethyl acetate/petroleum ether) to give (S)-tert-butyl 3-(2-(3-bromo-2- methylphenoxy)ethyl)piperidine-1-carboxylate (1.6 g, 4.0 mmol, 92.1% yield) as a yellow oil. MS (ESI) m/z: 314.0 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.17 - 7.13 (m, 1H), 7.12 - 7.06 (m, 1H), 6.98 (d, J = 8.0 Hz, 1H), 4.03 (t, J = 6.0 Hz, 2H), 3.91 - 3.69 (m, 2H), 2.23 (s, 3H), 1.80 (d, J = 11.8 Hz, 1H), 1.71 - 1.53 (m, 5H), 1.47 - 1.24 (m, 12H) Step B. Procedure for preparation of (S)-3-(2-(3-bromo-2- methylphenoxy)ethyl)piperidine To a solution of (S)-tert-butyl 3-(2-(3-bromo-2-methylphenoxy)ethyl)piperidine-1-carboxylate (1.6 g, 4.02 mmol, 1 equiv.) in HCl/dioxane (20 mL). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to remove solvent, and then it was filtered and concentrated under reduced pressure to give compound (S)-3-(2-(3- bromo-2-methylphenoxy)ethyl)piperidine (1.03 g, 3.1 mmol, 76.6% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ = 9.06 - 8.78 (m, 2H), 7.18 - 7.14 (m, 1H), 7.12 - 7.06 (m, 1H), 6.98 (d, J = 8.0 Hz, 1H), 4.07 - 3.98 (m, 2H), 3.29 - 3.21 (m, 1H), 3.18 (d, J = 12.0 Hz, 1H), 2.80 - 2.68 (m, 1H), 2.60 (t, J = 12.0 Hz, 1H), 2.24 (s, 3H), 2.04 - 1.93 (m, 1H), 1.85 - 1.60 (m, 5H), 1.29 - 1.14 (m, 1H) Step C. Procedure for preparation of (S)-ethyl 2-(3-(2-(3-bromo-2- methylphenoxy)ethyl)piperidin-1-yl)acetate To a solution of (S)-3-(2-(3-bromo-2-methylphenoxy)ethyl)piperidine (500 mg, 1.49 mmol, 1 equiv., HCl) in CH 3 CN (5 mL) was added K 2 CO 3 (1.03 g, 7.47 mmol, 5 equiv.), KI (24.80 mg, 149.40 µmol, 0.1 equiv.), and ethyl 2-bromoacetate (249.49 mg, 1.49 mmol, 165.23 µL, 1 equiv.). The mixture was stirred at 60 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by flash silica gel chromatography (Eluent of 0~100% ethyl acetate/petroleum ether) to give (S)-ethyl 2-(3-(2-(3- bromo-2-methylphenoxy)ethyl)piperidin-1-yl)acetate (360 mg, 936.7 µmol, 62.7% yield) as a yellow oil. MS (ESI) m/z: 386.0 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.17 - 7.13 (m, 1H), 7.11 - 7.06 (m, 1H), 6.97 (d, J = 8.0 Hz, 1H), 4.09 - 4.03 (m, 2H), 4.03 - 3.97 (m, 2H), 3.16 (s, 2H), 2.80 (d, J = 10.4 Hz, 1H), 2.71 (d, J = 10.8 Hz, 1H), 2.22 (s, 3H), 2.18 - 2.10 (m, 1H), 1.95 (t, J = 10.0 Hz, 1H), 1.77 - 1.69 (m, 2H), 1.67 - 1.59 (m, 2H), 1.59 - 1.54 (m, 1H), 1.51 - 1.37 (m, 1H), 1.16 (t, J = 7.2 Hz, 3H), 0.99 - 0.88 (m, 1H) Step D. Procedure for preparation of (S)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(1-(2-ethoxy-2-oxoe thyl)piperidin-3-yl)ethoxy)- 2-methylphenyl)picolinate To a solution of (S)-ethyl 2-(3-(2-(3-bromo-2-methylphenoxy)ethyl)piperidin-1-yl)acetat e (300 mg, 780.63 µmol, 1 equiv.) in dioxane (3 mL) was added tert-butyl 6-(8-(benzo[d]thiazol- 2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-t etramethyl-1,3,2-dioxaborolan- 2-yl)picolinate (573.80 mg, 936.75 µmol, 1.2 equiv.), KF (1.5 M, 1.56 mL, 3 equiv.), and [2- (2-aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-butyl-p hosphane;methanesulfonate (56.85 mg, 78.06 µmol, 0.1 equiv.). The mixture was stirred at 100 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~40% ethyl acetate/petroleum ether) to give (S)- tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(1- (2-ethoxy-2-oxoethyl)piperidin-3-yl)ethoxy)-2-methylphenyl)p icolinate (450 mg, 541.1 µmol, 69.3% yield, 95% purity) as a yellow solid. MS (ESI) m/z: 790.5 [M+H] + Step E. Procedure for preparation of (S)-2-(3-(2-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)ethyl)piperidin-1-yl)acetic acid To a solution of (S)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(1-(2-ethoxy-2-oxoethyl)piperidin-3-yl)eth oxy)-2-methylphenyl)picolinate (450 mg, 569.64 µmol, 1 equiv.) in THF (5 mL) and H2O (1.25 mL) was added LiOH•H2O (71.71 mg, 1.71 mmol, 3 equiv.). The mixture was stirred at 40 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to remove solvent, the residue was added 5mL H2O. The pH of mixture was adjusted to 4 with 1M HCl. Then the reaction mixture was filtered and concentrated under reduced pressure to give (S)-2-(3-(2-(3-(6-(8-(benzo[d]thiazol- 2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-buto xycarbonyl)pyridin-3-yl)-2- methylphenoxy)ethyl)piperidin-1-yl)acetic acid (430 mg, 564.3 µmol, 99.1% yield) as a yellow solid. MS (ESI) m/z: 762.5 [M+H] + Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((3S)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-3-yl)ethoxy)-2-m ethylphenyl)picolinate To a solution of (S)-2-(3-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)ethyl)piperidin-1-yl)acetic acid (130 mg, 170.62 µmol, 1 equiv.) in DMF (1 mL) was added HATU (84.34 mg, 221.81 µmol, 1.3 equiv.) and DIEA (66.15 mg, 511.86 µmol, 89.16 µL, 3 equiv.). To the mixture was added 3-(6-amino-1-methyl-1H-indazol-3-yl)piperidine-2,6-dione (52.88 mg, 204.74 µmol, 1.2 equiv.) and stirred at 25 °C for 12 hours. The reaction mixture was added into water (5 mL), then filtered and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol- 2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((3S )-1-(2-((3-(2,6-dioxopiperidin- 3-yl)-1-methyl-1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-3 -yl)ethoxy)-2- methylphenyl)picolinate (130 mg, 129.7 µmol, 76.0% yield) as a yellow solid. MS (ESI) m/z: 1003.7 [M+H] + Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((3S)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-3-yl)ethoxy)-2-m ethylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-((3S)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1 -methyl-1H-indazol-6-yl)amino)- 2-oxoethyl)piperidin-3-yl)ethoxy)-2-methylphenyl)picolinate (130 mg, 129.72 µmol, 1 equiv.) in DCM (1.5 mL) was added TFA (14.79 mg, 129.72 µmol, 9.60 µL, 1 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(2-((3S)-1-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2- oxoethyl)piperidin-3- yl)ethoxy)-2-methylphenyl)picolinic acid (53.6 mg, 53.4 µmol, 41.2% yield, 98.8% purity) as an off-white solid. MS (ESI) m/z: 946.4 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 13.20 - 12.63 (m, 1H), 10.87 (s, 1H), 9.93 - 9.83 (m, 1H), 8.15 - 8.12 (m, 1H), 8.07 - 7.99 (m, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.67 - 7.57 (m, 2H), 7.52 - 7.42 (m, 2H), 7.41 - 7.32 (m, 3H), 7.17 (d, J = 8.4 Hz, 1H), 7.06 (t, J = 8.0 Hz, 1H), 6.92 (d, J = 8.4 Hz, 1H), 6.86 (d, J = 8.4 Hz, 1H), 6.65 - 6.57 (m, 1H), 4.98 (s, 2H), 4.37 - 4.26 (m, 1H), 4.06 - 3.97 (m, 2H), 3.95 - 3.85 (m, 5H), 3.21 - 3.09 (m, 3H), 3.04 - 2.98 (m, 2H), 2.95 - 2.86 (m, 1H), 2.84 - 2.75 (m, 1H), 2.68 - 2.60 (m, 2H), 2.35 - 2.28 (m, 1H), 2.24 - 2.11 (m, 2H), 2.07 - 1.98 (m, 1H), 1.88 (s, 3H), 1.86 - 1.50 (m, 6H) Example 40. Preparation of Compound 180b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-((3R)-1-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino )-2-oxoethyl)piperidin-3- yl)ethoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of (R)-tert-butyl 3-(2-(3-bromo-2- methylphenoxy)ethyl)piperidine-1-carboxylate A mixture of 3-bromo-2-methylphenol (0.91 g, 4.87 mmol, 1 equiv.), (R)-tert-butyl 3-(2- hydroxyethyl)piperidine-1-carboxylate (1.00 g, 4.38 mmol, 0.9 equiv.), 2-(tributyl-λ 5 - phosphanylidene)acetonitrile (1.76 g, 7.30 mmol, 1.5 equiv.) in toluene (10 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 120 °C for 2 h under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 10% ethyl acetate / petroleum ether) to give (R)-tert-butyl 3-(2-(3-bromo-2-methylphenoxy)ethyl)piperidine-1- carboxylate (1.64 g, crude) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.15 (d, J = 8.0 Hz, 1H), 7.09 (t, J = 8.0 Hz, 1H), 6.97 (d, J = 8.0 Hz, 1H), 4.03 (t, J = 5.6 Hz, 2H), 3.84 - 3.71 (m, 2H), 2.83 - 2.64 (m, 2H), 2.23 (s, 3H), 1.81 - 1.58 (m, 5H), 1.33 (s, 9H), 1.27 - 1.17 (m, 2H) Step B. Procedure for preparation of (R)-3-(2-(3-bromo-2- methylphenoxy)ethyl)piperidine A solution of (R)-tert-butyl 3-(2-(3-bromo-2-methylphenoxy)ethyl)piperidine-1-carboxylate (1.64 g, 4.12 mmol, 1 equiv.) in TFA (15 mL) and DCM (15 mL) was stirred at 25 °C for 1 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 10% MeOH / DCM gradient) to give (R)-3-(2-(3-bromo-2-methylphenoxy)ethyl)piperidine (1.34 g, crude) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 8.66 (s, 1H), 7.19 - 7.14 (m, 1H), 7.10 (t, J = 8.0 Hz, 1H), 6.98 (d, J = 7.6 Hz, 1H), 4.08 - 4.03 (m, 2H), 3.31 - 3.20 (m, 2H), 2.81 - 2.60 (m, 2H), 2.24 (s, 3H), 1.97 - 1.89 (m, 1H), 1.87 - 1.53 (m, 6H) Step C. Procedure for preparation of (R)-ethyl 2-(3-(2-(3-bromo-2- methylphenoxy)ethyl)piperidin-1-yl)acetate A mixture of (R)-3-(2-(3-bromo-2-methylphenoxy)ethyl)piperidine (600 mg, 1.41 mmol, 70% purity, 1 equiv.), ethyl 2-bromoacetate (235.20 mg, 1.41 mmol, 155.76 µL, 1 equiv.), K2CO3 (973.22 mg, 7.04 mmol, 5 equiv.), KI (23.38 mg, 140.84 µmol, 0.1 equiv.) in CH 3 CN (5 mL) was stirred at 60 °C for 1 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 100% ethyl acetate / petroleum ether gradient) to give (R)-ethyl 2-(3-(2-(3-bromo-2- methylphenoxy)ethyl)piperidin-1-yl)acetate (420 mg, 1.1 mmol, 77.6% yield, 100% purity) as a white oil. MS (ESI) m/z: 385.9 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.15 (d, J = 8.0 Hz, 1H), 7.09 (t, J = 8.0 Hz 1H), 6.97 (d, J = 8.0 Hz, 1H), 4.09 - 4.05 (m, 2H), 4.03 - 3.99 (m, 2H), 3.17 (s, 2H), 2.82 - 2.70 (m, 1H), 2.73 - 2.70 (m, 1H), 2.23 (s, 3H), 2.18 - 1.92 (m, 1H), 1.77 - 1.57 (m, 5H), 1.50 - 1.42 (m, 1H), 1.17 (t, J = 7.2 Hz, 3H), 0.99 - 0.92 (m, 1H) Step D. Procedure for preparation of (R)-tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(1-(2- ethoxy-2- oxoethyl)piperidin-3-yl)ethoxy)-2-methylphenyl)picolinate A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)picolinat e (621.62 mg, 1.01 mmol, 1.3 equiv.), (R)-ethyl 2-(3-(2-(3-bromo-2-methylphenoxy)ethyl)piperidin-1-yl)acetat e (300 mg, 780.63 µmol, 1 equiv.), [2-(2-aminophenyl)phenyl]palladium(1 + );bis(1-adamantyl)-butyl- phosphane;methanesulfonate (113.70 mg, 156.13 µmol, 0.2 equiv.), KF (1.5 M, 780.63 µL, 1.5 equiv.) in dioxane (3 mL) was stirred at 100 °C for 1 h under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 50% ethyl acetate / petroleum ether) to give (R)-tert- butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(1-(2- ethoxy-2-oxoethyl)piperidin-3-yl)ethoxy)-2-methylphenyl)pico linate (573 mg, 725.3 µmol, 92.9% yield) as a yellow oil. MS (ESI) m/z: 790.4 [M+H] + . Step E. Procedure for preparation of (R)-2-(3-(2-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)ethyl)piperidin-1-yl)acetic acid A mixture of (R)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(1-(2-ethoxy-2-oxoethyl)piperidin-3-yl)eth oxy)-2-methylphenyl)picolinate (523 mg, 662.04 µmol, 1 equiv.), LiOH•H2O (138.91 mg, 3.31 mmol, 5 equiv.) in THF (4 mL) and H 2 O (2 mL) was stirred at 40 °C for 1 h. The reaction mixture was diluted with H 2 O (2 mL) and concentrated as a turbid liquid. Then the pH of the turbid liquid was adjusted to 3 with 1 N HCl (2 mL). A residue was obtained after trituration and filtered. The residue was used for next step without other purification. The compound (R)-2-(3-(2-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)ethyl)piperidin-1-yl)acetic acid (458 mg, 601.1 µmol, 90.8% yield) was obtained as a yellow solid. MS (EIS) m/z: 762.6 [M+H] + . Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((3R)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-3-yl)ethoxy)-2-m ethylphenyl)picolinate A mixture of (R)-2-(3-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)ethyl)piperidin-1-yl)acetic acid (80 mg, 105.00 µmol, 1 equiv.), HATU (59.88 mg, 157.50 µmol, 1.5 equiv.), DIPEA (40.71 mg, 314.99 µmol, 54.87 µL, 3 equiv.) in DMF (0.8 mL) was stirred at 25°C for 0.25 h. Then 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (32.5 mg, 126 µmol, 1.2 equiv.) was added and the mixture was stirred at 40 °C for 2 h. The reaction mixture was quenched by addition brine 5 mL at 25 °C, and then diluted and extracted with ethyl acetate (10 mL), concentrated under reduced pressure to give a brown oil. The oil was used for next step without other purification. The compound tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((3R)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)piperidin-3-yl)ethoxy)-2-meth ylphenyl)picolinate (100 mg, crude) was obtained as a brown oil. MS (ESI) m/z: 1002.6 [M+H] + . Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((3R)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-3-yl)ethoxy)-2-m ethylphenyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(2-((3R)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-6-yl)amino)-2- oxoethyl)piperidin-3-yl)ethoxy)-2-methylphenyl)picolinate (80 mg, 79.83 µmol, 1 equiv.) in DCM (0.5 mL) and TFA (0.5 mL) was stirred at 25 °C for 16 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2- ((3R)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)amino)-2- oxoethyl)piperidin-3-yl)ethoxy)-2-methylphenyl)picolinic acid (46.1 mg, 47.2 µmol, 59.1% yield, 96.8% purity) as a white solid. MS (ESI) m/z: 946.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.98 - 12.83 (m, 2H), 10.89 (s, 1H), 10.19 - 9.64 (m, 1H), 8.04 - 8.01 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.68 - 7.63 (m, 2H), 7.50 - 7.45 (m, 2H), 7.40 - 7.33 (m, 3H), 7.16 - 7.07 (m, 2H), 6.97 - 6.88 (m, 2H), 6.63 (d, J = 7.6 Hz, 1H), 4.99 (s, 2H), 4.33 (dd, J = 4.4, 9.6 Hz, 1H), 4.05 - 4.00 (m, 2H), 3.94 - 3.91 (m, 5H), 3.03 (t, J = 5.6 Hz, 2H), 2.68 - 2.55 (m, 5H), 2.38 - 2.31 (m, 2H), 2.21 - 2.13 (m, 2H), 1.91 (s, 3H), 1.86 - 1.57 (m, 6H), 1.32 - 0.99 (m, 2H) Example 41. Preparation of Compound 181 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-4-piperidyl]-2,2-difluoro- propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 4-(3-ethoxy-2,2-difluoro-1-hydroxy-3- oxo-propyl)piperidine-1-carboxylate A mixture of Zn (5.21 g, 79.71 mmol, 2 equiv.) in THF (100 mL) was degassed and purged with N 2 three times and added dropwise 1,2-dibromoethane (748.72 mg, 3.99 mmol, 300.69 µL, 0.1 equiv.) and then the mixture was stirred at 60 °C for 0.5 h under N2 atmosphere. And then a solution of tert-butyl 4-formylpiperidine-1-carboxylate (8.5 g, 39.86 mmol, 1 equiv.) and ethyl 2-bromo-2,2-difluoro-acetate (11.33 g, 55.80 mmol, 7.17 mL, 1.4 equiv.) in THF (100 mL) was added dropwise to the above solution, and the resulting solution was stirred at 60 °C for 3 hours. The mixture was cooled to 20 °C and then quenched by 1N HCl (200 mL), and then extracted with EtOAc (100 mL × 3). The combined organic layers were washed with brine (100 mL × 2), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~20% ethyl acetate/petroleum ether) to give compound tert-butyl 4-(3-ethoxy-2,2-difluoro-1- hydroxy-3-oxo-propyl)piperidine-1-carboxylate (19.6 g, crude) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6,) δ = 4.35 - 4.21 (m, 2H), 3.94 (s, 2H), 3.80 - 3.64 (m, 1H), 3.18 - 3.16 (m, 1H), 2.67 (s, 2H), 1.82 - 1.68 (m, 2H), 1.59 (d, J = 12.4 Hz, 1H), 1.39 (s, 9H), 1.34 - 1.12 (m, 5H). Step B. Procedure for preparation of tert-butyl 4-(3-ethoxy-2,2-difluoro-1- methylsulfanylcarbothioyloxy-3-oxo-propyl)piperidine-1-carbo xylate To a solution of tert-butyl 4-(3-ethoxy-2,2-difluoro-1-hydroxy-3-oxo-propyl)piperidine-1 - carboxylate (10 g, 29.64 mmol, 1 equiv.) in DMF (100 mL) was added DBU (18.05 g, 118.57 mmol, 17.87 mL, 4 equiv.) and CS2 (22.57 g, 296.42 mmol, 17.91 mL, 10 equiv.). The mixture was stirred at 20 °C for 1 hour. Then MeI (42.07 g, 296.42 mmol, 18.45 mL, 10 equiv.) was added to the mixture, and the mixture was stirred at 20 °C for 12 hours. The mixture was diluted with water (200 mL) and extracted with EtOAc (100 mL × 3). The combined organic layers were washed with brine (100 mL × 3), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~8% ethyl acetate/petroleum ether) to give compound tert-butyl 4-(3-ethoxy-2,2- difluoro-1-methylsulfanylcarbothioyloxy-3-oxo-propyl)piperid ine-1-carboxylate (11 g, 25.7 mmol, 86.8% yield) as a yellow oil. 1 H NMR (400 MHz, CDCl3) δ = 6.40 – 6.15 (m, 1H), 4.29 (q, J = 7.2 Hz,, 2H), 4.18 – 4.10 (m, 2H), 2.80 – 2.62 (m, 2H), 2.59 (s, 3H), 2.30 – 2.15 (m, 1H), 1.78 (t, J = 11.2 Hz, 2H), 1.45 (s, 10H), 1.33 (t, J = 7.2 Hz, 3H). Step C. Procedure for preparation of tert-butyl 4-(3-ethoxy-2,2-difluoro-3-oxo- propyl)piperidine-1-carboxylate A mixture of tert-butyl 4-(3-ethoxy-2,2-difluoro-1-methylsulfanylcarbothioyloxy-3-ox o- propyl)piperidine-1-carboxylate (10.5 g, 24.56 mmol, 1 equiv.), 2-tert-butylperoxy-2-methyl- propane (3.95 g, 27.02 mmol, 4.98 mL, 1.1 equiv.), phenylphosphonoylbenzene (5.96 g, 29.47 mmol, 1.2 equiv.) in dioxane (100 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 100 °C for 24 hours under N2 atmosphere. The reaction mixture was diluted with H2O (50 mL) and extracted with ethyl acetate (100mL × 2). The combined organic layers were washed with brine 50 mL (25 mL × 2), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~16% ethyl acetate/petroleum ether) to give compound tert- butyl 4-(3-ethoxy-2,2-difluoro-3-oxo-propyl)piperidine-1-carboxyla te (5.1 g, 15.8 mmol, 64.6% yield) as a colorless oil. 1 H NMR (400 MHz, CDCl3) δ = 4.37 - 4.31 (m, 2H), 4.07 (d, J = 12.8 Hz, 2H), 2.71 (t, J = 12.8 Hz, 2H), 2.12 - 1.93 (m, 2H), 1.81 - 1.71 (m, 3H), 1.46 (s, 9H), 1.37 (t, J = 7.2 Hz, 3H), 1.28 - 1.15 (m, 2H). Step D. Procedure for preparation of tert-butyl 4-(2,2-difluoro-3-hydroxy- propyl)piperidine-1-carboxylate A mixture of tert-butyl 4-(3-ethoxy-2,2-difluoro-3-oxo-propyl)piperidine-1-carboxyla te (5.1 g, 15.87 mmol, 1 equiv.) in THF (60 mL) was added LiAlH4 (602.27 mg, 15.87 mmol, 1 equiv.), and then the mixture was stirred at 0 °C for 1 hour under N 2 atmosphere. The reaction mixture was quenched by addition H 2 O (0.7 mL) at 0 °C. The aqueous phase was acidified to pH=4~5 with 1M HCl. The reaction mixture was filtered. The filter cake was diluted in ethyl acetate. The combined organic layers were dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~18% ethyl acetate/petroleum ether) to give compound tert-butyl 4-(2,2-difluoro-3- hydroxy-propyl)piperidine-1-carboxylate (3.5 g, 11.3 mmol, 71.1% yield, 90% purity) as a colorless oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 5.47 (t, J = 6.0 Hz, 1H), 3.89 (d, J = 12.4 Hz, 2H), 3.61 - 3.49 (m, 2H), 2.71 (s, 2H), 1.90 - 1.73 (m, 3H), 1.68 (d, J = 13.6 Hz, 2H), 1.38 (s, 9H), 1.14 - 1.00 (m, 2H). Step E. Procedure for preparation of tert-butyl 4-[3-(3-bromo-2-methyl-phenoxy)-2,2- difluoro-propyl]piperidine-1-carboxylate A mixture of tert-butyl 4-(2,2-difluoro-3-hydroxy-propyl)piperidine-1-carboxylate (500 mg, 1.79 mmol, 1 equiv.), 1,3-dibromo-2-methyl-benzene (894.77 mg, 3.58 mmol, 2 equiv.), 2,2,6,6-tetramethylheptane-3,5-dione (82.47 mg, 447.51 µmol, 92.14 µL, 0.25 equiv.), CuI (170.46 mg, 895.02 µmol, 0.5 equiv.) and Cs2CO3 (699.88 mg, 2.15 mmol, 1.2 equiv.) in NMP (5 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 120 °C for 12 hours under N2 atmosphere. The reaction mixture was quenched by addition water 50 mL, and then extracted with EtOAc (50 mL × 3). The combined organic layers were washed with brine (50 mL × 3), dried over anhydrous sulfate sodium, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate1/0 to 4/1) to give compound tert-butyl 4-[3-(3-bromo-2-methyl- phenoxy)-2,2-difluoro-propyl]piperidine-1-carboxylate (1.78 g, crude) as a yellow oil. MS (ESI) m/z: 394.2 [M-56+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 7.24 (d, J = 8.0 Hz, 1H), 7.13 (t, J = 8.4 Hz, 1H), 7.05 (d, J = 8.0 Hz, 1H), 4.40 - 4.25 (m, 2H), 3.89 (d, J = 12.0 Hz, 2H), 2.83 - 2.61 (m, 2H), 1.99 (s, 3H), 1.90 - 1.78 (m, 1H), 1.76 - 1.64 (m, 2H), 1.38 (s, 9H), 1.16 - 1.01 (m, 4H). Step F. Procedure for preparation of 4-[3-(3-bromo-2-methyl-phenoxy)-2,2-difluoro- propyl]piperidine A solution of tert-butyl 4-[3-(3-bromo-2-methyl-phenoxy)-2,2-difluoro-propyl]piperidi ne-1- carboxylate (1.78 g, 3.97 mmol, 1 equiv.) in HCl/EtOAc (4 M, 15 mL, 15.11 equiv.) was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give 4- [3-(3-bromo-2-methyl-phenoxy)-2,2-difluoro-propyl]piperidine (2 g, crude) as a yellow solid. MS (ESI) m/z: 348.2 [M+H] + Step G. Procedure for preparation of ethyl 2-[4-[3-(3-bromo-2-methyl-phenoxy)-2,2- difluoro-propyl]-1-piperidyl]acetate To a solution of 4-[3-(3-bromo-2-methyl-phenoxy)-2,2-difluoro-propyl]piperidi ne (500 mg, 1.44 mmol, 1 equiv.) and ethyl 2-bromoacetate (239.79 mg, 1.44 mmol, 158.80 µL, 1 equiv.) in CH 3 CN (5 mL) was added K 2 CO 3 (595.33 mg, 4.31 mmol, 3 equiv.). The mixture was stirred at 60 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate1/0 to 1/1) to give compound ethyl 2-[4-[3-(3-bromo-2-methyl-phenoxy)- 2,2-difluoro-propyl]-1-piperidyl]acetate (380 mg, 874.9 µmol, 60.9% yield) as a yellow oil. MS (ESI) m/z: 436.0 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 7.23 (d, J = 8.0 Hz, 1H), 7.12 (t, J = 8.0 Hz, 1H), 7.05 (d, J = 7.6 Hz, 1H), 4.38 - 4.25 (m, 2H), 4.06 (q, J = 7.2 Hz, 2H), 3.15 (s, 2H), 2.78 (d, J = 10.8 Hz, 2H), 2.25 (s, 2H), 2.14 (t, J = 11.2 Hz, 2H), 2.06 - 1.91 (m, 2H), 1.72 - 1.63 (m, 2H), 1.34 - 1.21 (m, 2H), 1.21 - 1.14 (m, 5H). Step H. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-(2-ethoxy-2-oxo- ethyl)-4-piperidyl]-2,2- difluoro-propoxy]-2-methyl-phenyl]pyridine-2-carboxylate Ethyl 2-[4-[3-(3-bromo-2-methyl-phenoxy)-2,2-difluoro-propyl]-1-pi peridyl]acetate (380 mg, 874.94 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (535.94 mg, 874.94 µmol, 1 equiv.), [2-(2-aminophenyl)phenyl]palladium(1+);bis(1- adamantyl)-butyl-phosphane;methanesulfonate (63.71 mg, 874.94 µmol, 0.1 equiv.), and KF (1.5 M, 1.75 mL, 3 equiv.) were taken up into a microwave tube in dioxane (2 mL). The sealed tube was heated at 100 °C for 60 minutes under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate10/1 to 1/1) to give compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[1-(2- ethoxy-2-oxo-ethyl)-4-piperidyl]-2,2-difluoro-propoxy]-2-met hyl-phenyl]pyridine-2- carboxylate (512 mg, 609.5 µmol, 69.6% yield) as a yellow solid. MS (ESI) m/z: 840.5 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ,) δ = 12.92 - 12.81 (m, 1H), 8.02 (d, J = 8.4 Hz, 1H), 7.77 (d, J = 8.4 Hz, 1H), 7.60 (d, J = 7.6 Hz, 1H), 7.45 (d, J = 8.8 Hz, 3H), 7.40 - 7.31 (m, 3H), 7.14 (t, J = 8.0 Hz, 1H), 7.00 (d, J = 8.4 Hz, 1H), 6.95 (d, J = 8.4 Hz, 1H), 6.65 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.35 - 4.21 (m, 2H), 4.06 - 4.03 (m, 2H), 3.87 (t, J = 5.6 Hz, 2H), 3.14 (s, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.76 (d, J = 11.2 Hz, 2H), 2.12 (t, J = 10.8 Hz, 2H), 1.99 (s, 3H), 1.89 (s, 3H), 1.71 - 1.58 (m, 3H), 1.17 (d, J = 2.0 Hz, 3H), 1.00 (s, 9H). Step I. Procedure for preparation of 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyr idyl]-2-methyl-phenoxy]- 2,2-difluoro-propyl]-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[3-[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]-2,2-dif luoro-propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (512 mg, 609.53 µmol, 1 equiv.) in H2O (2.5 mL) and THF (2.5 mL) was added LiOH (72.99 mg, 3.05 mmol, 5 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was quenched by addition HCl (1M, 10 mL) and then extracted with CH2Cl2 (20 mL × 3). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give compound 2-[4-[3-[3-[6-[8- (1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin- 2-yl]-2-tert-butoxycarbonyl-3- pyridyl]-2-methyl-phenoxy]-2,2-difluoro-propyl]-1-piperidyl] acetic acid (600 mg, crude) as a yellow solid. Step J. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-diox o-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-4-piperidyl]-2,2-difluoro-p ropoxy]-2-methyl- phenyl]pyridine-2-carboxylate To a solution of 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]-2,2-difluoro-propyl]- 1-piperidyl]acetic acid (60 mg, 73.90 µmol, 1 equiv.) and 3-(6-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (20.99 mg, 81.29 µmol, 1.1 equiv.) in DMF (1 mL) was added DIEA (28.65 mg, 221.69 µmol, 38.61 µL, 3 equiv.) and HATU (42.15 mg, 110.85 µmol, 1.5 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was quenched by addition water 10 mL, filtered, and concentrated under reduced pressure to give compound tert-butyl 6- [8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinol in-2-yl]-3-[3-[3-[1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-4-piperidyl]-2,2-difluoro- propoxy]-2-methyl-phenyl]pyridine-2-carboxylate (120 mg, crude) as a yellow solid. MS (ESI) m/z: 1052.5 [M+H] + Step K. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]-2,2-difluoro-propoxy]-2- methyl-phenyl]pyridine-2- carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-ind azol-6-yl]amino]-2-oxo-ethyl]-4- piperidyl]-2,2-difluoro-propoxy]-2-methyl-phenyl]pyridine-2- carboxylate (120 mg, 114.05 µmol, 1 equiv.) in DCM (1 mL) was added TFA (13.00 mg, 114.05 µmol, 8.44 µL, 1 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[3-[1-[2-[[3-(2,6-dioxo- 3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]-4-pip eridyl]-2,2-difluoro-propoxy]- 2-methyl-phenyl]pyridine-2-carboxylic acid (35.7 mg, 34.8 µmol, 30.5% yield, 97.1% purity) as a white solid. MS (ESI) m/z: 996.8 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ,) δ = 10.88 (s, 1H), 9.82 (s, 1H), 8.06 - 8.00 (m, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 8.8 Hz, 2H), 7.49 - 7.42 (m, 3H), 7.40 - 7.31 (m, 2H), 7.24 - 7.18 (m, 1H), 7.12 (t, J = 8.0 Hz, 1H), 7.00 - 6.92 (m, 2H), 6.71 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.35 - 4.24 (m, 3H), 3.95 - 3.88 (m, 5H), 3.12 (s, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.86 (d, J = 11.2 Hz, 2H), 2.68 - 2.59 (m, 2H), 2.39 - 2.28 (m, 2H), 2.23 - 2.14 (m, 3H), 2.11 - 1.97 (m, 2H), 1.93 (s, 3H), 1.78 - 1.71 (m, 2H), 1.49 - 1.35 (m, 2H). Example 42. Preparation of Compound 187a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((3S)-4-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino )-2-oxoethyl)-3- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linic acid Step A. Procedure for preparation of (S)-tert-butyl 4-(2-ethoxy-2-oxoethyl)-3- (trifluoromethyl)piperazine-1-carboxylate A mixture of (S)-tert-butyl 3-(trifluoromethyl) piperazine-1-carboxylate (300 mg, 1.18 mmol, 1 equiv.), ethyl 2-bromoacetate (1.18 g, 7.08 mmol, 782.99 µL, 6 equiv.) and DIEA (762.48 mg, 5.90 mmol, 1.03 mL, 5 equiv.) in THF (3 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 60 °C for 3 hours under N 2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (20 mL × 3). The combined organic layers were washed with brine (20 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~20% ethyl acetate/petroleum ether) to give (S)- tert-butyl 4-(2-ethoxy-2-oxoethyl)-3-(trifluoromethyl)piperazine-1-carb oxylate (140 mg, 411.36 µmol, 34.86% yield) as a colorless oil. 1 H NMR (400 MHz, CD 3 Cl) δ = 4.20 - 4.11 (m, 2H), 4.01 - 3.79 (m, 1H), 3.76 - 3.61 (m, 2H), 3.60 - 3.50 (m, 1H), 3.49 - 3.40 (m, 2H), 3.39 -3.24 (m, 1H), 3.04 - 2.77 (m, 2H), 1.43 (s, 9H), 1.25 (t, J = 7.2 Hz, 3H). Step B. Procedure for preparation of (S)-ethyl 2-(2-(trifluoromethyl)piperazin-1-yl) acetate A mixture of (S)-tert-butyl 4-(2-ethoxy-2-oxoethyl)-3-(trifluoromethyl)piperazine-1- carboxylate (140 mg, 411.36 µmol, 1 equiv.) in HCl/dioxane (2 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 25 °C for 4 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give (S)-ethyl 2-(2- (trifluoromethyl)piperazin-1-yl) acetate (150 mg, crude) as a yellow solid. Step C. Procedure for preparation of 1-bromo-3-(3-bromopropoxy)-2-methylbenzene A mixture of 1,3-dibromopropane (16.19 g, 80.20 mmol, 8.18 mL, 5 equiv.), 3-bromo-2- methylphenol (3 g, 16.04 mmol, 1 equiv.), and K 2 CO 3 (6.65 g, 48.12 mmol, 3 equiv.) in CH 3 CN (30 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 70 °C for 16 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~5% ethyl acetate/petroleum ether) to give compound 1-bromo-3-(3-bromopropoxy)-2- methylbenzene (3.6 g, 11.69 mmol, 72.87% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.20 - 7.15 (m, 1H), 7.10 (t, J = 8.0 Hz, 1H), 6.99 (d, J = 8.0 Hz, 1H), 4.09 (t, J = 6.0 Hz, 2H), 3.69 (t, J = 6.4 Hz, 2H), 3.61 (t, J = 6.4 Hz, 1H), 2.30 - 2.26 (m, 2H), 2.24 (s, 3H). Step D. Procedure for preparation of (S)-ethyl 2-(4-(3-(3-bromo-2- methylphenoxy)propyl)-2-(trifluoromethyl)piperazin-1-yl)acet ate A mixture of (S)-ethyl 2-(2-(trifluoromethyl)piperazin-1-yl)acetate (150 mg, 542.14 µmol, 1 equiv., HCl), 1-bromo-3-(3-bromopropoxy)-2-methylbenzene (200.38 mg, 650.56 µmol, 1.2 equiv.), and K 2 CO 3 (449.56 mg, 3.25 mmol, 6 equiv.) in CH 3 CN (4 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 60 °C for 16 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~30% ethyl acetate/petroleum ether) to give compound (S)-ethyl 2-(4-(3-(3-bromo-2- methylphenoxy)propyl)-2-(trifluoromethyl)piperazin-1-yl)acet ate (100 mg, 205.1 µmol, 37.5% yield, 95.8% purity) as a colorless oil. MS (ESI) m/z: 469.0 [M+H] + Step E. Procedure for preparation of (S)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(4-(2-ethoxy-2-oxoe thyl)-3- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linate (S)-ethyl 2-(4-(3-(3-bromo-2-methylphenoxy)propyl)-2-(trifluoromethyl) piperazin-1- yl)acetate (100 mg, 213.99 µmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3, 2-dioxaborolan-2-yl)picolinate (157.29 mg, 256.78 µmol, 1.2 equiv.), Ad2nBuP Pd G3(cataCXium® A Pd G3 ) (15.58 mg, 21.40 µmol, 0.1 equiv.), and KF (1.5 M, 427.97 uL, 3 equiv.) were taken up into a microwave tube in dioxane (2.5 mL) .The sealed tube was heated at 100 °C for 1 hour under microwave. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (25 mL × 3). The combined organic layers were washed with brine (15 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , petroleum ether : ethyl acetate= 2:1) to give (S)-tert-butyl 6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2( 1H)-yl)-3-(3-(3-(4-(2-ethoxy- 2-oxoethyl)-3-(trifluoromethyl)piperazin-1-yl)propoxy)-2-met hylphenyl)picolinate (150 mg, 171.82 µmol, 80.30% yield) as a yellow oil. MS (ESI) m/z: 873.3 [M+H] + Step F. Procedure for preparation of (S)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)propyl)-2-(trifluoromethyl)piperazin-1-yl)acet ic acid A mixture of (S)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4-(2-ethoxy-2-oxoethyl)-3-(trifluoromethy l)piperazin-1-yl)propoxy)-2- methylphenyl)picolinate (155 mg, 177.55 µmol, 1 equiv.) and LiOH·H2O (22.35 mg, 532.65 µmol, 3 equiv.) in THF (2 mL) and H2O (0.5 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 16 hours under N 2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (25 mL × 3). The combined organic layers were washed with brine (15 mL × 3), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give compound (S)-2-(4-(3-(3-(6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)propyl)-2-(trif luoromethyl)piperazin-1- yl)acetic acid (150 mg, crude) as a yellow solid. MS (ESI) m/z: 845.3 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 13.06 - 12.60 (m, 1H), 10.73 - 10.30 (m, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.60 (d, J = 7.6 Hz, 1H), 7.49 - 7.41 (m, 3H), 7.40 - 7.31 (m, 2H), 7.12 (t, J = 8.0 Hz, 1H), 6.98 - 6.90 (m, 2H), 6.61 (d, J = 7.6 Hz, 1H), 4.98 (d, J = 4.0 Hz, 2H), 4.38 - 4.13 (m, 1H), 3.87 (t, J = 5.6 Hz, 2H), 3.64 - 3.47 (m, 4H), 3.09 - 2.97 (m, 4H), 2.28 - 2.10 (m, 2H), 1.99 (s, 3H), 1.90 (s, 2H), 1.17 (t, J = 7.2 Hz, 4H), 1.02 (s, 9H). Step G. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3S)-4-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-3-(trifluoromethyl)pipera zin-1-yl)propoxy)-2- methylphenyl)picolinate A mixture of (S)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)-2- (trifluoromethyl)piperazin-1-yl)acetic acid (75 mg, 88.76 µmol, 1 equiv.), 3-(6-amino-1- methyl-indazol-3-yl)piperidine-2,6-dione (25.22 mg, 97.64 µmol, 1.1 equiv.), and DIEA (34.42 mg, 266.29 µmol, 46.38 µL, 3 equiv.) in DMF (1 mL) was degassed and purged with N 2 three times. The mixture was then stirred at 25°C for 5 minutes. After 5 minutes, HATU (40.50 mg, 106.52 µmol, 1.2 equiv.) was added, and the mixture was stirred at 25 °C for 1 hour under N2 atmosphere. The resulting reaction mixture was diluted with water (20 mL) and extracted with EtOAc (20 mL × 3). The combined organic layers were washed with brine (20 mL × 6), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((3S)-4-(2-((3-(2,6-dioxopiperidin-3-yl)-1 -methyl-1H-indazol-6-yl)amino)- 2-oxoethyl)-3-(trifluoromethyl)piperazin-1-yl)propoxy)-2-met hylphenyl)picolinate (100 mg, crude) as a brown solid. MS (ESI) m/z: 1085.4 [M+H] + Step H. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3S)-4-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-3-(trifluoromethyl)pipera zin-1-yl)propoxy)-2- methylphenyl)picolinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-((3S)-4-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-6-yl)amino)-2- oxoethyl)-3-(trifluoromethyl)piperazin-1-yl)propoxy)-2-methy lphenyl)picolinate (100 mg, 92.15 µmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL) as degassed and purged with N 2 three times, and then the mixture was stirred at 25 °C for 1 hour under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give compound 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3S)-4-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)-3-(trifluoromethyl)piperazin -1-yl)propoxy)-2- methylphenyl)picolinic acid (32.1 mg, 30.7 µmol, 33.4% yield, 98.4% purity) as an off-white solid. MS (ESI) m/z: 1029.4 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 13.03 - 12.32 (m, 2H), 10.90 (s, 1H), 9.79 (s, 1H), 8.14 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.65 - 7.57 (m, 2H), 7.50 - 7.41 (m, 3H), 7.40 - 7.32 (m, 2H), 7.25 - 7.19 (m, 1H), 7.13 - 7.04 (m, 2H), 6.96 (d, J = 8.8 Hz, 1H), 6.86 (d, J = 8.0 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.41 - 4.34 (m, 1H), 4.08 (s, 3H), 4.01 (t, J = 5.6 Hz, 2H), 3.91 (t, J = 5.6 Hz, 2H), 3.79 - 3.67 (m, 2H), 3.48 (d, J = 16.8 Hz, 1H), 3.06 -2.97 (t, J = 5.2 Hz, 3H), 2.87 - 2.75 (m, 2H), 2.75 - 2.62 (m, 2H), 2.61 - 2.53 (m, 3H), 2.48 -2.41 (m, 2H), 2.40 - 2.31 (m, 1H), 2.21 - 2.12 (m, 1H), 1.96 - 1.85 (m, 5H). Example 43. Preparation of Compound 187b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((3R)-4-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino )-2-oxoethyl)-3- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linic acid Step A. Procedure for preparation of (R)-tert-butyl 4-(2-ethoxy-2-oxoethyl)-3- (trifluoromethyl)piperazine-1-carboxylate To a solution of (R)-tert-butyl 3-(trifluoromethyl)piperazine-1-carboxylate (700 mg, 2.75 mmol, 1 equiv.) in THF (5 mL) was added DIEA (711.65 mg, 5.51 mmol, 959.10 µL, 2.0 equiv.) and ethyl 2-bromoacetate (597.73 mg, 3.58 mmol, 395.84 µL, 1.3 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove solvent to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=10/1 to 4/1) to give a compound (R)-tert- butyl 4-(2-ethoxy-2-oxoethyl)-3-(trifluoromethyl)piperazine-1-carb oxylate (300 mg, 537.7 µmol, 45.7% yield, 61% purity) as a white oil. MS (ESI) m/z: 240.5 [M-100+1] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 4.14 - 4.01 (m, 2H), 3.70 - 3.52 (m, 3H), 3.32 (s, 3H), 1.38 (s, 9H), 1.23 - 1.11 (m, 4H), 0.94 (d, J = 6.4 Hz, 2H) Step B. Procedure for preparation of (R)-ethyl 2-(2-(trifluoromethyl)piperazin-1- yl)acetate To a solution of (R)-tert-butyl 4-(2-ethoxy-2-oxoethyl)-3-(trifluoromethyl)piperazine-1- carboxylate (250 mg, 734.56 µmol, 1 equiv.) was added HCl/dioxane (4 M, 183.64 µL, 1 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to remove solvent and provide (R)-ethyl 2-(2- (trifluoromethyl)piperazin-1-yl)acetate (240 mg, crude, HCl) as a white solid. Step C. Procedure for preparation of (R)-ethyl 2-(4-(3-(3-bromo-2- methylphenoxy)propyl)-2-(trifluoromethyl)piperazin-1-yl)acet ate To a solution of 1-bromo-3-(3-bromopropoxy)-2-methylbenzene (307.72 mg, 999.08 µmol, 1.2 equiv.) in CH 3 CN (10 mL) was added K 2 CO 3 (345.20 mg, 2.50 mmol, 3 equiv.) and (R)-ethyl 2-(2-(trifluoromethyl)piperazin-1-yl)acetate (200 mg, 832.56 µmol, 1 equiv.). The mixture was stirred at 60 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The resulting residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=10/1 to 4/1) to give (R)-ethyl 2-(4-(3-(3-bromo-2- methylphenoxy)propyl)-2-(trifluoromethyl)piperazin-1-yl)acet ate (200 mg, 425.53 µmol, 51.11% yield, 99.43% purity) as a white oil. MS (ESI) m/z: 469.3 [M+H] + Step D. Procedure for preparation of (R)-tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(4-(2- ethoxy-2-oxoethyl)-3- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linate To a solution of (R)-ethyl 2-(4-(3-(3-bromo-2-methylphenoxy)propyl)-2- (trifluoromethyl)piperazin-1-yl)acetate (180 mg, 385.18 µmol, 1 equiv.) in dioxane (5 mL) was added K2CO3 (1.5 M, 385.18 µL, 1.5 equiv.), [2-(2-aminophenyl)phenyl]palladium(1+); bis(1- adamantyl)-butyl-phosphane;methanesulfonate (56.10 mg, 77.04 µmol, 0.2 equiv.), and tert- butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)picolinate (283.13 mg, 462.22 µmol, 1.2 equiv.). The mixture was stirred at 100 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=10/1 to 2/1) to give (R)-tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(4-(2- ethoxy-2-oxoethyl)-3- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linate (160 mg, 174.1 µmol, 45.2% yield, 95% purity) as a colorless oil. MS (ESI) m/z: 873.7 [M+H] + Step E. Procedure for preparation of (R)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)propyl)-2-(trifluoromethyl)piperazin-1-yl)acet ic acid To a solution of (R)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4-(2-ethoxy-2-oxoethyl)-3-(trifluoromethy l)piperazin-1-yl)propoxy)-2- methylphenyl)picolinate (150 mg, 171.82 µmol, 1 equiv.) in THF (4 mL) was added LiOH (12.34 mg, 515.47 µmol, 3 equiv.) and H 2 O (1 mL). The mixture was stirred at 25 °C for 2 hours. The pH was adjusted to 6 with 1M HCl. The residue was diluted with H 2 O (10 mL) and extracted with DCM (10 mL × 3). The combined organic layers were washed with aqueous brine (10 mL × 3), dried over Na2SO4, filtered and concentrated under reduced pressure to give a compound (R)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)-2- (trifluoromethyl)piperazin-1-yl)acetic acid (130 mg, crude) was obtained as a yellow solid. MS (ESI) m/z: 845.5 [M+H] + Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3R)-4-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-3-(trifluoromethyl)pipera zin-1-yl)propoxy)-2- methylphenyl)picolinate To a solution of (R)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)-2- (trifluoromethyl)piperazin-1-yl)acetic acid (65 mg, 76.93 µmol, 1 equiv.) in DMF (2 mL) was added HATU (35.10 mg, 92.31 µmol, 1.2 equiv.), 3-(6-amino-1-methyl-1H-indazol-3- yl)piperidine-2,6-dione (23.84 mg, 92.31 µmol, 1.2 equiv.), and DIEA (29.83 mg, 230.79 µmol, 40.20 µL, 3 equiv.). The mixture was stirred at 25 °C for 1 hour. The residue was diluted with H2O (5 mL) and extracted with DCM (5 mL × 3). The combined organic layers were washed with aqueous brine (5 mL × 3), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a compound tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3R)-4-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)-3-(trifluoromethyl)piperazin -1-yl)propoxy)-2- methylphenyl)picolinate (100 mg, crude) as a brown oil. MS (ESI) m/z: 543.1 [(M+H)/2] + Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3R)-4-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-3-(trifluoromethyl)pipera zin-1-yl)propoxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((3R)-4-(2-((3-(2,6-dioxopiperidin-3-yl)-1 -methyl-1H-indazol-6-yl)amino)- 2-oxoethyl)-3-(trifluoromethyl)piperazin-1-yl)propoxy)-2-met hylphenyl)picolinate (100 mg, 92.15 µmol, 1 equiv.) in DCM (1 mL) was added TFA (10.51 mg, 92.15 µmol, 6.82 µL, 1 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by prep-HPLC to give a compound 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3- ((3R)-4-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)amino)-2-oxoethyl)-3- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linic acid (37.52 mg, 35.6 µmol, 38.8% yield, 97.7% purity) as a yellow gum. MS (ESI) m/z: 1029.8 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.87 (d, J = 2.0 Hz, 1H), 12.73 - 12.39 (m, 1H), 10.88 (s, 1H), 9.90 (s, 1H), 8.10 - 8.00 (m, 2H), 7.79 (d, J = 7.6 Hz, 1H), 7.67 - 7.59 (m, 2H), 7.49 - 7.31 (m, 5H), 7.16 - 7.06 (m, 2H), 6.98 (d, J = 8.8 Hz, 1H), 6.88 (d, J = 8.0 Hz, 1H), 6.64 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.35 – 4.28 (m, 1H), 4.02 (t, J = 5.6 Hz, 2H), 3.96 - 3.89 (m, 5H), 3.76 - 3.61 (m, 2H), 3.49 (m, 2H), 3.08 - 2.88 (m, 4H), 2.86 - 2.71 (m, 2H), 2.69 - 2.56 (m, 4H), 2.34 - 2.31 (m, 2H), 2.20 - 2.13 (m, 1H), 1.96 - 1.85 (m, 5H) Example 44. Preparation of Compound 190 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(3-(1-(2-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin -1-yl)ethyl)piperidin-4- yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 4-(3-(4-bromo-3- methylphenoxy)propyl)piperidine-1-carboxylate To a solution of tert-butyl 4-(3-bromopropyl)piperidine-1-carboxylate (2.0 g, 6.53 mmol, 1.0 equiv.) in CH3CN (20 mL) was added K2CO3 (2.71 g, 19.59 mmol, 3.0 equiv.) and 4-bromo-3- methyl-phenol (1.83 g, 9.80 mmol, 1.5 equiv.). The mixture was stirred at 60 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give tert-butyl 4-(3-(4- bromo-3-methylphenoxy)propyl)piperidine-1-carboxylate (2.5 g, crude) as a yellow solid. MS (ESI) m/z: 313.8 [M-100+H] + . Step B. Procedure for preparation of 4-(3-(4-bromo-3-methylphenoxy)propyl)piperidine To a solution of tert-butyl 4-(3-(4-bromo-3-methylphenoxy)propyl)piperidine-1-carboxylat e (2.5 g, 6.06 mmol, 1.0 equiv.) in HCl/EtOAc (15 mL). The mixture was stirred at 25 °C for 1 hour. The mixture was filtered and concentrated under reduced pressure to give compound 4- (3-(4-bromo-3-methylphenoxy)propyl)piperidine (1.2 g, 3.4 mmol, 56.7% yield) as an off- white solid. Step C. Procedure for preparation of 4-[3-(4-bromo-3-methyl-phenoxy)propyl]-1-(2,2- diethoxyethyl)piperidine To a solution of 4-(3-(4-bromo-3-methylphenoxy)propyl)piperidine (500 mg, 1.43 mmol, 1.0 equiv., HCl) in CH3CN (5 mL) was added K2CO3 (594.51 mg, 4.30 mmol, 3.0 equiv.) and 2- bromo-1,1-diethoxyethane (282.57 mg, 1.43 mmol, 215.70 µL, 1.0 equiv.). The mixture was stirred at 80 °C for 5 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by flash silica gel chromatography (Eluent of 0~5% DCM/MeOH) to give 4-[3-(4-bromo-3-methyl-phenoxy)propyl]-1-(2,2- diethoxyethyl)piperidine (400 mg, 924.3 µmol, 64.4% yield, 99% purity) as a white solid. MS (ESI) m/z: 430.0 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 7.42 (d, J = 8.8 Hz, 1H), 6.94 (d, J = 2.8 Hz, 1H), 6.73 - 6.68 (m, 1H), 4.55 (t, J = 5.2 Hz, 1H), 3.91 (t, J = 6.4 Hz, 2H), 3.62 - 3.52 (m, 2H), 3.50 - 3.39 (m, 3H), 2.85 (d, J = 11.2 Hz, 2H), 2.36 (d, J = 5.2 Hz, 2H), 2.29 (s, 3H), 1.95 (t, J = 11.2 Hz, 2H), 1.73 - 1.64 (m, 2H), 1.60 (d, J = 11.6 Hz, 2H), 1.36 - 1.25 (m, 2H), 1.10 (t, J = 7.2 Hz, 8H) Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(3-(1-(2,2-diethoxyethyl)p iperidin-4-yl)propoxy)-2- methylphenyl)picolinate A mixture of 4-[3-(4-bromo-3-methyl-phenoxy)propyl]-1-(2,2-diethoxyethyl) piperidine (300 mg, 700.28 µmol, 1.0 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)picolinate (514.74 mg, 840.33 µmol, 1.2 equiv.), and KF (1.5 M, 1.40 mL, 3.0 equiv.) in dioxane (3 mL) was degassed and purged with N 2 three times, and then [2-(2- aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-butyl-phos phane;methanesulfonate (51.00 mg, 70.03 µmol, 0.1 equiv.) was added into the mixture. The resulting mixture was stirred at 100 °C for 1 hour under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by flash silica gel chromatography (Eluent of 0~100% ethyl acetate/petroleum ether) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(4-(3-(1-(2,2- diethoxyethyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolin ate (400 mg, 479.5 µmol, 68.4% yield) as a yellow oil. MS (ESI) m/z: 834.2 [M+H] + . Step E. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(2-methyl-4-(3-(1-(2-oxoethyl )piperidin-4- yl)propoxy)phenyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4-(3-(1-(2,2-diethoxyethyl)piperidin-4-yl)propoxy)-2- methylphenyl)picolinate (200 mg, 239.79 µmol, 1 equiv.) in HCOOH (1.5 mL) was stirred at 105 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(2-methyl-4- (3-(1-(2-oxoethyl)piperidin-4- yl)propoxy)phenyl)picolinic acid (150 mg, crude) as a yellow solid. MS (ESI) m/z: 704.4 [M+H] + Step F. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(3-(1-(2-(4-(3-(2,6-dioxop iperidin-3-yl)-1-methyl-1H- indazol-7-yl)piperazin-1-yl)ethyl)piperidin-4-yl)propoxy)-2- methylphenyl)picolinic acid To a solution of 3-(1-methyl-6-(piperazin-1-yl)-1H-indazol-3-yl)piperidine-2, 6-dione (93.05 mg, 255.74 µmol, 1.2 equiv., HCl) in DCM (3 mL) was added NaBH(OAc)3 (135.50 mg, 639.34 µmol, 3.0 equiv.) and 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2-methyl-4-(3-(1-(2-oxoethyl)piperidin-4-yl)pro poxy)phenyl)picolinic acid (150 mg, 213.11 µmol, 1.0 equiv.) at 0 °C. The mixture was stirred at 0 °C for 1 hour. Then the mixture was stirred at 25 °C for 11 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(4-(3-(1-(2-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin-1-yl )ethyl)piperidin-4-yl)propoxy)- 2-methylphenyl)picolinic acid (22.6 mg, 21.8 µmol, 10.2% yield, 97.7% purity) as a white solid. MS (ESI) m/z: 1015.7 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.88 (s, 1H), 8.01 (d, J = 8.0 Hz, 1H), 7.77 (d, J = 8.0 Hz, 1H), 7.61 (d, J = 7.2 Hz, 1H), 7.48 - 7.30 (m, 6H), 7.01 (d, J = 4.4 Hz, 2H), 6.94 (d, J = 8.4 Hz, 1H), 6.88 (d, J = 9.2 Hz, 1H), 6.77 (s, 1H), 6.71 - 6.66 (m, 1H), 4.95 (s, 2H), 4.36 - 4.30 (m, 1H), 4.23 (s, 3H), 3.94 (s, 2H), 3.90 - 3.86 (m, 2H), 2.99 (s, 6H), 2.93 - 2.77 (m, 4H), 2.70 - 2.53 (m, 8H), 2.36 - 2.26 (m, 2H), 2.17 - 2.10 (m, 2H), 2.04 (s, 3H), 1.69 (d, J = 6.8 Hz, 2H), 1.66 - 1.56 (m, 2H), 1.30 (s, 2H), 1.25 - 1.20 (m, 1H), 1.18 - 1.06 (m, 2H) Example 45. Preparation of Compound 193a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((4R)-1-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino )-2-oxoethyl)-3,3- difluoropiperidin-4-yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of (R)-4-(3-(3-bromo-2-methylphenoxy)propyl)-3,3- difluoropiperidine To a solution of (R)-tert-butyl 4-(3-(3-bromo-2-methylphenoxy)propyl)-3,3- difluoropiperidine-1-carboxylate (500.00 mg, 1.12 mmol, 1 equiv.) was added HCl/dioxane (4 M, 278.81 µL, 1 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The compound (R)-4-(3-(3- bromo-2-methylphenoxy)propyl)-3,3-difluoropiperidine (350 mg, crude, HCl) was obtained as a white solid. Step B. Procedure for preparation of (R)-ethyl 2-(4-(3-(3-bromo-2- methylphenoxy)propyl)-3,3-difluoropiperidin-1-yl)acetate To a solution of (R)-4-(3-(3-bromo-2-methylphenoxy)propyl)-3,3-difluoropiperi dine (700.00 mg, 2.01 mmol, 1 equiv.) in CH 3 CN (10 mL) was added K 2 CO 3 (833.46 mg, 6.03 mmol, 3 equiv.) and ethyl 2-bromoacetate (302.13 mg, 1.81 mmol, 200.09 µL, 0.9 equiv.). The mixture was stirred at 60 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate 1/0 to 2/1). The compound (R)-ethyl 2-(4-(3-(3-bromo-2- methylphenoxy)propyl)-3,3-difluoropiperidin-1-yl)acetate (600 mg, 1.3 mmol, 68.7% yield, 100% purity) was obtained as a white solid. MS (ESI) m/z: 436.0 [M+H] + Step C. Procedure for preparation of (R)-tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2- ethoxy-2-oxoethyl)-3,3- difluoropiperidin-4-yl)propoxy)-2-methylphenyl)picolinate To a solution of (R)-ethyl 2-(4-(3-(3-bromo-2-methylphenoxy)propyl)-3,3-difluoropiperid in- 1-yl)acetate (400 mg, 920.99 µmol, 1 equiv.) in dioxane (10 mL) was added [2-(2- aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-butyl-phos phane;methanesulfonate (67.07 mg, 92.10 µmol, 0.2 equiv.), K2CO3 (1 M, 2.76 mL, 3 equiv.) and tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)picolinate (564.15 mg, 920.99 µmol, 1 equiv.). The mixture was stirred at 100 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=10/1 to 2/1). The compound (R)-tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2- ethoxy-2-oxoethyl)-3,3- difluoropiperidin-4-yl)propoxy)-2-methylphenyl)picolinate (600 mg, 707.1 µmol, 76.8% yield, 99% purity) was obtained as a yellow solid. MS (ESI) m/z: 840.3 [M+H] + Step D. Procedure for preparation of (R)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)propyl)-3,3-difluoropiperidin-1-yl)acetic acid To a solution of (R)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-ethoxy-2-oxoethyl)-3,3-difluoropiper idin-4-yl)propoxy)-2- methylphenyl)picolinate (600 mg, 714.30 µmol, 1 equiv.) in THF (10 mL) and H2O (4 mL) was added LiOH•H 2 O (51.32 mg, 2.14 mmol, 3 equiv.). The mixture was stirred at 25 °C for 1 hour. The pH was adjusted to 6 with 1 M HCl. The residue was diluted with H2O (10 mL) and extracted with (10 mL × 3). The combined organic layers were washed with aqueous NaCl (10 mL × 3), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The compound (R)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)-3,3- difluoropiperidin-1-yl)acetic acid (500 mg, crude) was obtained as a yellow solid. MS (ESI) m/z: 812.8 [M+H] + Step E. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((4R)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-3,3-difluoropiperidin-4-y l)propoxy)-2- methylphenyl)picolinate To a solution of (R)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)-3,3- difluoropiperidin-1-yl)acetic acid (100.00 mg, 123.16 µmol, 1 equiv.) in DMF (2 mL) was added HATU (56.20 mg, 147.79 µmol, 1.2 equiv.), DIEA (47.75 mg, 369.48 µmol, 64.36 µL, 3 equiv.), and 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (38.17 mg, 147.79 µmol, 1.2 equiv.). The mixture was stirred at 25 °C for 1 hour. The mixture was diluted with H2O (10 mL) and extracted with DCM (10 mL × 3). The combined organic layers were washed with aqueous NaCl (10mL × 3), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The compound tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((4R)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)-3,3-difluoropiperidin-4-yl)p ropoxy)-2- methylphenyl)picolinate (120 mg, crude) was obtained as a brown oil. MS (ESI) m/z: 527.2 [(M+2)/2] + Step F. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((4R)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-3,3-difluoropiperidin-4-y l)propoxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((4R)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1 -methyl-1H-indazol-6- yl)amino)-2-oxoethyl)-3,3-difluoropiperidin-4-yl)propoxy)-2- methylphenyl)picolinate (120.00 mg, 114.05 µmol, 1 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 118.42 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC. The compound 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (3-((4R)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-inda zol-6-yl)amino)-2-oxoethyl)- 3,3-difluoropiperidin-4-yl)propoxy)-2-methylphenyl)picolinic acid (28.9 mg, 28.3 µmol, 24.8% yield, 97.7% purity) was obtained as a yellow solid. MS (ESI) m/z: 996.7 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.87 (d, J = 5.2 Hz, 1H), 12.74 - 12.23 (m, 1H), 10.88 (s, 1H), 10.04 - 9.85 (m, 1H), 8.08 - 8.00 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.66 - 7.59 (m, 2H), 7.50 - 7.42 (m, 3H), 7.40 - 7.32 (m, 2H), 7.14 - 7.06 (m, 2H), 6.97 (d, J = 8.8 Hz, 1H), 6.89 (d, J = 8.4 Hz, 1H), 6.64 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.32 (dd, J = 5.2, 9.6 Hz, 1H), 3.99 (s, 2H), 3.94 - 3.89 (m, 5H), 3.03 (t, J = 5.6 Hz, 2H), 2.74 - 2.54 (m, 4H), 2.33 (d, J = 2.0 Hz, 2H), 2.26 - 2.06 (m, 2H), 1.94 (s, 1H), 1.92 - 1.66 (m, 9H), 1.51 - 1.35 (m, 2H) Example 46. Preparation of Compound 193b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((4S)-1-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino )-2-oxoethyl)-3,3- difluoropiperidin-4-yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((4S)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-3,3-difluoropiperidin-4-y l)propoxy)-2- methylphenyl)picolinate To a solution of (S)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)-3,3- difluoropiperidin-1-yl)acetic acid (100 mg, 123.16 µmol, 1 equiv.) in DMF (2 mL) was added HATU (56.20 mg, 147.79 µmol, 1.2 equiv.), DIEA (47.75 mg, 369.48 µmol, 64.36 µL, 3 equiv.), and 3-(6-amino-1-methyl-indazol-3-yl) piperidine-2, 6-dione (38.17 mg, 147.79 µmol, 1.2 equiv.). The mixture was stirred at 25 °C for 1 hour. The residue was diluted with H 2 O (10 mL) and extracted with DCM (10 mL × 3). The combined organic layers were washed with aqueous NaCl (10 mL × 3), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The compound tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((4S)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)-3,3-difluoropiperidin-4-yl)p ropoxy)-2- methylphenyl)picolinate (120 mg, crude) was obtained as a yellow oil. MS (ESI) m/z: 527.1 [(M+2)/2] + Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((4S)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-3,3-difluoropiperidin-4-y l)propoxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((4S)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1 -methyl-1H-indazol-6-yl)amino)- 2-oxoethyl)-3,3-difluoropiperidin-4-yl)propoxy)-2-methylphen yl)picolinate (120.00 mg, 114.05 µmol, 1 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 118.42 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC. The compound 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3- ((4S)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)amino)-2-oxoethyl)-3,3- difluoropiperidin-4-yl)propoxy)-2-methylphenyl)picolinic acid (23.5 mg, 22.7 µmol, 19.8% yield, 95.8% purity) was obtained as a yellow solid. MS (ESI) m/z: 996.7 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.01 - 12.76 (m, 1H), 12.69 - 12.32 (m, 1H), 10.88 (s, 1H), 10.04 (d, J = 12.0 Hz, 1H), 8.06 - 8.00 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.67 - 7.59 (m, 2H), 7.48 - 7.42 (m, 3H), 7.40 - 7.31 (m, 2H), 7.14 - 7.06 (m, 2H), 6.97 (d, J = 8.8 Hz, 1H), 6.89 (d, J = 8.0 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.32 (dd, J = 5.2, 9.6 Hz, 1H), 3.99 (s, 2H), 3.92 (s, 5H), 3.03 (s, 2H), 2.67 - 2.60 (m, 4H), 2.33 (s, 2H), 2.23 - 2.10 (m, 2H), 2.06 - 1.98 (m, 1H), 1.90 (s, 9H), 1.52 - 1.35 (m, 2H) Example 47. Preparation of Compound 197a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((4S)-1-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino )-2-oxoethyl)-3,3- difluoropiperidin-4-yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl (4E)-4-(3-benzyloxypropylidene)-3, 3- difluoro-piperidine-1-carboxylate A mixture of tert-butyl 3, 3-difluoro-4-oxo-piperidine-1-carboxylate (8 g, 34.01 mmol, 1 equiv.) in THF (100 mL) was added NaHMDS (1 M, 44.21 mL, 1.3 equiv.) at -70 °C under N2 atmosphere. Then the 3-benzyloxypropyl (triphenyl) phosphonium; bromide (25.07 g, 51.01 mmol, 1.5 equiv.) was added the mixture at -70 °C under N 2 atmosphere. The mixture was stirred at 25 °C for 20 hours under N2 atmosphere. The reaction mixture was quenched by addition NH4Cl 100 mL at 0 °C. Then diluted with H2O 100 mL and extracted with ethyl acetate 300 mL (100 mL × 3). The combined organic layers were washed with aqueous NaCl 300 mL (100 mL × 3), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=10/1 to 3/1). The compound tert-butyl (4E)-4-(3-benzyloxypropylidene)-3, 3-difluoro- piperidine-1-carboxylate (4.6 g, 11.9 mmol, 35.0% yield, 95.2% purity) was obtained as a white oil. MS (ESI) m/z: 268.3 [M-100] + Step B. Procedure for preparation of tert-butyl 3,3-difluoro-4-(3- hydroxypropyl)piperidine-1-carboxylate To a solution of tert-butyl (4E)-4-(3-benzyloxypropylidene)-3, 3-difluoro-piperidine-1- carboxylate (3.6 g, 9.80 mmol, 1 equiv.) in MeOH (50 mL) was added Pd/C (2 g, 19.60 mmol, 10% purity, 0.5 equiv.) and Pd(OH)2 (2 g, 2.85 mmol, 20% purity, 0.5 equiv.) under N2 atmosphere. The suspension was degassed and purged with H 2 three times. The mixture was stirred under H2 (15 psi.) at 25 °C for 5 hours. The reaction mixture was filtered and concentrated under reduced pressure to remove solvent. The compound tert-butyl 3, 3-difluoro- 4-(3-hydroxypropyl) piperidine-1-carboxylate (2.1 g, crude) was obtained as a white oil. MS (ESI) m/z: 180.4 [M+H-100] + Step C. Procedure for preparation of tert-butyl 4-(3-(3-bromo-2-methylphenoxy)propyl)- 3,3-difluoropiperidine-1-carboxylate To a solution of 3-bromo-2-methyl-phenol (1.41 g, 7.52 mmol, 1 equiv.) in toluene (50 mL) was added 2-(tributyl-λ 5 -phosphanylidene)acetonitrile (2.72 g, 11.28 mmol, 1.5 equiv.) and tert-butyl 3,3-difluoro-4-(3-hydroxypropyl)piperidine-1-carboxylate (2.1 g, 7.52 mmol, 1 equiv.). The suspension was degassed and purged with N 2 three times. The mixture was stirred under N2 at 120 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=1/0 to 10/1). The compound tert-butyl 4-(3-(3-bromo-2- methylphenoxy)propyl)-3,3-difluoropiperidine-1-carboxylate (2.1 g, 4.4 mmol, 58.5% yield, 94% purity) was obtained as a white solid. Step D. Procedure for preparation of (S)-tert-butyl 4-(3-(3-bromo-2- methylphenoxy)propyl)-3,3-difluoropiperidine-1-carboxylate The tert-butyl 4-(3-(3-bromo-2-methylphenoxy)propyl)-3,3-difluoropiperidine -1-carboxylate residue was purified by prep-HPLC. The compound (S)-tert-butyl 4-(3-(3-bromo-2- methylphenoxy)propyl)-3,3-difluoropiperidine-1-carboxylate (1 g, 2.2 mmol, 47.1% yield, 99% purity) was obtained as a white oil. MS (ESI) m/z: 348.3 [M+H-100] + 1 H NMR (400 MHz, DMSO-d6) δ = 7.18 - 7.13 (m, 1H), 7.09 (t, J = 2.4, 8.0 Hz, 1H), 6.97 (d, J = 8.0 Hz, 1H), 4.13 - 3.95 (m, 4H), 3.90 (d, J = 11.2 Hz, 1H), 3.16 - 2.69 (m, 2H), 2.23 (d, J = 2.4 Hz, 3H), 1.99 (d, J = 3.2 Hz, 1H), 1.85 (d, J = 8.4 Hz, 5H), 1.39 (d, J = 2.4 Hz, 9H) Step E. Procedure for preparation of (S)-4-(3-(3-bromo-2-methylphenoxy)propyl)-3,3- difluoropiperidine To a solution of (S)-tert-butyl 4-(3-(3-bromo-2-methylphenoxy)propyl)-3,3- difluoropiperidine-1-carboxylate (1 g, 2.23 mmol, 1 equiv.) was added HCl/dioxane (4 M, 557.61 µL, 1 equiv.).The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The compound (S)-4-(3-(3-bromo-2- methylphenoxy)propyl)-3,3-difluoropiperidine (700 mg, crude) was obtained as a white solid. Step F. Procedure for preparation of (S)-ethyl 2-(4-(3-(3-bromo-2- methylphenoxy)propyl)-3,3-difluoropiperidin-1-yl)acetate To a solution of (S)-4-(3-(3-bromo-2-methylphenoxy)propyl)-3,3-difluoropiperi dine (700 mg, 2.01 mmol, 1 equiv.) in CH3CN (10 mL) was added K2CO3 (833.46 mg, 6.03 mmol, 3 equiv.) and ethyl 2-bromoacetate (302.13 mg, 1.81 mmol, 200.09 µL, 0.9 equiv.). The mixture was stirred at 60 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate, from 1/0 to 2/1). The compound (S)-ethyl 2-(4-(3-(3-bromo-2- methylphenoxy)propyl)-3,3-difluoropiperidin-1-yl)acetate (600 mg, 1.4 mmol, 68.7% yield) was obtained as a white solid. MS (ESI) m/z: 436.0 [M+H] + Step G. Procedure for preparation of (S)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-ethoxy-2-oxoe thyl)-3,3-difluoropiperidin- 4-yl)propoxy)-2-methylphenyl)picolinate To a solution of (S)-ethyl 2-(4-(3-(3-bromo-2-methylphenoxy)propyl)-3,3-difluoropiperid in-1- yl)acetate (400 mg, 920.99 µmol, 1 equiv.) in dioxane (10 mL) was added [2-(2- aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-butyl-phos phane;methanesulfonate (67.07 mg, 92.10 µmol, 0.1 equiv.), K2CO3 (1.5 M, 3760 µL, 3 equiv.), and tert-butyl 6-[8- (1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin- 2-yl]-3-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)pyridine-2-carboxylate (564.15 mg, 920.99 µmol, 1 equiv.). The mixture was stirred at 100 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate, 10/1 to 2/1). The compound (S)-tert-butyl 6-(8-(benzo[d]thiazol- 2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-( 2-ethoxy-2-oxoethyl)-3,3- difluoropiperidin-4-yl)propoxy)-2-methylphenyl)picolinate (670 mg, 789.6 µmol, 85.7% yield, 99% purity) was obtained as a yellow solid. MS (ESI) m/z: 840.4 [M+H] + Step H. Procedure for preparation of (S)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)propyl)-3,3-difluoropiperidin-1-yl)acetic acid To a solution of (S)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-ethoxy-2-oxoethyl)-3,3-difluoropiper idin-4-yl)propoxy)-2- methylphenyl)picolinate (580 mg, 690.49 µmol, 1 equiv.) in THF (10 mL) and H 2 O (4 mL) was added LiOH•H2O (49.61 mg, 2.07 mmol, 3 equiv.). The mixture was stirred at 25 °C for 1 hour. The pH was adjusted to 6 with 1M HCl. The residue was diluted with H 2 O (10 mL) and extracted with (10 mL × 3). The combined organic layers were washed with aqueous NaCl (10 mL × 3), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The compound (S)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)-3,3- difluoropiperidin-1-yl)acetic acid (430 mg, crude) was obtained as a yellow solid. MS (ESI) m/z: 812.5 [M+H] + Step I. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((4S)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-2-oxoethyl)-3,3-difluoropiperidin-4-y l)propoxy)-2- methylphenyl)picolinate To a solution of (S)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)-3,3- difluoropiperidin-1-yl)acetic acid (100 mg, 123.16 µmol, 1 equiv.) in DMF (2 mL) was added HATU (56.20 mg, 147.80 µmol, 1.2 equiv.), DIEA (47.75 mg, 369.49 µmol, 64.36 µL, 3 equiv.), and 3-(7-amino-1-methyl-indazol-3-yl) piperidine-2, 6-dione (38.17 mg, 147.80 µmol, 1.2 equiv.). The mixture was stirred at 25 °C for 1 hour. The residue was diluted with H2O (10 mL) and extracted with DCM (10 mL × 3). The combined organic layers were washed with aqueous NaCl (10mL × 3), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The compound tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((4S)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl-1H- indazol-7-yl)amino)-2-oxoethyl)-3,3-difluoropiperidin-4-yl)p ropoxy)-2- methylphenyl)picolinate (120 mg, crude) was obtained as a brown oil. MS (ESI) m/z: 527.0 [M/2+H] + Step J. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((4S)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-2-oxoethyl)-3,3-difluoropiperidin-4-y l)propoxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((4S)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1 -methyl-1H-indazol-7-yl)amino)- 2-oxoethyl)-3,3-difluoropiperidin-4-yl)propoxy)-2-methylphen yl)picolinate (120.00 mg, 114.05 µmol, 1 equiv.) in DCM (1.5 mL) was added TFA (2.31 g, 20.26 mmol, 1.5 mL, 177.64 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC. The compound 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3- ((4S)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -7-yl)amino)-2-oxoethyl)-3,3- difluoropiperidin-4-yl)propoxy)-2-methylphenyl)picolinic acid (29.1 mg, 28.9 µmol, 25.3% yield, 99.0% purity) was obtained as a yellow solid. MS (ESI) m/z: 996.7 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 12.90 - 12.80 (m, 1H), 12.71 - 12.47 (m, 1H), 10.90 (s, 1H), 9.89 - 9.81 (m, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 7.6 Hz, 1H), 7.64 - 7.56 (m, 2H), 7.49 - 7.43 (m, 3H), 7.40 - 7.33 (m, 2H), 7.28 (d, J = 7.2 Hz, 1H), 7.09 (q, J = 7.6 Hz, 2H), 6.97 (d, J = 8.8 Hz, 1H), 6.89 (d, J = 8.0 Hz, 1H), 6.63 (d, J = 8.0 Hz, 1H), 4.98 (s, 2H), 4.37 (dd, J = 4.8, 10.0 Hz, 1H), 4.08 (s, 3H), 3.99 (s, 2H), 3.92 (s, 2H), 3.03 (s, 2H), 2.67 (s, 4H), 2.33 (d, J = 2.0 Hz, 2H), 2.23 - 2.08 (m, 2H), 2.06 - 1.96 (m, 1H), 1.96 - 1.75 (m, 9H), 1.53 - 1.36 (m, 2H) Example 48. Preparation of Compound 197b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((4R)-1-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino )-2-oxoethyl)-3,3- difluoropiperidin-4-yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((4R)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-2-oxoethyl)-3,3-difluoropiperidin-4-y l)propoxy)-2- methylphenyl)picolinate To a solution of (R)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)-3,3- difluoropiperidin-1-yl)acetic acid (100.00 mg, 123.16 µmol, 1 equiv.) in DMF (2 mL) was added HATU (56.20 mg, 147.80 µmol, 1.2 equiv.), DIEA (47.75 mg, 369.49 µmol, 64.36 µL, 3 equiv.), and 3-(7-amino-1-methyl-indazol-3-yl) piperidine-2, 6-dione (38.17 mg, 147.80 µmol, 1.2 equiv.). The mixture was stirred at 25 °C for 1 hour. The residue was diluted with H2O (10 mL) and extracted with DCM (10 mL × 3). The combined organic layers were washed with aqueous NaCl (10mL × 3), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The compound tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((4R)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl-1H- indazol-7-yl)amino)-2-oxoethyl)-3,3-difluoropiperidin-4-yl)p ropoxy)-2- methylphenyl)picolinate (120 mg, crude) was obtained as a brown oil. MS (ESI) m/z: 527.2 [(M+2)/2] + Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((4R)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-2-oxoethyl)-3,3-difluoropiperidin-4-y l)propoxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((4R)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1 -methyl-1H-indazol-7-yl)amino)- 2-oxoethyl)-3,3-difluoropiperidin-4-yl)propoxy)-2-methylphen yl)picolinate (120.00 mg, 114.05 µmol, 1 equiv.) in DCM (1.5 mL) was added TFA (2.31 g, 20.26 mmol, 1.5 mL, 177.64 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC. The compound 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3- ((4R)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -7-yl)amino)-2-oxoethyl)-3,3- difluoropiperidin-4-yl)propoxy)-2-methylphenyl)picolinic acid (27.7 mg, 27.2 µmol, 23.8% yield, 97.8% purity) was obtained as a yellow solid. MS (ESI) m/z: 996.7 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 12.87 (d, J = 5.2 Hz, 1H), 12.74 - 12.23 (m, 1H), 10.88 (s, 1H), 10.04 - 9.85 (m, 1H), 8.08 - 8.00 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.66 - 7.59 (m, 2H), 7.50 - 7.42 (m, 3H), 7.40 - 7.32 (m, 2H), 7.14 - 7.06 (m, 2H), 6.97 (d, J = 8.8 Hz, 1H), 6.89 (d, J = 8.4 Hz, 1H), 6.64 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.32 (dd, J = 5.2, 9.6 Hz, 1H), 3.99 (s, 2H), 3.94 - 3.89 (m, 5H), 3.03 (t, J = 5.6 Hz, 2H), 2.74 - 2.54 (m, 4H), 2.33 (d, J = 2.0 Hz, 2H), 2.26 - 2.06 (m, 2H), 1.94 (s, 1H), 1.92 - 1.66 (m, 9H), 1.51 - 1.35 (m, 2H) Example 49. Preparation of Compound 200 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[4-[[4-[3- (2, 6-dioxo-3-piperidyl) -1-methyl-indazol-6-yl]piperazin-1-yl]methyl]-1-piperidyl]pr opoxy]- 2-methyl-phenyl] pyridine-2-carboxylic acid Step A. Procedure for preparation of methyl piperidine-4-carboxylate To a solution 1-(tert-butyl) 4-methyl piperidine-1,4-dicarboxylate (10 g, 41.10 mmol, 1 equiv.) in DCM (10 mL) and HCl/dioxane (4 M, 10 mL, 9.73e-1 equiv.) was stirred at 25 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The compound methyl piperidine-4-carboxylate (7.7 g, crude) was obtained as a white solid. Step B. Procedure for preparation of 1-bromo-3-(3-bromopropoxy) -2-methyl-benzene A mixture of 3-bromo-2-methyl-phenol (2 g, 10.69 mmol, 1 equiv.), 1, 3-dibromopropane (10.79 g, 53.47 mmol, 5.45 mL, 5 equiv.), Cs 2 CO 3 (10.45 g, 32.08 mmol, 3 equiv.), and KI (177.51 mg, 1.07 mmol, 0.1 equiv.) in CH 3 CN (15 mL) was stirred at 25 °C for 5 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~20% ethyl acetate/Petroleum). The compound 1-bromo-3-(3-bromopropoxy) -2-methyl-benzene (2.2 g, 7.1 mmol, 66.8% yield) was obtained as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.19 - 7.15 (m, 1H), 7.10 (t, J = 8.0 Hz, 1H), 6.99 (d, J = 8.0 Hz, 1H), 4.09 (t, J = 5.6 Hz, 2H), 3.69 (t, J = 6.4 Hz, 2H), 2.34 - 2.18 (m, 6H) Step C. Procedure for preparation of methyl 1-[3-(3-bromo-2-methyl-phenoxy) propyl]piperidine-4-carboxylate To a solution of 1-bromo-3-(3-bromopropoxy) -2-methyl-benzene (1.82 g, 5.91 mmol, 1 eq) in CH3CN (20 mL) was added methyl piperidine-4-carboxylate (1.17 g, 6.50 mmol, 1.1 equiv. HCl), K2CO3 (2.45 g, 17.73 mmol, 3 eq) and KI (98.09 mg, 590.89 μmol, 0.1 eq) at 25 °C. The reaction mixture was stirred at 60 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 100% ethyl acetate/petroleum ether). The compound methyl 1-[3-(3-bromo-2-methyl-phenoxy) propyl]piperidine-4-carboxylate (1.9 g, 5.0 mmol, 85.1% yield, 98% purity) was obtained as a colorless oil. MS (ESI) m/z: 370.2 [M+H] + . Step D. Procedure for preparation of [1-[3-(3-bromo-2-methyl-phenoxy) propyl]-4- piperidyl]methanol To a solution of methyl 1-[3-(3-bromo-2-methyl-phenoxy) propyl]piperidine-4-carboxylate (1.9 g, 5.13 mmol, 1 equiv.) in THF (20 mL) was added LiAlH 4 (214.23 mg, 5.64 mmol, 1.1 equiv.) at 0 °C. The reaction mixture was stirred at 0 °C for 1 hour. The reaction mixture was quenched with Na2SO4•10 H2O (20 mg) and slowly warmed to 20 °C. The mixture was filtered and washed with DCM (50 mL). Then the organic layers were concentrated under reduced pressure to give [1-[3-(3-bromo-2-methyl-phenoxy) propyl]-4-piperidyl]methanol (1.55 g, crude) as a colorless oil. MS (ESI) m/z: 342.2 [M+H] + . Step E. Procedure for preparation of 1-[3-(3-bromo-2-methyl-phenoxy) propyl]piperidine-4-carbaldehyde A solution of DMSO (1.23 g, 15.78 mmol, 1.23 mL, 4 equiv.) in DCM (10 mL) was added dropwise to a solution of oxalyl dichloride (1.00 g, 7.89 mmol, 690.54 μL, 2 equiv.) in DCM (1 mL) at -70 °C under N2 atmosphere. The mixture was stirred at -70 °C for 30 minutes. After which time [1-[3-(3-bromo-2-methyl-phenoxy) propyl]-4-piperidyl]methanol (1.35 g, 3.94 mmol, 1 equiv.) in DCM (1 mL) was added dropwise at -70 °C. The solution was stirred for 30 minutes at -70 °C. Then TEA (2.39 g, 23.67 mmol, 3.29 mL, 6 equiv.) was added into the solution. The solution was stirred at 25 °C for 1 hour under N2 atmosphere. The reaction mixture was diluted with water (10 mL) and extracted with DCM (10 mL × 3). The combined organic layers were washed with brine (10 mL × 1), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 20/1 to 10/1). The compound 1-[3-(3- bromo-2-methyl-phenoxy) propyl]piperidine-4-carbaldehyde (1.4 g, crude) was obtained as a yellow oil. 1 H NMR (400 MHz, CDCl3-d) δ = 9.68 - 9.63 (m, 1H), 7.13 (d, J = 8.0 Hz, 1H), 6.98 (t, J = 8.0 Hz, 1H), 6.76 (d, J = 8.0 Hz, 1H), 4.01 - 3.97 (m, 2H), 2.91 - 2.81 (m, 2H), 2.56 - 2.51 (m, 2H), 2.30 (s, 3H), 2.28 - 2.22 (m, 1H), 2.17 - 2.09 (m, 2H), 2.03 - 1.95 (m, 2H), 1.94 - 1.87 (m, 2H), 1.76 - 1.64 (m, 2H) Step F. Procedure for preparation of 3-[6-[4-[[1-[3-(3-bromo-2-methyl-phenoxy) propyl]- 4-piperidyl]methyl]piperazin-1-yl]-1-methyl-indazol-3-yl]pip eridine-2, 6-dione To a solution of 1-[3-(3-bromo-2-methyl-phenoxy) propyl]piperidine-4-carbaldehyde (200 mg, 587.80 μmol, 1 equiv.) in DCM (2 mL) was added 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl) piperidine-2, 6-dione (106.93 mg, 293.90 μmol, 0.5 equiv., HCl) and AcOH (3.53 mg, 58.78 μmol, 0.1 equiv.) at 25 °C. The mixture was stirred at 25 °C for 18 hours. NaBH(OAc)3 (373.73 mg, 1.76 mmol, 3 equiv.) was added to the mixture at 25 °C. The reaction mixture was stirred at 25 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH = 20:1). The compound 3-[6-[4-[[1-[3-(3-bromo-2-methyl-phenoxy) propyl]-4-piperidyl]methyl] piperazin-1-yl]-1-methyl-indazol-3-yl]piperidine-2, 6-dione (100 mg, 135.0 μmol, 22.9% yield, 88% purity) was obtained as a yellow oil. MS (ESI) m/z: 653.0 [M+H] + . Step G. Procedure for preparation of tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) - 3, 4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[4-[[4-[3-(2, 6-dioxo-3-piperidyl) -1-methyl- indazol-6-yl]piperazin-1-yl] methyl]-1-piperidyl]propoxy]-2-methyl-phenyl]pyridine-2- carboxylate 3-[6-[4-[[1-[3-(3-bromo-2-methyl-phenoxy) propyl]-4-piperidyl]methyl]piperazin-1-yl]-1- methyl-indazol-3-yl]piperidine-2, 6-dione (100 mg, 153.46 μmol, 1 equiv.), tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-3-(4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridine-2-carboxylate (122.20 mg, 199.50 μmol, 1.3 equiv.), KF (26.75 mg, 460.38 μmol, 10.79 μL, 3 equiv.) and [2-(2-aminophenyl) phenyl]palladium (1+);bis (1-adamantyl) -butyl-phosphane;methanesulfonate (11.18 mg, 15.35 μmol, 0.1 equiv.) were taken up into a microwave tube in dioxane (1 mL) and H 2 O (0.1 mL). The sealed tube was heated at 100 °C for 1 hour under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , DCM : MeOH = 10:1). The compound tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[4-[[4-[3-(2, 6-dioxo-3-piperidyl) -1-methyl- indazol-6-yl]piperazin-1-yl] methyl]-1-piperidyl]propoxy]-2-methyl-phenyl]pyridine-2- carboxylate (40 mg, 34.5 μmol, 22.5% yield, 91.2% purity) was obtained as a yellow oil. MS (ESI) m/z: 1057.4 [M+H] + . Step H. Procedure for preparation of 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4- dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[4-[[4-[3-(2, 6-dioxo-3-piperidyl) -1-methyl-indazol- 6-yl]piperazin-1-yl]methyl]-1-piperidyl]propoxy]-2-methyl-ph enyl] pyridine-2- carboxylic acid A solution of tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin- 2-yl]-3-[3-[3-[4-[[4-[3-(2, 6-dioxo-3-piperidyl) -1-methyl-indazol-6-yl]piperazin-1- yl]methyl]-1-piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-c arboxylate (40 mg, 37.83 μmol, 1 equiv.) in DCM (0.5 mL) and TFA (0.5 mL) was stirred at 25 °C for 18 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC. The compound 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4- dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[4-[[4-[3-(2, 6-dioxo-3-piperidyl) -1-methyl-indazol-6- yl]piperazin-1-yl]methyl]-1-piperidyl]propoxy]-2-methyl-phen yl] pyridine-2-carboxylic acid (21.1 mg, 20.9 μmol, 55.4% yield, 99.3% purity) was obtained as a pink solid. MS (ESI) m/z: 1001.4 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.85 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 8.0 Hz, 1H), 7.52 - 7.31 (m, 6H), 7.14 - 7.06 (m, 1H), 6.96 (d, J = 8.4 Hz, 1H), 6.93 - 6.86 (m, 2H), 6.83 (s, 1H), 6.65 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.25 (dd, J = 4.8, 8.8 Hz, 1H), 4.04 - 3.99 (m, 2H), 3.93 - 3.86 (m, 5H), 3.22 - 3.20 (m, 4H), 3.04 - 2.99 (m, 4H), 2.90 - 2.81 (m, 4H), 2.67 (s, 1H), 2.63 - 2.58 (m, 2H), 2.33 - 2.28 (m, 2H), 2.23 - 2.19 (m, 2H), 2.18 - 2.11 (m, 2H), 2.06 - 1.97 (m, 3H), 1.91 (s, 3H), 1.80 (d, J = 12.8 Hz, 2H), 1.69 (d, J = 3.2 Hz, 1H), 1.29 - 1.17 (m, 2H) Example 50. Preparation of compound 202a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[(3S)-1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-o xo-ethyl]-3-piperidyl]butoxy]- 2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl (3S)-3-formylpiperidine-1-carboxylate A mixture of tert-butyl (3S)-3-(hydroxymethyl)piperidine-1-carboxylate (4 g, 18.58 mmol, 1 equiv.) and DMP (8.67 g, 20.44 mmol, 6.33 mL, 1.1 equiv.) in DCM (50 mL) was stirred at 20 °C for 1 hour under N2 atmosphere. The mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=50/1 to 3/1) to give tert-butyl (3S)-3-formylpiperidine-1- carboxylate (3 g, 14.0 mmol, 75.7% yield) as a colorless oil. 1 H NMR 1 H NMR (400 MHz, DMSO-d 6 ) δ = 9.59 (s, 1H), 3.72 (d, J = 8.0 Hz, 1H), 3.43 (s, 1H), 3.14 - 3.01 (m, 1H), 2.49 - 2.41 (m, 1H), 1.89 - 1.80 (m, 1H), 1.65 - 1.48 (m, 2H), 1.39 (s, 11H) Step B. Procedure for preparation of tert-butyl (3R)-3-[(E)-4-benzyloxybut-1- enyl]piperidine-1-carboxylate A mixture of 3-benzyloxypropyl(triphenyl)phosphonium;bromide (3.46 g, 7.03 mmol, 1 equiv.) in THF (5 mL) was degassed and purged with N 2 three times and cooled to -70 °C. LiHMDS (1 M, 10.55 mL, 1.5 equiv.) was added dropwise at -70 °C. After stirring at -70 °C for 1 hour, tert-butyl (3S)-3-formylpiperidine-1-carboxylate (1.5 g, 7.03 mmol, 1 equiv.) in THF (3 mL) was added dropwise at -70 °C. The mixture was stirred at -70 °C for 1 hour and was quenched by addition of saturated NH4Cl solution 20 mL dropwise at 0°C. The resulting mixture was extracted with EtOAc (20 mL × 3). The combined organic layers were washed with brine (10 mL × 3), dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate 200/1 to 10/1) to give tert-butyl (3R)-3-[(E)-4-benzyloxybut-1- enyl]piperidine-1-carboxylate (1.8 g, 4.6 mmol, 65.9% yield, 89% purity) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.39 - 7.18 (m, 5H), 5.48 - 5.36 (m, 1H), 5.28 - 5.18 (m, 1H), 4.46 (s, 2H), 3.84 - 3.64 (m, 2H), 3.44 (t, J = 6.8 Hz, 2H), 2.72 (s, 1H), 2.63 - 2.51 (m, 1H), 2.41 - 2.25 (m, 3H), 1.66 (d, J = 12.4 Hz, 1H), 1.60 - 1.51 (m, 1H), 1.38 (s, 10H), 1.28 - 1.17 (m, 1H) Step C. Procedure for preparation of tert-butyl (3S)-3-(4-hydroxybutyl)piperidine-1- carboxylate A mixture of tert-butyl (3R)-3-[(E)-4-benzyloxybut-1-enyl]piperidine-1-carboxylate (1.8 g, 5.21 mmol, 1 equiv.), Pd/C (0.1 g, 521.02 µmol, 10% purity, 0.1 equiv.) and Pd(OH) 2 (0.1 g, 71.21 µmol, 10% purity, 0.01.37 equiv.) in MeOH (200 mL) was degassed and purged with H 2 (15 Psi) three times, and then the mixture was stirred at 20 °C for 16 hours under H2 (15 Psi) atmosphere. The mixture was filtered and concentrated under reduced pressure to give crude tert-butyl (3S)-3-(4-hydroxybutyl)piperidine-1-carboxylate (1.3 g, 4.0 mmol, 77.5% yield, 80% purity) as a gray oil. 1 H NMR (400 MHz, DMSO-d6) δ = 3.72 (s, 2H), 3.38 (q, J = 6.0 Hz, 1H), 2.84 - 2.70 (m, 1H), 2.50 (s, 1H), 1.74 (d, J = 10.4 Hz, 1H), 1.62 - 1.50 (m, 1H), 1.38 (s, 11H), 1.34 - 1.11 (m, 7H), 1.08 (d, J = 7.6 Hz, 1H), 0.87 (t, J = 6.4 Hz, 1H) Step D. Procedure for preparation of tert-butyl (3S)-3-[4-(3-bromo-2-methyl- phenoxy)butyl]piperidine-1-carboxylate A mixture of 3-bromo-2-methyl-phenol (399.69 mg, 2.14 mmol, 1.1 equiv.), tert-butyl (3S)-3- (4-hydroxybutyl)piperidine-1-carboxylate (0.5 g, 1.94 mmol, 1 equiv.) in toluene (5 mL) was degassed and purged with N 2 three times, and then 2-(tributyl-λ 5 -phosphanylidene)acetonitrile (515.78 mg, 2.14 mmol, 2.14 mL, 1.1 equiv.) was added dropwise. The mixture was stirred at 110 °C for 16 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~25% ethyl acetate/petroleum ether) to give tert-butyl (3S)-3-[4-(3-bromo-2- methyl-phenoxy)butyl]piperidine-1-carboxylate (0.5 g, 1.1 mmol, 55.5% yield, 92% purity) as a colorless oil. MS (ESI) m/z: 326.2 [M-100+H] + Step E. Procedure for preparation of (3S)-3-[4-(3-bromo-2-methyl- phenoxy)butyl]piperidine To a solution of tert-butyl (3S)-3-[4-(3-bromo-2-methyl-phenoxy)butyl]piperidine-1- carboxylate (0.5 g, 1.17 mmol, 1 equiv.) in HCl/dioxane (5 mL) was stirred at 20 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product was triturated with petroleum ether/ethyl acetate=5:1 (2 mL) at 20°C for 30 min to give (3S)-3-[4-(3-bromo-2-methyl-phenoxy)butyl]piperidine (0.3 g, 919.4 µmol, 78.4% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 8.81 - 8.58 (m, 1H), 8.52 - 8.30 (m, 1H), 7.19 - 7.14 (m, 1H), 7.13 - 7.07 (m, 1H), 6.97 (d, J = 7.6 Hz, 1H), 3.99 (br t, J = 6.4 Hz, 2H), 2.83 - 2.68 (m, 2H), 2.24 (s, 3H), 1.82 - 1.71 (m, 4H), 1.63 - 1.55 (m, 1H), 1.50 - 1.41 (m, 2H), 1.35 - 1.22 (m, 5H), 1.18 - 1.05 (m, 1H) Step F. Procedure for preparation of ethyl 2-[(3S)-3-[4-(3-bromo-2-methyl- phenoxy)butyl]-1-piperidyl]acetate To a solution of ethyl 2-bromoacetate (91.16 mg, 545.86 µmol, 60.37 µL, 0.9 equiv.) and (3S)- 3-[4-(3-bromo-2-methyl-phenoxy)butyl]piperidine (220 mg, 606.51 µmol, 1 equiv., HCl) in CH 3 CN (2 mL) was added K 2 CO 3 (251.47 mg, 1.82 mmol, 3 equiv.). The mixture was stirred at 60 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~25% ethyl acetate/petroleum ether) to give ethyl 2-[(3S)-3-[4-(3-bromo-2-methyl-phenoxy)butyl]- 1-piperidyl]acetate (0.18 g, 436.5 µmol, 71.9% yield) as a light yellow oil 1 H NMR (400 MHz, CDCl 3 ) δ = 7.06 (d, J = 8.0 Hz, 1H), 6.91 (t, J = 8.0 Hz, 1H), 6.68 (d, J = 8.0 Hz, 1H), 4.11 (q, J = 7.2 Hz, 2H), 3.86 (t, J = 6.4 Hz, 2H), 3.12 (s, 2H), 2.85 - 2.77 (m, 2H), 2.23 (s, 3H), 2.08 - 1.99 (m, 1H), 1.73 - 1.67 (m, 3H), 1.63 - 1.55 (m, 3H), 1.48 - 1.36 (m, 2H), 1.23 - 1.18 (m, 5H), 0.84 - 0.75 (m, 2H) Step G. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[(3S)-1-(2-ethoxy-2 -oxo-ethyl)-3- piperidyl]butoxy]-2-methyl-phenyl]pyridine-2-carboxylate A mixture of ethyl 2-[(3S)-3-[4-(3-bromo-2-methyl-phenoxy)butyl]-1-piperidyl]ac etate (0.18 g, 436.51 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (267.38 mg, 436.51 µmol, 1 equiv.), K3PO4 (1.5 M, 873.02 µL, 3 equiv.), and [2-(2- aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-butyl-phos phane;methanesulfonate (31.79 mg, 43.65 µmol, 0.1 equiv.) in dioxane (3 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 80 °C for 1 hour under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~30% ethyl acetate/petroleum ether) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4- [(3S)-1-(2-ethoxy-2-oxo-ethyl)-3-piperidyl]butoxy]-2-methyl- phenyl]pyridine-2-carboxylate (0.24 g, 228.8 µmol, 52.4% yield, 78% purity) as a yellow oil. MS (ESI) m/z: 381.7 [(M-56)/2+H] + Step H. Procedure for preparation of 2-[(3S)-3-[4-[3-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-2- methyl-phenoxy]butyl]-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[4-[(3S)-1-(2-ethoxy-2-oxo-ethyl)-3-piperidyl]but oxy]-2-methyl-phenyl]pyridine- 2-carboxylate (0.24 g, 228.84 µmol, 78% purity, 1 equiv.) in THF (1 mL) was added LiOH•H2O (1.5 M, 762.81 µL, 5 equiv.).The mixture was stirred at 20 °C for 1 hour. The mixture was concentrated under reduced pressure, adjusted to pH<7, and filtered to give crude 2-[(3S)-3-[4-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H-isoquinolin-2-yl]-2- tert-butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]butyl]-1-pip eridyl]acetic acid (0.16 g, 202.5 µmol, 88.5% yield) as a brown solid 1 H NMR (400 MHz, DMSO-d6) δ = 13.32 - 12.25 (m, 1H), 8.01 (d, J = 7.6 Hz, 1H), 7.77 (d, J = 8.0 Hz, 1H), 7.60 (d, J = 7.6 Hz, 1H), 7.49 - 7.40 (m, 3H), 7.40 - 7.31 (m, 2H), 7.14 - 7.06 (m, 1H), 6.93 (dd, J = 8.4, 12.4 Hz, 2H), 6.57 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 3.97 (t, J = 6.0 Hz, 2H), 3.87 (t, J = 6.0 Hz, 2H), 3.18 (s, 2H), 3.15 (d, J = 2.0 Hz, 2H), 3.03 (t, J = 6.0 Hz, 2H), 2.24 (t, J = 11.2 Hz, 1H), 1.87 (s, 3H), 1.76 - 1.56 (m, 7H), 1.50 - 1.37 (m, 2H), 1.29 - 1.18 (m, 3H), 1.00 (s, 9H), 0.95 - 0.88 (m, 1H) Step I. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[(3S)-1-[2-[[3-(2,6-dio xo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-3-piperidyl]butoxy]-2-methy l-phenyl]pyridine-2- carboxylate To a solution of 2-[(3S)-3-[4-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]butyl]-1- piperidyl]acetic acid (0.05 g, 63.29 µmol, 1 equiv.) in DMF (0.5 mL) was added HATU (28.88 mg, 75.95 µmol, 1.2 equiv.), TEA (19.21 mg, 189.88 µmol, 26.43 µL, 3 equiv.), and 3-(6- amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (16.35 mg, 63.29 µmol, 1 equiv.) .The mixture was stirred at 40 °C for 0.5 hour. The solution was poured into water (2 mL) and then filtered to give a crude tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[4-[(3S)-1-[2-[[3-(2,6-dioxo-3-piperi dyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-3-piperidyl]butoxy]-2-methyl-phenyl]p yridine-2-carboxylate (60 mg, 58.4 µmol, 92.0% yield) as a white solid. MS (ESI) m/z: 516.1 [M/2+H] + Step J. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[4-[(3S)-1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-3-piperidyl]butoxy]-2-methyl-phenyl]p yridine-2-carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[4-[(3S)-1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methy l-indazol-6-yl]amino]-2-oxo- ethyl]-3-piperidyl]butoxy]-2-methyl-phenyl]pyridine-2-carbox ylate (0.06 g, 58.24 µmol, 1 equiv.) in TFA (0.3 mL) was added DCM (0.5 mL) .The mixture was stirred at 20 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[(3S)-1-[2-[[3-(2,6-dio xo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-3-piperidyl]butoxy]-2-methy l-phenyl]pyridine-2- carboxylic acid (21.5 mg, 21.4 µmol, 36.8% yield, 96.9% purity) as a yellow solid MS (ESI) m/z: 975.0 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 13.09 - 12.58 (m, 1H), 10.88 (s, 1H), 10.03 - 9.69 (m, 1H), 8.07 - 8.00 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.67 - 7.57 (m, 2H), 7.50 - 7.32 (m, 5H), 7.23 - 7.17 (m, 1H), 7.07 (t, J = 8.0 Hz, 1H), 6.99 - 6.93 (m, 1H), 6.85 (d, J = 8.4 Hz, 1H), 6.62 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.32 (dd, J = 5.2, 9.6 Hz, 1H), 4.00 - 3.85 (m, 7H), 3.19 - 3.12 (m, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.90 - 2.77 (m, 2H), 2.67 - 2.59 (m, 2H), 2.38 - 2.30 (m, 1H), 2.21 - 2.07 (m, 2H), 1.95 - 1.83 (m, 4H), 1.78 - 1.68 (m, 3H), 1.67 - 1.56 (m, 3H), 1.52 - 1.42 (m, 2H), 1.32 - 1.20 (m, 2H), 0.96 - 0.82 (m, 1H) Example 51. Preparation of Compound 205 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[8-[4-[3-(2,6- dioxo-3-piperidyl)-1-methyl-indazol-6-yl]piperazin-1-yl]octo xy]-2-methyl-phenyl]pyridine- 2-carboxylic acid Step A. Procedure for preparation of ethyl 8-(3-bromo-2-methyl-phenoxy)octanoate A mixture of 3-bromo-2-methyl-phenol (2 g, 10.69 mmol, 1 equiv.), ethyl 8-bromooctanoate (3.22 g, 12.83 mmol, 1.2 equiv.), K2CO3 (2.96 g, 21.39 mmol, 2 equiv.) in CH3CN (20 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 60 °C for 2 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 8% ethyl acetate / petroleum ether) to give ethyl 8-(3-bromo-2-methyl-phenoxy)octanoate (3.5 g, 9.8 mmol, 91.6% yield) as a colorless. 1 H NMR (400 MHz, DMSO-d6) δ = 7.13 - 7.06 (m, 2H), 6.95 - 6.93 (m, 1H), 4.06 - 4.01 (m, 2H), 3.96 (s, 2H), 2.23 (s, 3H), 1.71 (s, 2H), 1.52 (s, 2H), 1.41 (s, 2H), 1.37 - 1.21 (m, 6H), 1.18 - 1.15 (m, 3H). Step B. Procedure for preparation of 8-(3-bromo-2-methyl-phenoxy) octan-1-ol To a solution of ethyl 8-(3-bromo-2-methyl-phenoxy)octanoate (3.5 g, 9.80 mmol, 1 equiv.) in THF (30 mL) was added LiAlH 4 (557.71 mg, 14.69 mmol, 1.5 equiv.). The mixture was stirred at 0 °C for 2 hours. The reaction mixture was quenched by addition H2O (0.64 mL), 15% NaOH (0.64 mL), and then extracted with ethyl acetate 30mL (10 mL × 3). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give The compound 8-(3-bromo-2-methyl-phenoxy) octan-1-ol (2.8 g, crude) as a colorless oil. Step C. Procedure for preparation of 8-(3-bromo-2-methyl-phenoxy)octanal DMSO (2.78 g, 35.53 mmol, 2.78 mL, 4 equiv.) in DCM (60 mL) was added a solution of (COCl)2 (2.25 g, 17.76 mmol, 1.55 mL, 2 equiv.) in DCM (60 mL) at -70 °C under N2 atmosphere. The mixture was stirred at -70 °C for 1 hour. After which time 8-(3-bromo-2- methyl-phenoxy) octan-1-ol (2.8 g, 8.88 mmol, 1 equiv.) in DCM (60 mL) was added dropwise at -70 °C. The solution was stirred for 1 hour at -70 °C. Then TEA (5.39 g, 53.29 mmol, 7.42 mL, 6 equiv.) was added into the solution. The solution was stirred at -70 °C for 1.5 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 30% ethyl acetate / petroleum ether) to give 8-(3-bromo-2-methyl-phenoxy)octanal (2.3 g, 7.3 mmol, 82.6% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 9.66 (d, J = 1.6 Hz, 1H), 7.16 - 7.07 (m, 2H), 6.96 (d, J = 8.0 Hz, 1H), 3.99 - 3.94 (m, 2H), 2.51 (d, J = 1.6 Hz, 2H), 2.23 (s, 3H), 1.46 - 1.16 (m, 12H). Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[2-methyl-3-(8-oxooctoxy) phenyl]pyridine-2- carboxylate A mixture of 8-(3-bromo-2-methyl-phenoxy)octanal (200 mg, 638.51 µmol, 1 equiv.), tert- butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-2-carboxylate (391.12 mg, 638.51 µmol, 1 equiv.), Ad2nBuP Pd G3(cataCXium® A Pd G3) (93.00 mg, 127.70 µmol, 0.2 equiv.), K 2 CO 3 (1.5 M, 638.51 µL, 1.5 equiv.) in 1,4-dioxane (6 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 100 °C for 1 hour under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[2-methyl-3-(8-oxooctoxy)phenyl]pyridine -2-carboxylate (65 mg, 90.4 µmol, 14.1% yield) as a white solid. MS (ESI) m/z: 719.8 [M+H] + Step E. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[8-[4-[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol- 6-yl]piperazin-1-yl]octoxy]-2-methyl-phenyl]pyridine-2-carbo xylate A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[2-methyl-3-(8-oxooctoxy)phenyl]pyridine-2-carboxylate (65 mg, 90.42 µmol, 1 equiv.), 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (35.52 mg, 108.50 µmol, 1.2 equiv.), NaBH(OAc)3 (57.49 mg, 271.25 µmol, 3 equiv.), 4A MS (10 mg, 1.00 equiv.) and AcOH (5.43 mg, 90.42 µmol, 5.18 µL, 1 equiv.) in DCM (2 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 25 °C for 4 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[8-[4-[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol-6-yl]piperazin-1-yl]octoxy]-2-me thyl-phenyl]pyridine-2- carboxylate (80 mg, crude) as a white solid. MS (ESI) m/z: 1030.9 [M+H] + Step F. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[8-[4-[3-(2,6-dioxo-3-piperidyl)-1 -methyl-indazol-6- yl]piperazin-1-yl]octoxy]-2-methyl-phenyl]pyridine-2-carboxy lic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[8-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol -6-yl]piperazin-1-yl]octoxy]-2- methyl-phenyl]pyridine-2-carboxylate (80 mg, 77.65 µmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL). The mixture was stirred at 40 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8- (1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin- 2-yl]-3-[3-[8-[4-[3-(2,6-dioxo- 3-piperidyl)-1-methyl-indazol-6-yl]piperazin-1-yl]octoxy]-2- methyl-phenyl]pyridine-2- carboxylic acid (27.8 mg, 27.3 µmol, 35.2% yield, 95.8% purity) as a white solid. MS (ESI) m/z: 974.5 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 10.85 (s, 1H), 8.14 (s, 1H), 8.04 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 6.8 Hz, 1H), 7.50 - 7.48 (m, 2H), 7.44 - 7.41 (m, 2H), 7.36 (d, J = 8.4 Hz, 2H), 7.11 - 7.07 (m, 1H), 6.95 - 6.83 (m, 5H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 - 4.96 (m, 2H), 4.27 - 4.24 (m, 1H), 3.98 - 3.95 (m, 2H), 3.90 - 3.88 (m, 5H), 3.24 - 3.21 (m, 6H), 3.01 - 2.98 (m, 2H), 2.62 - 2.59 (m, 6H), 2.31 - 2.26 (m, 1H), 2.19 - 2.13 (m, 1H), 1.90 (s, 3H), 1.77 - 1.73 (m, 2H), 1.48 - 1.44 (m, 4H), 1.36 – 1.29 (m, 6H). Example 52. Preparation of Compound 207a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((3S)-1-(2- ((3-(2,6-dioxo piperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2-oxoethyl)p iperidin-3- yl)propoxy)-2-methylphenyl) picolinic acid Step A. Procedure for preparation of (R)-tert-butyl 3-formylpiperidine-1-carboxylate To a solution of tert-butyl (3R) -3-(hydroxymethyl) piperidine-1-carboxylate (10 g, 46.45 mmol, 1 equiv.) in DCM (10 mL) was added DMP (19.70 g, 46.45 mmol, 14.38 mL, 1 equiv.) at 0 °C. The reaction mixture was degassed and purged with N2 three times, and then the mixture was stirred at 0 °C for 2 hours under N 2 atmosphere. The reaction mixture was diluted with Na2S2O3 (10 mL) and extracted with DCM (20 mL × 3). The combined organic layers were washed with brine (10 mL× 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give (R)-tert-butyl 3-formylpiperidine-1-carboxylate (9.29 g, crude) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 9.59 (s, 1H), 3.82 - 3.65 (m, 1H), 3.55 - 3.35 (m, 2H), 3.12 - 3.02 (m, 1H), 2.49 - 2.41 (m, 1H), 1.89 - 1.81 (m, 1H), 1.64 - 1.51 (m, 2H), 1.39 (s, 10H) Step B. Procedure for preparation of (S,E)-tert-butyl 3-(3-ethoxy-3-oxoprop-1-en-1- yl)piperidine-1-carboxylate To a solution of ethyl 2-(diethoxyphosphoryl)acetate (19.53 g, 87.12 mmol, 17.28 mL, 2 equiv.) in THF (100 mL) was added NaH (2.30 g, 57.50 mmol, 60% purity, 1.32 equiv.) in portions under N2 at 0 °C. The mixture was stirred at 0 °C for 1 hour. (R)-tert-butyl 3- formylpiperidine-1-carboxylate (9.29 g, 43.56 mmol, 1 equiv.) was added to the mixture, and then the mixture was stirred at 25 °C for 16 hours. The reaction mixture was quenched by addition NH4Cl (50 mL) at 0 °C, and extracted with ethyl acetate (150 mL × 3). The combined organic layers were washed with brine 100 mL (100 mL × 1), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate 1/0 to 20/1) to give (S,E)-tert-butyl 3-(3- ethoxy-3-oxoprop-1-en-1-yl)piperidine-1-carboxylate (6.7 g, 22.1 mmol, 50.8% yield, 93.7% purity) as a colorless oil. 1 H NMR (400 MHz, CDCl3) δ = 6.85 (dd, J = 6.8, 16.0 Hz, 1H), 5.87 (d, J = 16.0 Hz, 1H), 4.20 (q, J = 7.2 Hz, 2H), 3.95 - 3.86 (m, 1H), 2.96 - 2.61 (m, 2H), 2.46 - 2.28 (m, 1H), 1.93 - 1.85 (m, 1H), 1.74 - 1.57 (m, 2H), 1.56 - 1.49 (m, 1H), 1.47 (s, 9H), 1.44 - 1.34 (m, 1H), 1.30 (t, J = 7.2 Hz, 3H) Step C. Procedure for preparation of (S)-tert-butyl 3-(3-ethoxy-3-oxopropyl)piperidine- 1-carboxylate To a solution of (S,E)-tert-butyl 3-(3-ethoxy-3-oxoprop-1-en-1-yl)piperidine-1-carboxylate (2 g, 7.06 mmol, 1 equiv.) in EtOH (20 mL) was added PtO2 (160.27 mg, 705.81 μmol, 0.1 equiv.) under N2. The suspension was degassed under vacuum and purged with H2 three times. The mixture was stirred under H 2 (15 psi) at 25 °C for 24 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 10% Ethylacetate / petroleum ether) to give (S)-tert- butyl 3-(3-ethoxy-3-oxopropyl)piperidine-1-carboxylate (2.8 g, crude) as a colorless oil. 1 H NMR (400 MHz, CDCl3) δ = 4.18 - 4.09 (m, 2H), 3.96 - 3.88 (m, 1H), 2.85 - 2.74 (m, 1H), 2.51 (t, J = 11.6 Hz, 1H), 2.34 (t, J = 7.6 Hz, 2H), 1.87 - 1.78 (m, 1H), 1.67 - 1.59 (m, 2H), 1.59 - 1.49 (m, 2H), 1.46 (s, 9H), 1.45 - 1.38 (m, 1H), 1.26 (t, J = 7.1 Hz, 3H), 1.15 - 1.04 (m, 1H). SFC: Column: Chiralpak IC-350 × 4.6 mm I.D, 3 um, Mobile phase: Phase A for CO2, and Phase B for MeOH (0.05% DEA); Gradient elution: B in A from 5% to 40% Flow rate: 3 mL/min; Detector: PDA; Column Temp: 35 C; Back Pressure: 100 Bar Step D. Procedure for preparation of (S)-tert-butyl 3-(3-hydroxypropyl)piperidine-1- carboxylate To a solution of (S)-tert-butyl 3-(3-ethoxy-3-oxopropyl)piperidine-1-carboxylate (2.8 g, 9.81 mmol, 1 equiv.) in THF (30 mL) was added LiAlH4 (372.39 mg, 9.81 mmol, 1 equiv.) dropwise at 0 °C under N 2 . The reaction mixture was stirred under N 2 at 0 °C for 2 hours. The reaction mixture was quenched with Na 2 SO 4 •10 H 2 O (100 mg) and slowly warmed to 20 °C. The mixture was filtered and the filter cake was washed with DCM (50 mL). Then the organic layers were concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 10% Ethylacetate / petroleum ether) to give (S)- tert-butyl 3-(3-hydroxypropyl)piperidine-1-carboxylate (2.2 g, 7.5 mmol, 76.8% yield, 83.4% purity) as a colorless oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 4.51 - 4.23 (m, 1H), 3.86 - 3.63 (m, 2H), 3.46 - 3.34 (m, 3H), 2.80 - 2.71 (m, 1H), 1.78 - 1.70 (m, 1H), 1.60 - 1.52 (m, 1H), 1.46 - 1.40 (m, 2H), 1.38 (s, 9H), 1.35 - 1.24 (m, 2H), 1.22 - 1.12 (m, 2H), 1.08 - 0.99 (m, 1H) Step E. Procedure for preparation of (S)-tert-butyl 3-(3-(3-bromo-2- methylphenoxy)propyl)piperidine-1-carboxylate To a solution of (S)-tert-butyl 3-(3-hydroxypropyl)piperidine-1-carboxylate (2.2 g, 9.04 mmol, 1 equiv.) in toluene (25 mL) was added 3-bromo-2-methylphenol (1.69 g, 9.04 mmol, 1 equiv.) and 2-(tributyl-λ 5 -phosphanylidene) acetonitrile (2.62 g, 10.85 mmol, 1.2 equiv.) at 25 °C. The reaction mixture was degassed and purged with N2 three times, and then the mixture was stirred at 120 °C for 16 hours under N 2 atmosphere. The mixture was filtered, and the filter cake was washed with DCM (50 mL). Then the organic layers were concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate 1/0 to 20/1) to give (S)-tert-butyl 3-(3-(3-bromo-2- methylphenoxy)propyl)piperidine-1-carboxylate (3.1 g, 6.1 mmol, 68.0% yield, 80% purity) as a colorless oil. 1 H NMR (400 MHz, CDCl3) δ = 7.14 (d, J = 8.0 Hz, 1H), 6.99 (t, J = 8.0 Hz, 1H), 6.75 (d, J = 8.0 Hz, 1H), 4.03 - 3.89 (m, 4H), 2.83 - 2.75 (m, 1H), 2.55 - 2.45 (m, 1H), 2.31 (s, 3H), 1.92 - 1.80 (m, 3H), 1.70 - 1.62 (m, 1H), 1.55 - 1.48 (m, 1H), 1.47 (s, 9H), 1.45 - 1.32 (m, 3H), 1.17 - 1.05 (m, 1H) Step F. Procedure for preparation of (S)-3-(3-(3-bromo-2- methylphenoxy)propyl)piperidine To a solution of (S)-tert-butyl 3-(3-(3-bromo-2-methylphenoxy)propyl)piperidine-1- carboxylate (3.17 g, 6.15 mmol, 80% purity, 1 equiv.) in DCM (10 mL) was added HCl/dioxane (4 M, 15 mL, 9.76 equiv.) at 25 °C. The reaction mixture was stirred at 25 °C for 1 hour. The reaction mixture was filtered to give (S)-3-(3-(3-bromo-2-methylphenoxy)propyl)piperidine (2.24 g, crude) as a yellow solid. 1 H NMR (400 MHz, CD3OD) δ = 7.14 - 7.10 (m, 1H), 7.03 (t, J = 8.0 Hz, 1H), 6.89 (d, J = 8.0 Hz, 1H), 4.00 (t, J = 6.4 Hz, 2H), 3.41 - 3.32 (m, 2H), 2.95 - 2.86 (m, 1H), 2.66 (t, J = 12.0 Hz, 1H), 2.29 (s, 3H), 2.04 - 1.67 (m, 6H), 1.57 - 1.46 (m, 2H), 1.33 - 1.19 (m, 1H) Step G. Procedure for preparation of (S)-ethyl 2-(3-(3-(3-bromo-2- methylphenoxy)propyl)piperidin-1-yl)acetate To a solution of (S)-3-(3-(3-bromo-2-methylphenoxy)propyl)piperidine (2.2 g, 6.31 mmol, 1 equiv., HCl) in CH3CN (20 mL) was added ethyl 2-bromoacetate (948.26 mg, 5.68 mmol, 627.98 μL, 0.9 equiv.) and K2CO3 (2.62 g, 18.93 mmol, 3 equiv.) at 25 °C. The reaction mixture was stirred at 60 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 2% MeOH/DCM) to give (S)-ethyl 2-(3-(3-(3-bromo-2- methylphenoxy)propyl)piperidin-1-yl)acetate (1.9 g, 4.7 mmol, 75.6% yield) as a colorless oil. 1 H NMR (400 MHz, CDCl3-d) δ = 7.14 (d, J = 8.0 Hz, 1H), 6.99 (t, J = 8.0 Hz, 1H), 6.75 (d, J = 8.0 Hz, 1H), 4.19 (q, J = 7.2 Hz, 2H), 3.93 (t, J = 6.4 Hz, 2H), 3.21 (s, 2H), 2.95 - 2.86 (m, 2H), 2.31 (s, 3H), 2.18 - 2.08 (m, 1H), 1.90 - 1.76 (m, 4H), 1.75 - 1.65 (m, 3H), 1.44 - 1.32 (m, 2H), 1.31 - 1.26 (m, 3H), 0.98 - 0.84 (m, 1H) Step H. Procedure for preparation of (S)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-ethoxy-2-oxoe thyl)piperidin-3- yl)propoxy) -2-methylphenyl)picolinate tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)picolinate (600 mg, 979.52 μmol, 1 equiv.), (S)-ethyl 2- (3-(3-(3-bromo-2-methylphenoxy)propyl)piperidin-1-yl)acetate (390.18 mg, 979.52 μmol, 1 equiv.), KF (1.5 M, 1.96 mL, 3 equiv.) and [2-(2-aminophenyl) phenyl]palladium (1+) ;bis (1- adamantyl) -butyl-phosphane;methanesulfonate (71.34 mg, 97.95 μmol, 0.1 equiv.) were taken up into a microwave tube in dioxane (6 mL). The sealed tube was heated at 100 °C for 60 minutes under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~50% ethyl acetate/petroleum ether) to give (S)-tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2- ethoxy-2-oxoethyl)piperidin-3- yl)propoxy) -2-methylphenyl)picolinate (476 mg, 558.2 μmol, 57.0% yield, 94.3% purity) as a white solid. MS (ESI) m/z: 804.8 [M+H] + . Step I. Procedure for preparation of (S)-2-(3-(3-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)propyl)piperidin-1-yl)acetic acid To a solution of (S)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-ethoxy-2-oxoethyl)piperidin-3-yl)pro poxy)-2- methylphenyl)picolinate (476 mg, 592.04 μmol, 1 equiv.) in THF (5 mL) and H2O (0.5 mL) was added LiOH•H 2 O (74.53 mg, 1.78 mmol, 3 equiv.) at 25 °C. The reaction mixture was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give (S)-2-(3-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin-2(1H)-yl)- 2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)propyl) piperidin-1-yl)acetic acid (580 mg, crude) as a yellow solid. MS (ESI) m/z: 776.8 [M+H] + . Step J. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3S)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-3-yl)propoxy)-2- methylphenyl)picolinate To a solution of (S)-2-(3-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)piperidin-1- yl)acetic acid (150 mg, 193.31 μmol, 1 equiv.) in DMF (1.5 mL) was added 3-(6-amino-1- methyl-1H-indazol-3-yl)piperidine-2,6-dione (49.93 mg, 193.31 μmol, 1 equiv.), HATU (80.85 mg, 212.64 μmol, 1.1 equiv.), and DIPEA (74.95 mg, 579.93 μmol, 101.01 μL, 3 equiv.) at 25 °C. The reaction mixture was stirred at 25 °C for 16 hours. The mixture was triturated with water and filtered to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3S)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)piperidin-3-yl)propoxy)-2-met hylphenyl)picolinate (140 mg, 112.3 μmol, 58.0% yield, 81.5% purity) as a yellow solid. MS (ESI) m/z: 1016.8 [M+H] + . Step K. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3S)-1-(2-((3-(2,6-dio xo piperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-3-yl)propoxy)-2- methylphenyl) picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-((3S)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-6-yl)amino)-2- oxoethyl)piperidin-3-yl)propoxy)-2-methylphenyl)picolinate (140 mg, 112.28 μmol, 81.5% purity, 1 equiv.) in DCM (1 mL) and TFA (1 mL) was stirred at 25 °C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3S)-1-(2-((3-(2,6-dio xo piperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)piperidin-3-yl)propoxy)-2-met hylphenyl) picolinic acid (41.0 mg, 41.5 μmol, 37.0% yield, 97.1% purity) as a white solid. MS (ESI) m/z: 960.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.87 (s, 1H), 9.85 (s, 1H), 8.06 - 8.00 (m, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.65 - 7.58 (m, 2H), 7.49 - 7.32 (m, 5H), 7.20 (d, J = 8.4 Hz, 1H), 7.09 - 7.04 (m, 1H), 6.94 (d, J = 8.8 Hz, 1H), 6.85 (d, J = 8.4 Hz, 1H), 6.62 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.31 (dd, J = 5.2, 9.6 Hz, 1H), 3.97 - 3.91 (m, 3H), 3.90 (s, 4H), 3.20 - 3.06 (m, 4H), 3.02 (t, J = 5.6 Hz, 2H), 2.90 - 2.77 (m, 2H), 2.69 - 2.59 (m, 2H), 2.38 - 2.28 (m, 1H), 2.20 - 2.09 (m, 2H), 1.90 (s, 1H), 1.88 (s, 3H), 1.81 - 1.72 (m, 3H), 1.66 - 1.58 (m, 2H), 1.37 (d, J = 6.4 Hz, 2H) Example 53. Preparation of Compound 208 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[2-[4-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]piperazin-1-yl ]ethyl]phenoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of methyl 2-[4-(3-bromo-2-methyl- phenoxy)phenyl]acetate A mixture of methyl 2-(4-hydroxyphenyl)acetate (1.7 g, 10.23 mmol, 1 equiv.), 1,3-dibromo- 2-methyl-benzene (7.67 g, 30.69 mmol, 3 equiv.), CuI (974.18 mg, 5.12 mmol, 0.5 equiv.), cesium carbonate (4.00 g, 12.28 mmol, 1.2 equiv.), and 2,2,6,6-tetramethylheptane-3,5-dione (471.30 mg, 2.56 mmol, 526.59 µL, 0.25 equiv.) in NMP (20 mL) was stirred at 120 °C for 16 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~3% ethyl acetate/petroleum ether) to give methyl 2-[4-(3-bromo-2-methyl-phenoxy)phenyl]acetate (3 g, 8.9 mmol, 87.4% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.44 (d, J = 8.0 Hz, 1H), 7.26 (d, J = 8.8 Hz, 2H), 7.17 (t, J = 8.0 Hz, 1H), 6.91 (d, J = 8.4 Hz, 1H), 6.88 (d, J = 8.4 Hz, 2H), 3.65 (s, 2H), 3.61 (s, 3H), 2.26 (s, 3H) Step B. Procedure for preparation of 2-[4-(3-bromo-2-methyl-phenoxy)phenyl]ethanol To a solution of methyl 2-[4-(3-bromo-2-methyl-phenoxy)phenyl]acetate (1 g, 2.98 mmol, 1 equiv.) in THF (10 mL) was purged with N 2 three times and added in portions LiAlH 4 (90.59 mg, 2.39 mmol, 0.8 equiv.) under N2 at 0 °C. The mixture was stirred under N2 at 0 °C for 0.5 hour. The reaction mixture was quenched with Na 2 SO 4 •10 H 2 O (100 mg) and slowly warmed to 20 °C. The mixture was filtered and washed with DCM (10 mL). Then the organic layers were concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 30% ethyl acetate / petroleum ether) to give 2-[4-(3- bromo-2-methyl-phenoxy)phenyl]ethanol (760 mg, 2.4 mmol, 82.9% yield) as a white oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.41 (dd, J = 0.8, 8.0 Hz, 1H), 7.22 - 7.19 (m, 2H), 7.17 - 7.12 (m, 1H), 6.87 - 6.82 (m, 3H), 4.64 - 4.61 (m, 1H), 3.61 - 3.57 (m, 2H), 2.72 - 2.67 (m, 2H), 2.26 (s, 3H) Step C. Procedure for preparation of 2-[4-(3-bromo-2-methyl- phenoxy)phenyl]acetaldehyde To a solution of 2-[4-(3-bromo-2-methyl-phenoxy)phenyl]ethanol (760 mg, 2.47 mmol, 1 equiv.) in DCM (7.6 mL) was added DMP (1.26 g, 2.97 mmol, 919.16 µL, 1.2 equiv.) in an ice bath. The mixture was stirred at 0 °C for 2 hours. The reaction mixture was quenched by addition Na 2 S 2 O 3 (5 mL) at 0°C and extracted with DCM (5 mL), filtered, and concentrated under reduced pressure to give 2-[4-(3-bromo-2-methyl-phenoxy)phenyl]acetaldehyde (700 mg, crude) as a yellow solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 9.69 - 9.66 (m, 1H), 7.98 (dd, J = 1.2, 8.0 Hz, 1H), 7.70 (dd, J = 1.6, 7.6 Hz, 1H), 7.44 (dd, J = 0.8, 8.0 Hz, 1H), 7.25 - 7.22 (m, 2H), 6.92 - 6.89 (m, 2H), 3.77 - 3.71 (m, 2H), 1.91 (s, 3H) Step D. Procedure for preparation of 2-[[4-(3-bromo-2-methyl-phenoxy)phenyl]methyl]- 1,3-dioxolane To a solution of 2-[4-(3-bromo-2-methyl-phenoxy)phenyl]acetaldehyde (600 mg, 1.97 mmol, 1 equiv.) and ethylene glycol (1.85 g, 29.81 mmol, 1.67 mL, 15.16 equiv.) in toluene (50 mL) was added TsOH (67.71 mg, 393.23 µmol, 0.2 equiv.). The mixture was stirred at 140 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~10% ethyl acetate / petroleum ether) to give 2-[[4-(3-bromo-2-methyl-phenoxy)phenyl]methyl]-1,3-dioxolane (680 mg, crude) as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.42 (dd, J = 0.8, 8.0 Hz, 1H), 7.26 - 7.22 (m, 2H), 7.15 (t, J = 8.0 Hz, 1H), 6.90 - 6.85 (m, 2H), 6.85 - 6.83 (m, 1H), 4.96 - 4.92 (m, 1H), 3.89 - 3.85 (m, 2H), 3.77 - 3.73 (m, 2H), 2.84 (d, J = 5.2 Hz, 2H), 2.26 (s, 3H) Step E. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-(1,3-dioxolan-2-ylm ethyl)phenoxy]-2-methyl- phenyl]pyridine-2-carboxylate 2-[[4-(3-bromo-2-methyl-phenoxy)phenyl]methyl]-1,3-dioxolane (680 mg, 1.95 mmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-2-carb oxylate (1.19 g, 1.95 mmol, 1 equiv.), KF (1.5 M, 3.89 mL, 3 equiv.), and [2-(2-aminophenyl)phenyl]palladium(1+);bis(1- adamantyl)-butyl-phosphane;methanesulfonate (283.62 mg, 389.44 µmol, 0.2 equiv.) were taken up into a microwave tube in dioxane (6.8 mL).The sealed tube was heated at 100 °C for 1 hour under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~20% ethyl acetate/petroleum ether) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[4-(1,3-dioxolan-2-ylmethy l)phenoxy]-2-methyl- phenyl]pyridine-2-carboxylate (650 mg, 800.8 µmol, 41.1% yield, 93% purity) as a white solid. MS (ESI) m/z: 755.5[M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.86 (s, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.60 (d, J = 7.6 Hz, 1H), 7.52 (d, J = 8.8 Hz, 1H), 7.49 - 7.43 (m, 2H), 7.36 (d, J = 7.6, 12.4 Hz, 2H), 7.24 - 7.16 (m, 3H), 6.97 (d, J = 8.8 Hz, 1H), 6.89 - 6.79 (m, 4H), 4.99 (s, 2H), 4.93 (t, J = 5.2 Hz, 1H), 3.90 - 3.84 (m, 4H), 3.80 - 3.72 (m, 2H), 3.04 (t, J = 6.0 Hz, 2H), 2.83 (d, J = 4.8 Hz, 2H), 1.90 (s, 3H), 1.06 (d, J = 2.0 Hz, 9H) Step F. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[2-methyl-3-[4-(2-oxoethyl)phenoxy]ph enyl]pyridine-2- carboxylic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[4-(1,3-dioxolan-2-ylmethyl)phenoxy]-2-methyl-pheny l]pyridine-2-carboxylate (200 mg, 264.94 µmol, 1 equiv.) in HCOOH (3 mL) was stirred at 100 o C for 2 hours. The reaction mixture was concentrated under reduced pressure to give 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[2-methyl-3- [4-(2- oxoethyl)phenoxy]phenyl]pyridine-2-carboxylic acid (180 mg, 237.5 µmol, 89.6% yield, 86.4% purity) as a white solid. MS (ESI) m/z: 655.5[M+H]+. Step G. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[4-[2-[4-[3-(2,6-dioxo-3-piperidyl )-1-methyl-indazol-6- yl]piperazin-1-yl]ethyl]phenoxy]-2-methyl-phenyl]pyridine-2- carboxylic acid To a solution of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- [2-methyl-3-[4-(2-oxoethyl)phenoxy]phenyl]pyridine-2-carboxy lic acid (50 mg, 76.37 µmol, 1 equiv.) and 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (75.00 mg, 229.10 µmol, 3 equiv.) in EtOH (1 mL) and THF (1 mL) was added AcOH (458.58 µg, 7.64 µmol, 4.37e-1 µL, 0.1 equiv.). The mixture was stirred at 25 °C for 15.5 hours. Then was added NaBH3CN (14.40 mg, 229.10 µmol, 3 equiv.). The mixture was stirred at 25 °C for 0.5 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[4-[2-[4-[3-(2,6-dioxo-3-piperidyl)-1 -methyl-indazol-6-yl]piperazin-1- yl]ethyl]phenoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid (14.4 mg, 14.0 µmol, 36.6% yield, 93.7% purity) as a yellow solid. MS (ESI) m/z: 967.1[M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.85 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.53 - 7.48 (m, 2H), 7.48 - 7.43 (m, 2H), 7.39 - 7.33 (m, 2H), 7.22 (d, J = 8.4 Hz, 2H), 7.17 (t, J = 7.6 Hz, 1H), 7.00 (d, J = 8.8 Hz, 1H), 6.94 - 6.91 (m, 1H), 6.89 (d, J = 7.2 Hz, 2H), 6.85 - 6.81 (m, 3H), 5.00 (s, 2H), 4.26 (dd, J = 5.2, 9.2 Hz, 1H), 3.92 (t, J = 6.0 Hz, 2H), 3.89 (s, 3H), 3.24 (s, 4H), 3.03 (t, J = 5.6 Hz, 2H), 2.79 - 2.74 (m, 2H), 2.66 (s, 4H), 2.63 - 2.58 (m, 4H), 2.35 - 2.25 (m, 1H), 2.20 - 2.11 (m, 1H), 1.90 (s, 3H) Example 54. Preparation of Compound 219a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[2-[(3S)-1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-o xo-ethyl]pyrrolidin-3- yl]ethoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl (3S)-3-[2-(3-bromo-2-methyl- phenoxy)ethyl]pyrrolidine-1-carboxylate A mixture of 3-bromo-2-methyl-phenol (1.30 g, 6.97 mmol, 1 equiv.), tert-butyl (3S)-3-(2- hydroxyethyl)pyrrolidine-1-carboxylate (1.5 g, 6.97 mmol, 1 equiv.), 2-(tributyl-λ 5 - phosphanylidene)acetonitrile (2.52 g, 10.45 mmol, 1.5 equiv.) in toluene (20 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 120 °C for 6 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~10% ethyl acetate/petroleum ether) to give tert-butyl (3S)-3-[2-(3-bromo-2-methyl- phenoxy)ethyl]pyrrolidine-1-carboxylate (2.3 g, 6.1 mmol, 88.8% yield) as a yellow oil. Step B. Procedure for preparation of (3S)-3-[2-(3-bromo-2-methyl- phenoxy)ethyl]pyrrolidine A mixture of tert-butyl (3S)-3-[2-(3-bromo-2-methyl-phenoxy)ethyl]pyrrolidine-1- carboxylate (2.38 g, 6.19 mmol, 1 equiv.) in HCl/EtOAc (10 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 2 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The compound (3S)-3-[2-(3-bromo-2-methyl-phenoxy)ethyl]pyrrolidine (1.6 g, 4.9 mmol, 80.5% yield) was obtained as a white solid. Step C. Procedure for preparation of ethyl 2-[(3S)-3-[2-(3-bromo-2-methyl- phenoxy)ethyl]pyrrolidin-1-yl]acetate A mixture of (3S)-3-[2-(3-bromo-2-methyl-phenoxy)ethyl]pyrrolidine (1.6 g, 5.63 mmol, 1 equiv.), ethyl 2-bromoacetate (940.22 mg, 5.63 mmol, 622.66 µL, 1 equiv.), K 2 CO 3 (3.89 g, 28.15 mmol, 5 equiv.) in CH 3 CN (10 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 60 °C for 2 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~23% ethyl acetate/petroleum ether) to give ethyl 2- [(3S)-3-[2-(3-bromo-2-methyl-phenoxy)ethyl]pyrrolidin-1-yl]a cetate (1.1 g, 2.9 mmol, 51.6% yield, 97.9% purity) as a yellow oil. MS (ESI) m/z: 372.3 [M+2] + Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[(3S)-1-(2-ethoxy-2 -oxo-ethyl)pyrrolidin-3- yl]ethoxy]-2-methyl-phenyl]pyridine-2-carboxylate A mixture of ethyl 2-[(3S)-3-[2-(3-bromo-2-methyl-phenoxy)ethyl]pyrrolidin-1-yl ]acetate (300 mg, 810.20 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)pyridine-2- carboxylate (496.28 mg, 810.20 µmol, 1 equiv.), K 2 CO 3 (1.5 M, 810.20 µL, 1.5 equiv.), Ad2nBuP Pd G3 (118.01 mg, 162.04 µmol, 0.2 equiv.) in dioxane (5 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 100 °C for 1 hour in microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~100% ethyl acetate/petroleum ether) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[2-[(3S)-1-(2-ethoxy-2-oxo-ethyl)pyrrolidin-3-yl]et hoxy]-2-methyl-phenyl]pyridine- 2-carboxylate (430 mg, 554.1 µmol, 68.4% yield) as a yellow oil. MS (ESI) m/z: 776.9 [M+H] + . Step E. Procedure for preparation of 2-[(3S)-3-[2-[3-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-2- methyl-phenoxy]ethyl]pyrrolidin-1-yl]acetic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[2-[(3S)-1-(2-ethoxy-2-oxo-ethyl)pyrrolidin-3-yl]et hoxy]-2-methyl-phenyl]pyridine- 2-carboxylate (105 mg, 135.32 µmol, 1 equiv.), LiOH (1 M, 405.95 µL, 3 equiv.) in THF (1 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 25 °C for 2 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was taken up in HCl 0.5 mL and filtered. The solid was dissolved in DCM and concentrated under reduced pressure to give a residue. The compound 2-[(3S)-3-[2-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H-isoquinolin-2-yl]-2- tert-butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]ethyl]pyrrol idin-1-yl]acetic acid (98 mg, 131.0 µmol, 96.8% yield) was obtained as a white solid. Step F. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[(3S)-1-[2-[[3-(2,6 -dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]pyrrolidin-3-yl]ethoxy]-2-me thyl-phenyl]pyridine-2- carboxylate A mixture of 2-[(3S)-3-[2-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]ethyl]pyrrolidin-1- yl]acetic acid (80 mg, 106.97 µmol, 1 equiv.), 3-(6-amino-1-methyl-indazol-3-yl)piperidine- 2,6-dione (27.63 mg, 106.97 µmol, 1 equiv.), EDCI (30.76 mg, 160.45 µmol, 1.5 equiv.) in pyridine (1 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 2 hours under N2 atmosphere. The reaction mixture was then diluted with water 5 mL and extracted with ethyl acetate 30 mL (15 mL × 2). The combined organic layers were washed with brine 20 mL (10 mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The compound tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[(3S)- 1-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]pyrrolid in-3-yl]ethoxy]-2-methyl- phenyl]pyridine-2-carboxylate (150 mg, 74.8 µmol, 69.9% yield, 49.3% purity) was obtained as a red oil. MS (ESI) m/z: 989.4 [M+H] + . Step G. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[2-[(3S)-1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]pyrrolidin-3-yl]ethoxy]-2-methyl-pheny l]pyridine-2-carboxylic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[2-[(3S)-1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo- ethyl]pyrrolidin-3-yl]ethoxy]-2-methyl-phenyl]pyridine-2-car boxylate (150 mg, 151.80 µmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 12 hours under N 2 atmosphere. The reaction mixture was diluted with water 5 mL and extracted with ethyl acetate 10 mL (5 mL × 2). The combined organic layers were washed with brine 10 mL (5 mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[2- [(3S)-1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl ]amino]-2-oxo-ethyl]pyrrolidin- 3-yl]ethoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid (28.7 mg, 29.8 µmol, 19.6% yield, 96.7% purity) as a purple solid. MS (ESI) m/z: 933.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.01 - 12.71 (m, 1H), 10.88 (s, 1H), 10.49 - 9.89 (m, 1H), 8.14 (s, 1H), 8.06 - 8.00 (m, 2H), 7.81 - 7.76 (m, 1H), 7.66 - 7.61 (m, 2H), 7.48 - 7.43 (m, 2H), 7.40 (d, J = 4.8 Hz, 1H), 7.39 - 7.32 (m, 2H), 7.16 (d, J = 9.2 Hz, 1H), 7.12 (s, 1H), 6.93 (dd, J = 8.4, 18.4 Hz, 2H), 6.64 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.32 (dd, J = 5.2, 9.6 Hz, 1H), 4.01 (s, 2H), 3.93 (d, J = 1.6 Hz, 2H), 3.91 (s, 3H), 3.03 (t, J = 6.0 Hz, 4H), 2.71 - 2.57 (m, 4H), 2.46 (s, 2H), 2.37 - 2.27 (m, 2H), 2.20 - 2.11 (m, 2H), 1.91 (s, 5H), 1.68 - 1.58 (m, 1H) Example 55. Preparation of Compound 219b 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[2-[(3R)-1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-o xo-ethyl]pyrrolidin-3- yl]ethoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl (3R)-3-[2-(3-bromo-2-methyl- phenoxy)ethyl]pyrrolidine-1-carboxylate To a solution of 3-bromo-2-methyl-phenol (764.51 mg, 4.09 mmol, 1.1 equiv.) in THF (32 mL) was added tert-butyl (3R)-3-(2-hydroxyethyl)pyrrolidine-1-carboxylate (800 mg, 3.72 mmol, 1 equiv.), DIAD (1.20 g, 5.95 mmol, 1.16 mL, 1.6 equiv.) and PPh3 (1.56 g, 5.95 mmol, 1.6 equiv.). The mixture was stirred at 25 °C for 8 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 37% ethyl acetate / petroleum ether) to give tert-butyl (3R)-3- [2-(3-bromo-2-methyl-phenoxy)ethyl]pyrrolidine-1-carboxylate (400 mg, 822.2 µmol, 22.1% yield, 79% purity) as a white solid. MS (ESI) m/z: 329.7 [M-56+1] + 1 H NMR (400 MHz, CD3OD) δ = 7.14 - 7.12 (m, 1H), 7.06 - 7.00 (m, 1H), 6.90 - 6.88 (m, 1H), 4.08 - 3.99 (m, 2H), 3.65-3.57 (m, 1H), 3.49 - 3.44 (m, 1H), 3.38 - 3.35 (s, 1H), 3.03 - 2.96 (m, 1H), 2.31 (s, 3H), 2.15 - 2.06 (m, 1H), 1.94 - 1.87 (m, 2H), 1.69 - 1.56 (m, 1H), 1.47 (s, 9H), 1.32 - 1.24 (m, 1H). Step B. Procedure for preparation of (3R)-3-[2-(3-bromo-2-methyl-phenoxy)ethyl] pyrrolidine To a solution of tert-butyl (3R)-3-[2-(3-bromo-2-methyl-phenoxy)ethyl]pyrrolidine-1- carboxylate (300 mg, 208.17 µmol, 1 equiv.) in HCl / EtOAc (10 mL). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was used in the next step without purification. The compound (3R)-3-[2- (3-bromo-2-methyl-phenoxy)ethyl] pyrrolidine (180 mg, 561.3 µmol, 71.9% yield) was obtained as a white solid. MS (ESI) m/z: 285.8 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 7.18 - 7.17 (m, 1H), 7.11 (t, J = 8.0 Hz, 1H), 7.02 - 6.97 (m, 1H), 4.05 - 4.02 (m, 2H), 3.28 - 3.18 (m, 1H), 2.83 - 2.75 (m, 1H), 2.42 - 2.34 (m, 1H), 2.25 (s, 3H), 2.13 - 2.09 (m, 1H), 2.00 - 1.98 (s, 1H), 1.92 - 1.86 (m, 2H), 1.64 - 1.54 (m, 1H), 1.25 - 1.13 (m, 2H). Step C. Procedure for preparation of ethyl 2-[(3R)-3-[2-(3-bromo-2-methyl- phenoxy)ethyl]pyrrolidin-1-yl]acetate A mixture of (3R)-3-[2-(3-bromo-2-methyl-phenoxy)ethyl]pyrrolidine (160 mg, 479.02 µmol, 96% purity, 1 equiv., HCl), ethyl 2-bromoacetate (80.00 mg, 479.02 µmol, 52.98 µL, 1 equiv.), and K2CO3 (198.61 mg, 1.44 mmol, 3 equiv.) in CH3CN (4 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 60 °C for 2 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 37% ethyl acetate / petroleum ether) to give ethyl 2-[(3R)-3-[2-(3-bromo-2-methyl-phenoxy)ethyl]pyrrolidin-1-yl ]acetate (150 mg, 405.1 µmol, 84.5% yield) as a white solid. MS (ESI) m/z: 371.9 [M+H] + Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[(3R)-1-(2-ethoxy-2 -oxo-ethyl)pyrrolidin-3- yl]ethoxy]-2-methyl-phenyl]pyridine-2-carboxylate A mixture of ethyl 2-[(3R)-3-[2-(3-bromo-2-methyl-phenoxy)ethyl]pyrrolidin-1-yl ]acetate (80 mg, 216.05 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (158.81 mg, 259.26 µmol, 1.2 equiv.), Ad 2 nBuP Pd G3(cataCXium® A Pd G3 ) (31.47 mg, 43.21 µmol, 0.2 equiv.), and K2CO3 (1.5 M, 216.05 µL, 1.5 equiv.) in 1,4-dioxane (4 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 100 °C for 1 hour under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 37% ethyl acetate / petroleum ether) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[(3R)-1-(2-ethoxy-2-oxo -ethyl)pyrrolidin-3-yl]ethoxy]- 2-methyl-phenyl]pyridine-2-carboxylate (80 mg, 103.1 µmol, 47.7% yield) as a white solid. MS (ESI) m/z: 776.7 [M+H] + Step E. Procedure for preparation of 2-[(3R)-3-[2-[3-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-2- methyl-phenoxy]ethyl]pyrrolidin-1-yl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[2-[(3R)-1-(2-ethoxy-2-oxo-ethyl)pyrrolidin-3-yl] ethoxy]-2-methyl- phenyl]pyridine-2-carboxylate (80 mg, 103.10 µmol, 1 equiv.) in THF (2 mL) was added LiOH•H2O (12.98 mg, 309.30 µmol, 3 equiv.) and H2O (0.5 mL). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue used in the next step without purification. The compound 2-[(3R)-3-[2-[3- [6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoqui nolin-2-yl]-2-tert- butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]ethyl]pyrrolidin- 1-yl]acetic acid (80 mg, crude) was obtained as a white solid. MS (ESI) m/z: 748.7 [M+H] + Step F. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[(3R)-1-[2-[[3-(2,6 -dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]pyrrolidin-3-yl]ethoxy]-2-me thyl-phenyl]pyridine-2- carboxylate A mixture of 2-[(3R)-3-[2-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]ethyl]pyrrolidin-1- yl]acetic acid (80 mg, 106.97 µmol, 1 equiv.), 3-(6-amino-1-methyl-indazol-3-yl)piperidine- 2,6-dione (33.15 mg, 128.36 µmol, 1.2 equiv.), HATU (40.67 mg, 106.97 µmol, 1 equiv.), DIPEA (41.47 mg, 320.90 µmol, 55.89 µL, 3 equiv.) in DMF (3 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 25 °C for 2 hours under N 2 atmosphere. The reaction mixture was diluted with water (6 mL) and extracted with ethyl acetate (3 mL × 3). The combined organic layers were washed with water (3 mL × 3), filtered, and concentrated under reduced pressure to give a residue. The residue was used in the next step without purification. The compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[2-[(3R)-1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]pyrrolidin-3-yl]ethoxy]-2-methyl-pheny l]pyridine-2-carboxylate (70 mg, 70.8 µmol, 66.2% yield) was obtained as a white solid. MS (ESI) m/z: 721.5 [M+H] + Step G. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[2-[(3R)-1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]pyrrolidin-3-yl]ethoxy]-2-methyl-pheny l]pyridine-2-carboxylic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[2-[(3R)-1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo- ethyl]pyrrolidin-3-yl]ethoxy]-2-methyl-phenyl]pyridine-2-car boxylate (70 mg, 70.84 µmol, 1 equiv.) in DCM (1 mL) and TFA (1 mL) was stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep- HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3- [2-[(3R)-1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6 -yl]amino]-2-oxo- ethyl]pyrrolidin-3-yl]ethoxy]-2-methyl-phenyl]pyridine-2-car boxylic acid (32.4 mg, 34.5 µmol, 48.8% yield, 99.4% purity) as a white solid. MS (ESI) m/z: 932.8 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 10.87 (s, 1H), 9.89 (s, 1H), 8.14 (s, 1H), 8.04 - 8.02 (m, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.63 - 7.59 (m, 2H), 7.48 - 7.33 (m, 5H), 7.21 - 7.18 (m, 1H), 7.11 - 7.06 (m, 1H), 6.97 - 6.84 (m, 2H), 6.62 (d, J = 7.2 Hz, 1H), 5.04 - 4.95 (m, 2H), 4.33 - 4.29 (m, 1H), 4.00 - 3.90 (m, 7H), 3.04 - 3.02 (m, 2H), 2.98 - 2.94 (m, 1H), 2.81 - 2.77 (m, 1H), 2.70 - 2.60 (m, 4H), 2.40 - 2.27 (m, 4H), 2.18 - 2.14 (m, 1H), 2.08 – 1.98 (m, 1H), 1.93 - 1.85 (m, 5H), 1.55 - 1.50 (m, 1H). Example 56. Preparation of Compound 226 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[[7-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-oxo-e thyl]-7-azaspiro[3.5]nonan-2- yl]methoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 2-[(3-bromo-2-methyl-phenoxy)methyl]- 7-azaspiro[3.5]nonane-7-carboxylate A mixture of 3-bromo-2-methyl-phenol (1 g, 5.35 mmol, 1 equiv.), tert-butyl 2- (hydroxymethyl)-7-azaspiro[3.5]nonane-7-carboxylate (1.50 g, 5.88 mmol, 1.1 equiv.), and 2- (tributyl-λ5-phosphanylidene) acetonitrile (1.55 g, 6.42 mmol, 1.2 equiv.) in toluene (10 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 120 °C for 2 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 0/1 to 10/1) to give compound tert-butyl 2-[(3-bromo-2-methyl- phenoxy)methyl]-7-azaspiro[3.5]nonane-7-carboxylate (2.35 g, crude) as a green oil. 1 H NMR (400 MHz, CDCl 3 ) δ = 7.15 (d, J = 8.0 Hz, 1H), 6.99 (t, J = 8.0 Hz, 1H), 6.76 (d, J = 8.4 Hz, 1H), 3.91 (d, J = 6.0 Hz, 2H), 3.42 - 3.35 (m, 2H), 3.34 - 3.26 (m, 2H), 2.83 - 2.67 (m, 1H), 2.33 (s, 3H), 2.03 - 1.94 (m, 2H), 1.78 - 1.69 (m, 2H), 1.66 - 1.60 (m, 2H), 1.52 - 1.49 (m, 2H), 1.46 (s, 9H). Step B. Procedure for preparation of 2-[(3-bromo-2-methyl-phenoxy)methyl]-7- azaspiro[3.5]nonane To a solution of tert-butyl 2-[(3-bromo-2-methyl-phenoxy)methyl]-7-azaspiro[3.5]nonane-7 - carboxylate (1.85, 4.36 mmol, 1 equiv.) in DCM (10 mL) was added HCl/1,4-dioxane (4 M, 10 mL, 9.18 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , DCM: MeOH = 0/1 to 10/1) to give compound 2-[(3-bromo-2-methyl- phenoxy)methyl]-7-azaspiro[3.5]nonane (1.4 g, 4.32 mmol, 99.04% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ = 7.18 - 7.14 (m, 1H), 7.09 (t, J = 8.0 Hz, 1H), 6.96 (d, J = 8.0 Hz, 1H), 3.96 (d, J = 6.0 Hz, 2H), 3.02 - 2.97 (m, 2H), 2.93 - 2.88 (m, 2H), 2.74 - 2.66 (m, 1H), 2.24 (s, 3H), 2.01 - 1.92 (m, 2H), 1.81 - 1.76 (m, 2H), 1.75 - 1.70 (m, 2H), 1.70 - 1.67 (m, 2H). Step C. Procedure for preparation of ethyl 2-[2-[(3-bromo-2-methyl-phenoxy)methyl]-7- azaspiro[3.5]nonan-7-yl]acetate To a solution of 2-[(3-bromo-2-methyl-phenoxy)methyl]-7-azaspiro[3.5]nonane (500 mg, 1.54 mmol, 1 equiv.) and ethyl 2-bromoacetate (257.51 mg, 1.54 mmol, 170.54 µL, 1 equiv.) in CH 3 CN (5 mL) was added K 2 CO 3 (1.07 g, 7.71 mmol, 5 equiv.). The mixture was stirred at 60 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH = 10: 1) to give compound ethyl 2-[2-[(3-bromo-2-methyl-phenoxy)methyl]-7-azaspiro[3.5]nonan -7- yl]acetate (240 mg, 584.8 µmol, 37.9% yield) as a colorless oil. 1 H NMR (400 MHz, CDCl3) δ = 7.15 (d, J = 8.0 Hz, 1H), 6.99 (t, J = 8.0 Hz, 1H), 6.76 (d, J = 8.4 Hz, 1H), 4.25 - 4.15 (m, 2H), 3.90 (d, J = 6.4 Hz, 2H), 3.20 (s, 2H), 2.80 - 2.66 (m, 1H), 2.62 - 2.39 (m, 4H), 2.32 (s, 3H), 2.04 - 1.90 (m, 2H), 1.76 (t, J = 4.4 Hz, 2H), 1.58 (s, 4H), 1.28 (t, J = 7.2 Hz, 3H). Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[[7-(2-ethoxy-2-oxo-et hyl)-7-azaspiro[3.5]nonan- 2-yl]methoxy]-2-methyl-phenyl]pyridine-2-carboxylate Ethyl 2-[2-[(3-bromo-2-methyl-phenoxy)methyl]-7-azaspiro[3.5]nonan -7-yl]acetate (240 mg, 584.87 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (358.26 mg, 584.87 µmol, 1 equiv.) K 2 CO 3 (1.5 M, 1.17 mL, 3 equiv.) and Ad 2 nBuP Pd G3(cataCXium® A Pd G3) (85.19 mg, 116.97 µmol, 0.2 equiv.) were taken up into a microwave tube in 1,4-dioxane (3 mL). The sealed tube was heated at 100 °C for 1 hour under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate = 0/1 to 1/1) to give compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[[7-(2-ethoxy-2-oxo-ethyl)-7-azaspiro [3.5]nonan-2-yl]methoxy]-2- methyl-phenyl]pyridine-2-carboxylate (240 mg, 294.1 µmol, 50.2% yield) as a yellow solid. 1 H NMR (400 MHz, CDCl3) δ = 7.86 (d, J = 7.6 Hz, 1H), 7.72 (d, J = 8.0 Hz, 1H), 7.57 (d, J = 7.2 Hz, 1H), 7.47 - 7.41 (m, 1H), 7.38 (d, J = 8.4 Hz, 2H), 7.35 - 7.30 (m, 2H), 7.14 - 7.06 (m, 1H), 6.90 (d, J = 8.8 Hz, 1H), 6.80 (d, J = 8.0 Hz, 1H), 6.70 (d, J = 7.6 Hz, 1H), 5.16 - 4.90 (m, 2H), 4.23 - 4.16 (m, 2H), 4.11 (t, J = 6.0 Hz, 2H), 3.94 (d, J = 6.0 Hz, 2H), 3.32 - 3.14 (m, 2H), 3.07 (t, J = 5.6 Hz, 2H), 2.77 - 2.68 (m, 1H), 2.64 - 2.36 (m, 4H), 2.00 (s, 3H), 1.95 (d, J = 9.6 Hz, 2H), 1.82 - 1.66 (m, 6H), 1.28 (s, 3H), 1.15 (s, 9H). Step E. Procedure for preparation of 2-[2-[[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl ]-2-methyl- phenoxy]methyl]-7-azaspiro[3.5]nonan-7-yl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[[7-(2-ethoxy-2-oxo-ethyl)-7-azaspiro[3.5]nonan-2 -yl]methoxy]-2-methyl- phenyl]pyridine-2-carboxylate (240 mg, 294.11 µmol, 1 equiv.) in THF (0.5 mL) and H 2 O (0.5 mL) was added LiOH•H 2 O (61.71 mg, 1.47 mmol, 5 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove THF and adjusted pH to 5 with HCl (1M). The mixture was then filtered concentrated under reduced pressure to give compound 2-[2-[[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]methyl]-7- azaspiro[3.5]nonan-7-yl]acetic acid (270 mg, crude) as a yellow solid. MS (ESI) m/z: 788.9 [M+H] + Step F. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[[7-[2-[[3-(2,6-dioxo- 3-piperidyl)-1-methyl- indazol-7-yl]amino]-2-oxo-ethyl]-7-azaspiro[3.5]nonan-2-yl]m ethoxy]-2-methyl- phenyl]pyridine-2-carboxylate To a solution of 2-[2-[[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin- 2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]meth yl]-7-azaspiro[3.5]nonan-7- yl]acetic acid (80 mg, 101.53 µmol, 1 equiv.) and 3-(7-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (31.47 mg, 121.83 µmol, 1.2 equiv.) in pyridine (1 mL) was added EDCI (29.19 mg, 152.29 µmol, 1.5 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was quenched by addition water 2 mL, and then diluted with EtOAc 2 mL and extracted with EtOAc (2 mL × 3). The combined organic layers were washed with brine (2 mL × 3), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[[7-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indaz ol-7-yl]amino]-2-oxo-ethyl]-7- azaspiro[3.5]nonan-2-yl]methoxy]-2-methyl-phenyl]pyridine-2- carboxylate (100 mg, crude) as a brown oil. MS (ESI) m/z: 1028.8 [M+H] + Step G. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[[7-[2-[[3-(2,6-dioxo-3-piperidyl) -1-methyl-indazol-7- yl]amino]-2-oxo-ethyl]-7-azaspiro[3.5]nonan-2-yl]methoxy]-2- methyl-phenyl]pyridine- 2-carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[[7-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indaz ol-7-yl]amino]-2-oxo-ethyl]-7- azaspiro[3.5]nonan-2-yl]methoxy]-2-methyl-phenyl]pyridine-2- carboxylate (100 mg, 97.26 µmol, 1 equiv.) in DCM (0.5 mL) was added TFA (1.54 g, 13.51 mmol, 1.0 mL, 138.87 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[[7-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-o xo-ethyl]-7- azaspiro[3.5]nonan-2-yl]methoxy]-2-methyl-phenyl]pyridine-2- carboxylic acid (26.7 mg, 27.3 µmol, 28.1% yield, 99.3% purity) as a yellow solid. MS (ESI) m/z: 972.7 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.89 (s, 1H), 9.84 (s, 1H), 8.06 - 7.98 (m, 1H), 7.82 - 7.74 (m, 1H), 7.62 (d, J = 6.4 Hz, 1H), 7.56 (d, J = 8.0 Hz, 1H), 7.50 - 7.40 (m, 3H), 7.39 - 7.29 (m, 3H), 7.07 (t, J = 8.0 Hz, 2H), 6.98 - 6.82 (m, 2H), 6.64 (d, J = 8.0 Hz, 1H), 4.98 (s, 2H), 4.43 - 4.31 (m, 1H), 4.11 (s, 3H), 3.93 (d, J = 5.6 Hz, 4H), 3.13 (s, 2H), 3.02 (s, 2H), 2.82 - 2.67 (m, 2H), 2.65 - 2.60 (m, 1H), 2.46 - 2.34 (m, 4H), 2.23 - 2.11 (m, 2H), 1.96 - 1.87 (m, 5H), 1.75 - 1.65 (m, 4H), 1.61 (d, J = 4.8 Hz, 2H). Example 57. Preparation of Compound 229 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2- oxoethyl)piperidin-4-yl)-3,3- difluoropropoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 4- (methoxy(methyl)carbamoyl)piperidine-1-carboxylate A mixture of 1-(tert-butoxycarbonyl)piperidine-4-carboxylic acid (50.0 g, 218 mmol, 1.0 equiv.), N,O-dimethylhydroxylamine (27.7 g, 283 mmol, 1.3 equiv.), EDCI (100 g, 523 mmol, 2.4 equiv.), NMM (33.1 g, 327 mmol, 36.0 mL, 1.5 equiv.), and HOBt (14.7 g, 109 mmol, 0.50 equiv.) in CH2Cl2 (500 mL) was stirred at 20 °C for 5 hours. After completion, the reaction mixture was diluted with H 2 O (500 mL) and extracted with CH 2 Cl 2 (400 mL × 2). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by column chromatography (SiO 2 , dichloromethane/methanol = 100/1 to 20/1) to give compound tert-butyl 4- (methoxy(methyl)carbamoyl)piperidine-1-carboxylate (52.3 g, 192 mmol, 88.1% yield) as a yellow oil. 1 H NMR (400 MHz) δ = 4.30 - 4.00 (m, 2H), 3.71 (s, 3H), 3.18 (s, 3H), 2.90 - 2.67 (m, 3H), 1.75 - 1.62 (m, 4H), 1.45 (s, 9H). Step B. Procedure for preparation of tert-butyl 4-(but-3-enoyl)piperidine-1-carboxylate To a solution of tert-butyl 4-(methoxy(methyl)carbamoyl)piperidine-1-carboxylate (15.0 g, 55.1 mmol, 1.0 equiv.) in THF (200 mL) was added dropwise allyl magnesium bromide (1 M, 66.1 mL, 1.2 equiv.) at 0 °C. After addition, the mixture was stirred 20 °C for 3 hours. The reaction mixture was quenched by addition saturated NH4Cl aqueous solution (200 mL) at 0 °C and then extracted with CH2Cl2 (200 mL × 2). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, dichloromethane/methanol = 100/1 to 10/1) to give tert-butyl 4-(but-3-enoyl)piperidine-1-carboxylate (14.0 g, 49.7 mmol, 90.3% yield, 90% purity) as a brown oil. 1 H NMR (400 MHz, CDCl 3 ) δ = 5.99 - 5.81 (m, 1H), 5.22 - 5.06 (m, 2H), 4.11 (d, J = 7.2 Hz, 2H), 3.22 (d, J = 6.8 Hz, 2H), 2.77 (t, J = 12.0 Hz, 2H), 2.58 - 2.47 (m, 1H), 1.84 - 1.75 (m, 2H), 1.58 - 1.48 (m, 2H), 1.44 (s, 9H). Step C. Procedure for preparation of tert-butyl 4-(1,1-difluorobut-3-en-1-yl)piperidine-1- carboxylate To a solution of tert-butyl 4-(but-3-enoyl)piperidine-1-carboxylate (14.0 g, 55.3 mmol, 1.0 equiv.) in CH 2 Cl 2 (200 mL) was added DAST (44.5 g, 276 mmol, 36.5 mL, 5.0 equiv.) dropwise at 0 °C. After that, the mixture was stirred at 20 °C for 5 hours. The reaction mixture was quenched by addition saturated NaHCO 3 aqueous solution (200 mL) at 0°C and then extracted with CH 2 Cl 2 (200 mL × 3). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 50/1 to 5/1) to give tert-butyl 4-(1,1- difluorobut-3-en-1-yl)piperidine-1-carboxylate (4.40 g, 16.0 mmol, 28.9% yield) as a colorless oil. 1 H NMR (400 MHz, CDCl3) δ = 5.90 -5.70 (m, 1H), 5.35 - 5.01 (m, 2H), 4.32 - 4.14 (m, 2H), 2.80 - 2.49 (m, 4H), 1.96 - 1.85 (m, 1H), 1.76 (d, J = 12.8 Hz, 2H), 1.46 (s, 9H), 1.44 - 1.37 (m, 2H). Step D. Procedure for preparation of tert-butyl 4-(1,1-difluoro-3- hydroxypropyl)piperidine-1-carboxylate Ozone was bubbled into a solution of tert-butyl 4-(1,1-difluorobut-3-en-1-yl)piperidine-1- carboxylate (4.40 g, 16.0 mmol, 1.0 equiv.) in CH 2 Cl 2 (30.0 mL) and ethanol (10.0 mL) at -78 °C for 1 hour until the solution became blue. After excess ozone was degassed and purged by nitrogen, NaBH4 (1.81 g, 47.9 mmol, 3.0 equiv.) was added in portions at 0 °C, and the reaction was stirred at 0 °C for 1 hour. The reaction mixture was quenched by addition saturated ammonium chloride aqueous solution (80 mL) at 0 °C, and then extracted with CH 2 Cl 2 (40 mL × 2). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate = 40/1 to 1/1) to give compound tert- butyl 4-(1,1-difluoro-3-hydroxypropyl)piperidine-1-carboxylate (3.50 g, 11.3 mmol, 70.6% yield, 90.0% purity) as a yellow oil. 1 H NMR (400 MHz, CDCl 3 ) δ = 4.25 - 4.04 (m, 2H), 3.90 - 3.77 (m, 2H), 2.71 - 2.48 (m, 2H), 2.10 - 1.96 (m, 2H), 1.92 - 1.82 (m, 1H), 1.71 (d, J = 13.2 Hz, 2H), 1.39 (s, 9H), 1.37 - 1.27 (m, 2H). Step E. Procedure for preparation of tert-butyl 4-(3-(3-bromo-2-methylphenoxy)-1,1- difluoropropyl)piperidine-1-carboxylate To a solution of tert-butyl 4-(1,1-difluoro-3-hydroxypropyl)piperidine-1-carboxylate (3.50 g, 12.5 mmol, 1.0 equiv.) in toluene (40.0 mL) was added 3-bromo-2-methylphenol (2.58 g, 13.8 mmol, 1.1 equiv.) and CMPB (6.05 g, 25.1 mmol, 2.0 equiv.). The reaction was stirred at 120 °C for 2 hours. After completion, the mixture was concentrated under reduced pressure and the residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 30/1 to 3/1) to give tert-butyl 4-(3-(3-bromo-2-methylphenoxy)-1,1-difluoropropyl)piperidine -1- carboxylate (2.4 g, 3.5 mmol, 28.3% yield, 63.6% purity) as a yellow oil. MS (ESI) m/z: 448.3 [M+H] + 1 H NMR (400 MHz, CDCl 3 ) δ = 7.10 (d, J = 8.0 Hz, 1H), 6.94 (t, J = 8.4 Hz, 1H), 6.71 (d, J = 8.4 Hz, 1H), 4.19 - 4.06 (m, 4H), 2.64 - 2.54 (m, 2H), 2.38 - 2.27 (m, 2H), 2.23 (s, 3H), 1.97 - 1.89 (m, 1H), 1.76 (d, J = 13.2 Hz, 2H), 1.40 - 1.39 (m, 11H). Step F. Procedure for preparation of 4-(3-(3-bromo-2-methylphenoxy)-1,1- difluoropropyl)piperidine A mixture of tert-butyl 4-(3-(3-bromo-2-methylphenoxy)-1,1-difluoropropyl)piperidine -1- carboxylate (2.26 g, 5.04 mmol, 1.0 equiv.) in HCl/1,4-dioxane (20.0 mL) was stirred at 25 °C for 1 hour. After completion, the reaction mixture was concentrated under reduced pressure to give 4-(3-(3-bromo-2-methylphenoxy)-1,1-difluoropropyl)piperidine (1.76 g, crude) as a yellow oil. MS (ESI) m/z: 347.8 [M+H] + Step G. Procedure for preparation of ethyl 2-(4-(3-(3-bromo-2-methylphenoxy)-1,1- difluoropropyl)piperidin-1-yl)acetate To a solution of 4-(3-(3-bromo-2-methylphenoxy)-1,1-difluoropropyl)piperidine (1.76 g, 5.05 mmol, 1.0 equiv.) and ethyl 2-bromoacetate (802 mg, 4.80 mmol, 531 µL, 0.95 equiv.) in CH 3 CN (20 mL) was added K 2 CO 3 (1.40 g, 10.1 mmol, 2.0 equiv.). The mixture was stirred at 60 °C for 2 hours. After completion, the reaction mixture was diluted with H2O (30 mL) and extracted with CH2Cl2 (30 mL × 2). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate = 30/1 to 3/1) to give ethyl 2-(4-(3-(3-bromo-2-methylphenoxy)-1,1-difluoropropyl)piperid in-1-yl)acetate (1.10 g, 2.45 mmol, 48.5% yield, 96.8% purity) as a yellow oil. MS (ESI) m/z: 435.9 [M+H] + 1 H NMR (400 MHz, CDCl3) δ = 7.10 (d, J = 8.0 Hz, 1H), 6.94 (t, J = 8.0 Hz, 1H), 6.71 (d, J = 8.0 Hz, 1H), 4.18 - 4.05 (m, 4H), 3.15 (s, 2H), 2.97 (d, J = 11.2 Hz, 2H), 2.38 - 2.25 (m, 2H), 2.23 (s, 3H), 2.11 (t, J = 11.2 Hz, 2H), 1.85 - 1.72 (m, 3H), 1.69 - 1.58 (m, 2H), 1.21 (t, J = 7.2 Hz, 3H). Step H. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-ethoxy-2-oxoethyl )piperidin-4-yl)-3,3- difluoropropoxy)-2-methylphenyl)picolinate A mixture of ethyl 2-(4-(3-(3-bromo-2-methylphenoxy)-1,1-difluoropropyl)piperid in-1- yl)acetate (313 mg, 684 µmol, 95% purity, 1.0 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tet ramethyl-1,3,2-dioxaborolan-2- yl)picolinate (440 mg, 718. µmol, 1.05 equiv.), Ad 2 nBuP Pd G 3 (cataCXium® A Pd G 3 ) (99.6 mg, 137 µmol, 0.20 equiv.), K 3 PO 4 (436 mg, 2.05 mmol, 3.0 equiv.) and H 2 O (0.30 mL) in 1,4- dioxane (3.00 mL) was degassed and purged with nitrogen three times, and then the mixture was stirred at 90 °C for 2 hours under nitrogen atmosphere. The reaction mixture was diluted with H 2 O (10 mL) and extracted with CH 2 Cl 2 (10 mL × 3). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=30/1 to 1/1) to give compound tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-ethoxy-2-oxoethyl )piperidin-4-yl)-3,3- difluoropropoxy)-2-methylphenyl)picolinate (357 mg, 372 µmol, 54.4% yield, 87.6% purity) as a yellow solid. MS (ESI) m/z: 840.7 [M+H] + 1 H NMR (400 MHz, CDCl3) δ = 7.86 (d, J = 7.6 Hz, 1H), 7.66 (d, J = 8.0 Hz, 1H), 7.57 (d, J = 8.0 Hz, 1H), 7.44 - 7.28 (m, 5H), 7.12 (t, J = 8.0 Hz, 1H), 6.90 (d, J = 8.8 Hz, 1H), 6.83 (d, J = 8.0 Hz, 1H), 6.73 (d, J = 8.0 Hz, 1H), 5.16 - 4.93 (m, 2H), 4.26 - 4.14 (m, 4H), 4.13 - 4.08 (m, 2H), 3.24 (s, 2H), 3.11 - 3.01 (m, 4H), 2.45 - 2.33 (m, 2H), 2.29 - 2.12 (m, 2H), 1.98 (s, 3H), 1.89 - 1.83 (m, 2H), 1.79 - 1.70 (m, 2H), 1.28 - 1.25 (m, 4H), 1.15 (s, 9H). Step I. Procedure for preparation of 2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyri din-3-yl)-2- methylphenoxy)-1,1-difluoropropyl)piperidin-1-yl)acetic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-ethoxy-2-oxoethyl)piperidin-4-yl)-3, 3-difluoropropoxy)-2- methylphenyl)picolinate (307 mg, 365 µmol, 1.0 equiv.) in THF (2.00 mL) and H2O (2.00 mL) was added LiOH•H2O (43.8 mg, 1.83 mmol, 5.0 equiv.). The mixture was stirred at 20 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give compound 2- (4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydrois oquinolin-2(1H)-yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)-1,1-difluoropr opyl)piperidin-1-yl)acetic acid (190 mg, crude) as a yellow solid. MS (ESI) m/z: 812.4 [M+H] + Step J Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)-3,3-difluorop ropoxy)-2- methylphenyl)picolinate To a solution of 2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)-1,1- difluoropropyl)piperidin-1-yl)acetic acid (100 mg, 123 µmol, 1.0 equiv.) in DMF (1.00 mL) were added 3-(6-amino-1-methyl-1H-indazol-3-yl)piperidine-2,6-dione (35.0 mg, 135 µmol, 1.1 equiv.), HATU (56.2 mg, 148 µmol, 1.2 equiv.), and DIEA (47.8 mg, 369 µmol, 64.4 µL, 3.0 equiv.). The mixture was stirred at 20 °C for 1 hour. After completion, the reaction mixture was diluted with water (3 mL) and then extracted with CH 2 Cl 2 (3 mL × 2). The combined organic layers were washed with brine (3 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2- ((3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)-3,3 -difluoropropoxy)-2- methylphenyl)picolinate (100 mg, crude) as a brown solid. MS (ESI) m/z: 1052.8 [M+H] + Step K. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)-3,3-difluorop ropoxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-meth yl-1H-indazol-6-yl)amino)-2- oxoethyl)piperidin-4-yl)-3,3-difluoropropoxy)-2-methylphenyl )picolinate (100 mg, 95.0 µmol, 1.0 equiv.) in CH 2 Cl 2 (1.00 mL) was added TFA (3.08 g, 27.0 mmol, 2 mL, 284 equiv.). The mixture was stirred at 20 °C for 6 hours. After completion, the reaction mixture was concentrated under reduced pressure and the residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-(1-(2-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2-oxoet hyl)piperidin-4-yl)-3,3- difluoropropoxy)-2-methylphenyl)picolinic acid (23.9 mg, 23.8 µmol, 25.0% yield, 98.7% purity) as a white solid. MS (ESI) m/z: 996.3 [M+H] + 1 H NMR (400 MHz, DMSO-d6,) δ = 13.11 - 12.55 (m, 1H), 10.88 (s, 1H), 10.08 - 9.67 (m, 1H), 8.03 (d, J = 6.4 Hz, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.67 - 7.57 (m, 2H), 7.52 - 7.41 (m, 3H), 7.41 - 7.31 (m, 2H), 7.20 (d, J = 8.4 Hz, 1H), 7.16 - 7.07 (m, 1H), 7.01 - 6.89 (m, 2H), 6.66 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.38 - 4.27 (m, 1H), 4.16 (t, J = 6.0 Hz, 2H), 3.92 (s, 5H), 3.21 - 3.10 (m, 2H), 3.07 - 2.90 (m, 4H), 2.66 - 2.56 (m, 2H), 2.48 - 2.33 (m, 3H), 2.27 - 2.06 (m, 3H), 2.04 - 1.94 (m, 1H), 1.90 (s, 3H), 1.88 - 1.73 (m, 2H), 1.70 - 1.50 (m, 2H). Example 58. Preparation of Compound 235 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[4-[[4-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]piperazin-1-yl ]methyl]-1-piperidyl]butoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 1-bromo-3-(4-bromobutoxy)-2-methyl-benzene A mixture of 3-bromo-2-methyl-phenol (3.0 g, 16.04 mmol, 1.0 equiv.), 1,4-dibromobutane (10.39 g, 48.12 mmol, 5.80 mL, 3.0 equiv.), Cs 2 CO 3 (15.68 g, 48.12 mmol, 3.0 equiv.), and KI (7.99 g, 48.12 mmol, 3.0 equiv.) in CH 3 CN (30 mL) was stirred at 25 °C for 5 hours. The reaction mixture was concentrated under reduced pressure to give 1-bromo-3-(4- bromobutoxy)-2-methyl-benzene (3.3 g, 10.2 mmol, 63.9% yield) colorless oil. Step B. Procedure for preparation of [1-[4-(3-bromo-2-methyl-phenoxy) butyl]-4- piperidyl] methanol A mixture of 1-bromo-3-(4-bromobutoxy)-2-methyl-benzene (1.0 g, 3.11 mmol, 1.0 equiv.), 4- piperidylmethanol (565.03 mg, 3.73 mmol, 1.2 equiv., HCl), K2CO3 (1.29 g, 9.32 mmol, 3.0 equiv.), and KI (257.74 mg, 1.55 mmol, 0.5 equiv.) in CH3CN (30 mL) was stirred at 60 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by reverse-phase HPLC (0.1% NH 3 •H 2 O) to give [1-[4-(3-bromo-2- methyl-phenoxy) butyl]-4-piperidyl] methanol (666 mg, 1.8 mmol, 58.3% yield, 97% purity) as a primrose yellow solid. MS (ESI) m/z: 357.9 [M+H] + . Step C. Procedure for preparation of 1-[4-(3-bromo-2-methyl-phenoxy)butyl]piperidine- 4-carbaldehyde A solvent of DMSO (350.87 mg, 4.49 mmol, 350.87 µL, 4.0 equiv.) in DCM (4 mL) was added dropwise to a solution of oxalyl dichloride (284.99 mg, 2.25 mmol, 196.54 µL, 2.0 equiv.) in DCM (0.5 mL) at -70℃ under N 2 atmosphere. The mixture was stirred at -70 °C for 30 minutes. After which time [1-[4-(3-bromo-2-methyl-phenoxy) butyl]-4-piperidyl] methanol (400 mg, 1.12 mmol, 1.0 equiv.) in DCM (0.5 mL) was added dropwise at -70 °C. The solution was stirred at -70 °C for 30 minutes. Then TEA (681.61 mg, 6.74 mmol, 937.56 µL, 6.0 equiv.) was added into the solution. The solution was stirred at 25 °C for 1 hour under N 2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with DCM 60 (20 mL × 3). The combined organic layers were washed with brine (10 mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give 1-[4-(3-bromo-2-methyl- phenoxy)butyl]piperidine-4-carbaldehyde (300 mg, 846.7 µmol, 75.4% yield) as brown oil. MS (ESI) m/z: 355.1 [M+H] + . Step D. Procedure for preparation of 3-[6-[4-[[1-[4-(3-bromo-2-methyl-phenoxy)butyl]- 4-piperidyl]methyl]piperazin-1-yl]-1-methyl-indazol-3-yl]pip eridine-2,6-dione To a mixture of 1-[4-(3-bromo-2-methyl-phenoxy)butyl]piperidine-4-carbaldehy de (100 mg, 282.26 µmol, 1 equiv.), 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (110.89 mg, 338.71 µmol, 1.2 equiv.) and CH3COOH (169.50 ug, 2.82 µmol, 0.161 µL, 0.01 equiv.) in a solvent of DCM (1 mL) and t-BuOH (1.0 mL) was added NaBH(OAc)3 (179.47 mg, 846.79 µmol, 3.0 equiv.). Then the mixture was stirred at 25 °C for 2.5 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH = 10:1) to give 3-[6-[4-[[1-[4-(3-bromo-2-methyl- phenoxy)butyl]-4-piperidyl]methyl]piperazin-1-yl]-1-methyl-i ndazol-3-yl]piperidine-2,6- dione (80 mg, 81.5 µmol, 28.8% yield, 67.8% purity) as a white solid. MS (ESI) m/z: 667.2 [M+H] + . Step E. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[4-[[4-[3-(2,6-diox o-3-piperidyl)-1-methyl- indazol-6-yl]piperazin-1-yl]methyl]-1-piperidyl]butoxy]-2-me thyl-phenyl]pyridine-2- carboxylate A mixture of 3-[6-[4-[[1-[4-(3-bromo-2-methyl-phenoxy)butyl]-4-piperidyl] methyl]piperazin- 1-yl]-1-methyl-indazol-3-yl]piperidine-2,6-dione (80 mg, 120.18 µmol, 1.0 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-2-carboxylate (95.70 mg, 156.24 µmol, 1.3 equiv.), [2-(2-aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-buty l- phosphane;methanesulfonate (8.75 mg, 12.02 µmol, 0.1 equiv.), and KF (34.91 mg, 600.91 µmol, 14.08 µL, 5.0 equiv.) in a solvent of dioxane (2.0 mL) was stirred at 100 °C for 1 hour under microwave. The reaction mixture was concentrated under reduced pressure to give a residue, which was purified by prep-TLC (SiO2, DCM: MeOH = 10:1) to give tert-butyl 6-[8- (1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin- 2-yl]-3-[3-[4-[4-[[4-[3-(2,6- dioxo-3-piperidyl)-1-methyl-indazol-6-yl]piperazin-1-yl]meth yl]-1-piperidyl]butoxy]-2- methyl-phenyl]pyridine-2-carboxylate (40 mg, 27.3 µmol, 22.7% yield, 73.3% purity) as brown oil. MS (ESI) m/z: 1071.6 [M+H] + . Step F. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[4-[4-[[4-[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6- yl]piperazin-1-yl]methyl]-1-piperidyl]butoxy]-2-methyl-pheny l]pyridine-2-carboxylic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[4-[4-[[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indaz ol-6-yl]piperazin-1-yl]methyl]-1- piperidyl]butoxy]-2-methyl-phenyl]pyridine-2-carboxylate (30 mg, 28.00 µmol, 1.0 equiv.) in TFA (0.8 mL) and DCM (0.8 mL) was stirred at 25°C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue, which was purified by reverse-phase HPLC to give the compound 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[4-[4-[[4-[3-(2,6-dioxo-3-piperidyl)- 1-methyl-indazol-6-yl]piperazin- 1-yl]methyl]-1-piperidyl]butoxy]-2-methyl-phenyl]pyridine-2- carboxylic acid (22.9 mg, 21.4 µmol, 76.5% yield, 99.1% purity, FA) as a yellow solid. MS (ESI) m/z: 1015.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ= 10.84 (s, 1H), 8.14 (s, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.51 - 7.48 (m, 1H), 7.47 - 7.40 (m, 3H), 7.39 - 7.30 (m, 2H), 7.13 - 7.07 (m, 1H), 6.96 - 6.87 (m, 3H), 6.83 (s, 1H), 6.65 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.28 - 4.22 (m, 1H), 4.02 (s, 2H), 3.94 - 3.90 (m, 2H), 3.88 (s, 3H), 3.21 (d, J = 0.8 Hz, 4H), 3.04 - 3.00 (m, 3H), 2.77 - 2.70 (m, 2H), 2.65 - 2.57 (m, 4H), 2.54 (s, 2H), 2.31 - 2.26 (m, 1H), 2.23 - 2.11 (m, 4H), 1.92 (s, 3H), 1.86 - 1.61 (m, 9H), 1.30 - 1.19 (m, 2H). Example 59. Preparation of Compound 239a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1-yl)-2,2-difluoro-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of methyl 4-(3-bromo-2-methyl-phenoxy) cyclohexanecarboxylate To a solution of 3-bromo-2-methyl-phenol (30 g, 160.40 mmol, 1 equiv.) in toluene (300 mL) was added methyl 4-hydroxycyclohexanecarboxylate (27.91 g, 176.44 mmol, 1.1 equiv.) and 2-(tributyl-λ 5 -phosphanylidene) acetonitrile (46.46 g, 192.48 mmol, 1.2 equiv.) at 25 °C. The reaction mixture was degassed and purged with N 2 three times, and then the mixture was stirred at 120 °C for 16 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 1/0 to 10/1) to give methyl 4-(3-bromo-2-methyl- phenoxy) cyclohexanecarboxylate (49 g, 80.5 mmol, 50.2% yield, 53.8% purity) as a colorless oil. Step B. Procedure for preparation of [4-(3-bromo-2-methyl-phenoxy) cyclohexyl]methanol To a solution of methyl 4-(3-bromo-2-methyl-phenoxy) cyclohexanecarboxylate (49 g, 80.57 mmol, 53.8% purity, 1 equiv.) in THF (500 mL) was added LiAlH 4 (3.97 g, 104.73 mmol, 1.3 equiv.) dropwise at 0 °C under N 2 . The reaction mixture was stirred under N 2 at 0 °C for 2 hours. The reaction mixture was quenched with Na2SO4•10 H2O (2 g) and slowly warmed to 20 °C. The mixture was filtered and washed with DCM (500 mL). Then the organic layers were concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 20/1 to 5/1) to give [4-(3-bromo-2- methyl-phenoxy) cyclohexyl]methanol (15.7 g, 52.4 mmol, 65.1% yield) as a yellow oil. 1 H NMR (400 MHz, CDCl 3 -d) δ = 7.14 (d, J = 8.0 Hz, 1H), 6.98 (t, J = 8.0 Hz, 1H), 6.82 (d, J = 8.4 Hz, 1H), 4.12 - 4.07 (m, 1H), 3.51 (d, J = 6.4 Hz, 2H), 2.31 (s, 3H), 2.23 - 2.14 (m, 2H), 1.96 - 1.88 (m, 2H), 1.63 - 1.54 (m, 1H), 1.50 - 1.46 (m, 2H), 1.16 - 1.05 (m, 2H) Step C. Procedure for preparation of 4-(3-bromo-2-methyl-phenoxy) cyclohexanecarbaldehyde A solution of DMSO (16.40 g, 209.89 mmol, 16.40 mL, 4 equiv.) in DCM (10 mL) was added dropwise to a solution of oxalyl dichloride (13.32 g, 104.95 mmol, 9.19 mL, 2 equiv.) in DCM (130 mL) at -70 °C under N2 atmosphere. The mixture was stirred at -70 °C for 30 minutes. After which time [4-(3-bromo-2-methyl-phenoxy) cyclohexyl]methanol (15.7 g, 52.47 mmol, 1 equiv.) in DCM (10 mL) was added dropwise at -70 °C. The solution was stirred for 30 minutes at -70 °C. Then TEA (31.86 g, 314.84 mmol, 43.82 mL, 6 equiv.) was added into the solution. The solution was stirred at 25 °C for 1 hour under N2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with DCM (20 mL × 3). The combined organic layers were washed with brine (10 mL × 2), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give 4-(3-bromo-2-methyl-phenoxy) cyclohexanecarbaldehyde (22 g, crude) as a yellow oil. Step D. Procedure for preparation of ethyl 3-[4-(3-bromo-2-methyl-phenoxy) cyclohexyl]-2, 2-difluoro-3-hydroxy-propanoate To a mixture of Zn (2.50 g, 38.28 mmol, 2.5 equiv.) in THF (5 mL) was added 1, 2- dibromoethane (287.62 mg, 1.53 mmol, 115.51 μL, 0.1 equiv.), ethyl 2-bromo-2, 2-difluoro- acetate (4.35 g, 21.43 mmol, 2.75 mL, 1.4 equiv.) at 60 °C. The reaction mixture was degassed and purged with N 2 three times, and then the mixture was stirred at 60 °C for 0.5 hours under N 2 atmosphere. Then a solution of 4-(3-bromo-2-methyl-phenoxy) cyclohexanecarbaldehyde (1.5 g, 5.05 mmol, 0.33 equiv.) in THF (1 mL) was added dropwise at 60 °C. The mixture was stirred at 60 °C for 16 hours. The mixture was cooled to 20 °C and then filtered. The filtrate was quenched by 1N HCl (100 mL), and then extracted with EtOAc (100 mL × 3). The combined organic layers were washed with brine saturated NaHCO3 (300 mL) and brine (100 mL × 1), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 10% ethyl acetate/petroleum ether) to give ethyl 3-[4-(3-bromo-2-methyl-phenoxy) cyclohexyl]-2, 2- difluoro-3-hydroxy-propanoate (670 mg, 1.5 mmol, 10.3% yield) as a colorless oil. 1 H NMR (400 MHz, CDCl3-d) δ = 7.15 (d, J = 8.0 Hz, 1H), 6.98 (t, J = 8.0 Hz, 1H), 6.81 (d, J = 8.0 Hz, 1H), 4.38 (q, J = 7.2 Hz, 2H), 4.15 - 4.07 (m, 1H), 3.95 - 3.85 (m, 1H), 2.30 (s, 3H), 2.25 - 2.17 (m, 2H), 2.13 - 2.05 (m, 2H), 1.94 - 1.87 (m, 1H), 1.82 - 1.73 (m, 1H), 1.52 - 1.43 (m, 3H), 1.39 (t, J = 7.2 Hz, 3H) Step E. Procedure for preparation of ethyl 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]- 2,2-difluoro-3-methylsulfanylcarbothioyloxy-propanoate To a solution of ethyl 3-[4-(3-bromo-2-methyl-phenoxy) cyclohexyl]-2, 2-difluoro-3-hydroxy- propanoate (670 mg, 1.59 mmol, 1 equiv.) in DMF (8.5 mL) was added DBU (968.50 mg, 6.36 mmol, 958.91 μL, 4 equiv.) and CS2 (1.21 g, 15.90 mmol, 961.07 μL, 10 equiv.) at 20 °C. The mixture was stirred at 20 °C for 1 hour under N2 atmosphere. Then MeI (2.26 g, 15.90 mmol, 990.11 μL, 10 equiv.) was added dropwise at 20 °C and the mixture was stirred at 20 °C for 1 hour under N 2 atmosphere. The mixture was diluted with water 10 mL and extracted with EtOAc (20 mL × 3). The combined organic layers were washed with brine (10 mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 10% ethyl acetate/petroleum ether) to give ethyl 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-2,2-difluoro-3- methylsulfanylcarbothioyloxy-propanoate (764 mg, 1.4 mmol, 93.9% yield) as a colorless oil. 1 H NMR (400 MHz, CDCl 3 -d) δ = 7.15 (d, J = 7.6 Hz, 1H), 6.97 (t, J = 8.0 Hz, 1H), 6.79 (d, J = 8.4 Hz, 1H), 6.31 - 6.22 (m, 1H), 4.30 (q, J = 7.2 Hz, 2H), 4.15 - 4.04 (m, 1H), 2.63 - 2.56 (m, 3H), 2.29 (s, 3H), 2.23 - 2.17 (m, 2H), 2.16 - 2.08 (m, 1H), 2.04 - 1.95 (m, 2H), 1.52 - 1.37 (m, 4H), 1.34 (t, J = 7.2 Hz, 3H) Step F. Procedure for preparation of ethyl 3-[4-(3-bromo-2-methyl-phenoxy) cyclohexyl]- 2, 2-difluoro-propanoate A solution of ethyl 3-[4-(3-bromo-2-methyl-phenoxy) cyclohexyl]-2, 2-difluoro-3- methylsulfanyl carbothioyloxy-propanoate (764 mg, 1.49 mmol, 1 equiv.), 2-tert-butylperoxy- 2-methyl-propane (240.28 mg, 1.64 mmol, 302.62 μL, 1.1 equiv.) and phenylphosphonoylbenzene (664.48 mg, 3.29 mmol, 2.2 equiv.) in dioxane (8 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 100 °C for 24 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 10% ethyl acetate/petroleum ether) to give ethyl 3-[4-(3-bromo-2-methyl-phenoxy) cyclohexyl]-2, 2-difluoro-propanoate (580 mg, 1.4 mmol, 95.8% yield) as a colorless oil. 1 H NMR (400 MHz, CDCl3-d) δ = 7.15 (d, J = 7.6 Hz, 1H), 6.97 (t, J = 8.0 Hz, 1H), 6.80 (d, J = 8.0 Hz, 1H), 4.34 (q, J = 7.2 Hz, 2H), 4.14 - 4.05 (m, 1H), 2.30 (s, 3H), 2.18 - 2.10 (m, 2H), 2.08 - 1.98 (m, 2H), 1.95 (d, J = 12.8 Hz, 2H), 1.77 - 1.64 (m, 1H), 1.54 - 1.45 (m, 2H), 1.37 (t, J = 7.2 Hz, 3H), 1.23 - 1.10 (m, 2H) Step G. Procedure for preparation of 3-[4-(3-bromo-2-methyl-phenoxy) cyclohexyl]-2, 2- difluoro-propanoic acid To a solution of ethyl 3-[4-(3-bromo-2-methyl-phenoxy) cyclohexyl]-2, 2-difluoro-propanoate (550 mg, 1.36 mmol, 1 equiv.) in THF (5 mL) and H 2 O (0.5 mL) was added LiOH•H 2 O (170.85 mg, 4.07 mmol, 3 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was treated with water (4 mL). The pH was adjusted to around 3 by progressively adding diluted HCl. The mixture was filtered to give 3-[4-(3-bromo-2-methyl-phenoxy) cyclohexyl]-2, 2-difluoro- propanoic acid (510 mg, 1.30 mmol, 95.94% yield, 96.3% purity) as a white solid. 1 H NMR (400 MHz, CDCl 3 -d) δ = 7.09 (d, J = 8.0 Hz, 1H), 6.86 (t, J = 8.0 Hz, 1H), 6.68 (d, J = 8.4 Hz, 1H), 4.02 - 3.90 (m, 1H), 2.22 (s, 3H), 2.05 - 1.98 (m, 2H), 1.96 - 1.91 (m, 2H), 1.87 - 1.84 (m, 2H), 1.71 - 1.60 (m, 1H), 1.43 - 1.34 (m, 2H), 1.10 - 0.96 (m, 2H) Step H. Procedure for preparation of 3-[6-[4-[3-[4-(3-bromo-2-methyl-phenoxy) cyclohexyl]-2, 2-difluoro-propanoyl]piperazin-1-yl]-1-methyl-indazol-3-yl]p iperidine-2, 6-dione To a solution of 3-[4-(3-bromo-2-methyl-phenoxy) cyclohexyl]-2, 2-difluoro-propanoic acid (80 mg, 212.08 μmol, 1 equiv.) in DMF (1 mL) was added HATU (104.83 mg, 275.70 μmol, 1.3 equiv.), DIPEA (82.23 mg, 636.23 μmol, 110.82 μL, 3 equiv.), and 3-(1-methyl-6- piperazin-1-yl-indazol-3-yl) piperidine-2, 6-dione (90.26 mg, 248.07 μmol, 1.17 equiv., HCl) at 25 °C. The reaction mixture was stirred at 40 °C for 16 hours. The reaction mixture was diluted with water (5 mL). The mixture was triturated with water (5 mL) at 25 °C for 10 min. The mixture was filtered to give 3-[6-[4-[3-[4-(3-bromo-2-methyl-phenoxy) cyclohexyl]-2, 2- difluoro-propanoyl]piperazin-1-yl]-1-methyl-indazol-3-yl]pip eridine-2, 6-dione (110 mg, 155.2 μmol, 73.2% yield, 96.9% purity) as a yellow solid. MS (ESI) m/z: 688.6 [M+H] + . Step I. Procedure for preparation of tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[4-[3-(2, 6-dioxo-3-piperidyl) -1-methyl- indazol-6-yl]piperazin-1-yl]-2, 2-difluoro-3-oxo-propyl]cyclohexoxy]-2-methyl- phenyl]pyridine-2-carboxylate 3-[6-[4-[3-[4-(3-bromo-2-methyl-phenoxy) cyclohexyl]-2, 2-difluoro-propanoyl]piperazin-1- yl]-1-methyl-indazol-3-yl]piperidine-2, 6-dione (110 mg, 160.21 μmol, 1 equiv.), tert-butyl 6- [8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-3-(4, 4, 5, 5- tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridine-2-carboxylate (127.58 mg, 208.28 μmol, 1.3 equiv.), KF (27.92 mg, 480.64 μmol, 11.26 μL, 3 equiv.), and [2-(2-aminophenyl) phenyl]palladium (1+) ;bis (1-adamantyl) -butyl-phosphane; methanesulfonate (23.34 mg, 32.04 μmol, 0.2 equiv.) were taken up into a microwave tube in dioxane (1 mL) and H2O (0.1 mL). The sealed tube was heated at 100 °C for 60 min under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , DCM: MeOH = 10:1) to give tert-butyl 6-[8-(1, 3-benzothiazol- 2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[4-[3-(2, 6-dioxo-3-piperidyl) - 1-methyl-indazol-6-yl]piperazin-1-yl]-2, 2-difluoro-3-oxo-propyl]cyclohexoxy]-2-methyl- phenyl]pyridine-2-carboxylate (95 mg, 75.8 μmol, 47.1% yield, 86.9% purity) as a white solid. MS (ESI) m/z: 1092.9 [M+H] + . Step J. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-2,2-difluoro-3-oxopro pyl)cyclohexyl)oxy)-2- methylphenyl)picolinic acid solution of tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2- yl]-3-[3-[4-[3-[4-[3-(2, 6-dioxo-3-piperidyl) -1-methyl-indazol-6-yl]piperazin-1-yl]-2, 2- difluoro-3-oxo-propyl]cyclohexoxy]-2-methyl-phenyl]pyridine- 2-carboxylate (95 mg, 86.98 μmol, 1 equiv.) in DCM (0.5 mL) and TFA (0.5 mL) was stirred at 25 °C for 40 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1-yl)-2,2-difluoro-3-oxopropyl)cyc lohexyl)oxy)-2- methylphenyl)picolinic acid (37.8 mg, 35.6 μmol, 40.9% yield, 97.4% purity) as an off-white solid. MS (ESI) m/z: 1036.9 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.95 - 12.29 (m, 2H), 10.85 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.54 (d, J = 8.8 Hz, 1H), 7.49 - 7.42 (m, 3H), 7.40 - 7.32 (m, 2H), 7.10 - 7.04 (m, 1H), 6.98 - 6.89 (m, 4H), 6.61 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.27 (dd, J = 5.2, 9.2 Hz, 1H), 4.23 - 4.14 (m, 1H), 3.90 (s, 5H), 3.85 - 3.79 (m, 2H), 3.76 - 3.69 (m, 2H), 3.28 - 3.28 (m, 2H), 3.02 (t, J = 5.2 Hz, 2H), 2.68 - 2.58 (m, 2H), 2.35 - 2.27 (m, 2H), 2.21 - 1.99 (m, 6H), 1.86 (s, 5H), 1.73 - 1.64 (m, 1H), 1.46 - 1.33 (m, 2H), 1.30 - 1.20 (m, 2H) Example 60. Preparation of Compound 240b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((3S)-1-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino )-2-oxoethyl)pyrrolidin-3- yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of (S)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-methoxy-2-oxo ethyl)pyrrolidin-3- yl)propoxy)-2-methylphenyl)picolinate (S)-ethyl 2-(3-(3-(3-bromo-2-methylphenoxy)propyl)pyrrolidin-1-yl)acet ate (126 mg, 327.86 µmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pic olinate (220.91 mg, 360.65 µmol, 1.1 equiv.), Ad 2 nBuP Pd G 3 (47.75 mg, 65.57 µmol, 0.2 equiv.), and K 2 CO 3 (1.5 M, 327.86 µL, 1.5 equiv.) in dioxane (3 mL) and were taken up into a microwave tube in dioxane (3 mL). The sealed tube was heated at 100 °C for 2 hours under microwave. The reaction mixture was partitioned between H2O (15 mL) and ethyl acetate (20 mL × 3). The organic phase was separated, washed with aqueous NaCl (15 mL × 3), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, petroleum ether : ethyl acetate = 0 : 1) to give (S)-tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2- methoxy-2- oxoethyl)pyrrolidin-3-yl)propoxy)-2-methylphenyl)picolinate (220 mg, 238.1 µmol, 72.6% yield, 85.5% purity) as a yellow solid MS (ESI) m/z: 790.5 [M+H] + . Step B. Procedure for preparation of (S)-2-(3-(3-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)propyl)pyrrolidin-1-yl)acetic acid A mixture of (S)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-methoxy-2-oxoethyl)pyrrolidin-3-yl)p ropoxy)-2- methylphenyl)picolinate (220 mg, 278.49 µmol, 1 equiv.), and LiOH.H 2 O (1 M, 835.46 µL, 3 equiv.) in THF (6 mL) was stirred at 25 °C for 15 hours. The mixture was concentrated and diluted with H2O (2 mL), then the pH of the mixture was adjusted to 3 with 1 M HCl. Then the mixture was filtered and concentrated to give (S)-2-(3-(3-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)propyl)pyrrolidin-1-yl)acetic acid (130 mg, 162.2 µmol, 58.2% yield, 95.1% purity) as a white solid. MS (ESI) m/z: 762.1 [M+H] + . Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3S)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-2-oxoethyl)pyrrolidin-3-yl)propoxy)-2 -methylphenyl)picolinate A mixture of (S)-2-(3-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)pyrrolidin-1- yl)acetic acid (130 mg, 170.62 µmol, 1 equiv.), 3-(7-amino-1-methyl-1H-indazol-3- yl)piperidine-2,6-dione (52.88 mg, 204.74 µmol, 1.2 equiv.) in pyridine (2 mL) was added EDCI (49.06 mg, 255.93 µmol, 1.5 equiv.), and then the mixture was stirred at 25 °C for 3 hours. The reaction mixture was partitioned between H2O (10 mL) and DCM (10 mL × 3). The organic phase was separated, washed with aqueous NaCl (10 mL × 2), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3S)- 1-(2-((3-(2,6-dioxopiperidin-3- yl)-1-methyl-1H-indazol-7-yl)amino)-2-oxoethyl)pyrrolidin-3- yl)propoxy)-2- methylphenyl)picolinate (130 mg, crude) as a yellow solid. MS (ESI) m/z: 1002.4 [M+H] + . Step D. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3S)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-2-oxoethyl)pyrrolidin-3-yl)propoxy)-2 -methylphenyl)picolinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-((3S)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-7-yl)amino)-2- oxoethyl)pyrrolidin-3-yl)propoxy)-2-methylphenyl)picolinate (126.15 mg, 125.88 µmol, 1 equiv.) in DCM (3 mL) and TFA (1 mL) was stirred at 40 °C for 39 hours. The reaction mixture was concentrated under the reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)- 3-(3-(3-((3S)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H -indazol-7-yl)amino)-2- oxoethyl)pyrrolidin-3-yl)propoxy)-2-methylphenyl)picolinic acid (24.9 mg, 25.3 µmol, 20.1% yield, 96.1% purity) as a yellow solid MS (ESI) m/z: 946.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.89 (s, 1H), 9.88 (s, 1H), 8.16 (s, 1H), 8.02 (d, J = 7.2 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 8.0 Hz, 1H), 7.56 (d, J = 8.0 Hz, 1H), 7.49 - 7.41 (m, 3H), 7.39 - 7.32 (m, 2H), 7.28 (d, J = 7.2 Hz, 1H), 7.07 (q, J = 8.0 Hz, 2H), 6.96 - 6.92 (m, 1H), 6.88 (d, J = 8.0 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.36 (dd, J = 5.2, 10 Hz, 1H), 4.08 (s, 3H), 3.98 (s, 2H), 3.91 (t, J = 5.6 Hz, 2H), 3.02 (s, 2H), 2.99 - 2.93 (m, 2H), 2.86 - 2.76 (m, 2H), 2.62 - 2.58 (m, 2H), 2.26 - 2.11 (m, 3H), 2.01 (dd, J = 1.6, 5.6 Hz, 2H), 1.90 (s, 3H), 1.78 - 1.71 (m, 2H), 1.55 -1.40 (m, 4H) Example 61. Preparation of Compound 241 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[2-[4-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]piperazin-1-yl ]ethyl]phenoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 1-bromo-3-(4-(2-chloroethyl)phenoxy)-2- methylbenzene To a solution of 2-(4-(3-bromo-2-methylphenoxy)phenyl)ethan-1-ol (100 mg, 325.54 µmol, 1 equiv.) in SOCl2 (3 mL). The mixture was stirred at 80 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a product 1-bromo-3-(4-(2- chloroethyl)phenoxy)-2-methylbenzene (100 mg, crude) was obtained as a yellow oil. Step B. Procedure for preparation of 3-(7-(4-(4-(3-bromo-2- methylphenoxy)phenethyl)piperazin-1-yl)-1-methyl-1H-indazol- 3-yl)piperidine-2,6- dione To a solution of 1-bromo-3-(4-(2-chloroethyl)phenoxy)-2-methylbenzene (100 mg, 307.10 µmol, 1 equiv.), 3-(1-methyl-7-(piperazin-1-yl)-1H-indazol-3-yl)piperidine-2, 6-dione (100.54 mg, 307.10 µmol, 1 equiv.) in NMP (1 mL) was added KI (50.98 mg, 307.10 µmol, 1 equiv.) and DIEA (198.45 mg, 1.54 mmol, 267.46 µL, 5 equiv.). The mixture was stirred at 60 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , by prep-TLC (SiO 2 , DCM: MeOH = 20:1) to give 3-(7-(4-(4-(3-bromo-2-methylphenoxy)phenethyl)piperazin-1-yl )-1-methyl-1H- indazol-3-yl)piperidine-2,6-dione (65 mg, 82.2 µmol, 26.8% yield, 78% purity) as a yellow oil. MS (ESI) m/z: 618.1 [M+H] +. Step C. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[2-[4-[3-(2,6-dioxo -3-piperidyl)-1-methyl- indazol-7-yl]piperazin-1-yl]ethyl]phenoxy]-2-methyl-phenyl]p yridine-2-carboxylate 3-(7-(4-(4-(3-bromo-2-methylphenoxy)phenethyl)piperazin-1-yl )-1-methyl-1H-indazol-3- yl)piperidine-2,6-dione (60 mg, 97.32 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tet ramethyl-1,3,2-dioxaborolan-2- yl)pyridine-2-carboxylate (65.57 mg, 107.05 µmol, 1.1 equiv.) and [2-(2- aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-butyl-phos phane;methanesulfonate (7.09 mg, 9.73 µmol, 0.1 equiv.), and KF (16.96 mg, 291.95 µmol, 6.84 µL, 3 equiv.) were taken up into a microwave tube in dioxane (2 mL) and H 2 O (0.2 mL). The sealed tube was heated at 100 °C for 1 hour under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 ,by prep-TLC (SiO 2 , DCM: MeOH = 20:1) to give tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[2-[4- [3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-7-yl]piperazin-1-yl]ethyl]phenoxy]-2-methyl-p henyl]pyridine-2-carboxylate (55 mg, 45.5 µmol, 46.7% yield, 84.6% purity) as a white oil. MS (ESI) m/z: 1022.3 [M+H] + . Step D. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[4-[2-[4-[3-(2,6-dioxo-3-piperidyl )-1-methyl-indazol-7- yl]piperazin-1-yl]ethyl]phenoxy]-2-methyl-phenyl]pyridine-2- carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[4-[2-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-inda zol-7-yl]piperazin-1- yl]ethyl]phenoxy]-2-methyl-phenyl]pyridine-2-carboxylate (50 mg, 48.91 µmol, 1 equiv.) in DCM (0.5 mL) was added TFA (770.00 mg, 6.75 mmol, 0.5 mL, 138.06 equiv.). The mixture was stirred at 25 °C for 3 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[2-[4- [3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-7-yl]piperazin-1-yl]ethyl]phenoxy]-2-methyl-p henyl]pyridine-2-carboxylic acid (4.1 mg, 4.1 µmol, 8.4% yield, 95% purity) as a yellow solid. MS (ESI) m/z: 966.6[M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.94 - 10.82 (m, 1H), 8.29 (s, 1H), 8.00 (d, J = 7.6 Hz, 1H), 7.76 (d, J = 8.0 Hz, 1H), 7.64 (d, J = 6.8 Hz, 1H), 7.43 (d, J = 6.4 Hz, 2H), 7.39 - 7.36 (m, 1H), 7.36 - 7.29 (m, 2H), 7.22 (d, J = 8.4 Hz, 2H), 7.16 - 7.10 (m, 1H), 7.06 - 7.00 (m, 2H), 6.94 (d, J = 8.0 Hz, 1H), 6.87 - 6.78 (m, 4H), 4.99 (s, 2H), 4.34 (dd, J = 5.2, 9.6 Hz, 1H), 4.25 (s, 3H), 3.91 (t, J = 5.6 Hz, 2H), 3.04 - 2.99 (m, 4H), 2.78 - 2.74 (m, 2H), 2.67 (d, J = 1.6 Hz, 1H), 2.63 - 2.57 (m, 4H), 2.52 (s, 2H), 2.43 (s, 1H), 2.36 - 2.30 (m, 2H), 2.20 - 2.14 (m, 1H), 1.94 (s, 3H) Example 62. Preparation of Compound 245a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((2R)-4-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino )-2-oxoethyl)-2- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linic acid Step A. Procedure for preparation of tert-butyl (R)-4-allyl-3-(trifluoromethyl)piperazine- 1-carboxylate To a solution of tert-butyl (R)-3-(trifluoromethyl)piperazine-1-carboxylate (500 mg, 1.97 mmol, 1.0 equiv.) in CH 3 CN (7 mL) was added K 2 CO 3 (815.38 mg, 5.90 mmol, 3.0 equiv.), KI (32.65 mg, 196.66 µmol, 0.1 equiv.), and 3-bromoprop-1-ene (237.91 mg, 1.97 mmol, 1.0 equiv.). The mixture was stirred at 80 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=1/0 to 50/1) to give tert-butyl (R)-4-allyl- 3-(trifluoromethyl)piperazine-1-carboxylate (400 mg, crude) as a yellow oil. 1 H NMR (400 MHz, CDCl3) δ = 5.95 - 5.64 (m, 1H), 5.41 - 5.07 (m, 2H), 4.23 - 3.55 (m, 2H), 3.50 - 3.28 (m, 3H), 3.24 - 3.08 (m, 2H), 2.91 (t, J = 10.8 Hz, 1H), 2.65 (d, J = 10.8 Hz, 1H), 1.46 (s, 9H). Step B. Procedure for preparation of tert-butyl (R)-4-(3-hydroxypropyl)-3- (trifluoromethyl) piperazine-1-carboxylate To a mixture of tert-butyl (R)-4-allyl-3-(trifluoromethyl)piperazine-1-carboxylate (400 mg, 1.36 mmol, 1 equiv.) in THF (5 mL) was added BH 3 •THF (1 M, 4.08 mL, 3.0 equiv.). The mixture was degassed and purged with N2 three times, and then the mixture was stirred at 0 °C for 2 hours. Afterwards, NaOH (2 M, 1.36 mL, 2.0 equiv.) was added slowly, and the reaction was stirred for 2 hours under N 2 atmosphere. Then H 2 O 2 (924.58 mg, 8.15 mmol, 783.55 µL, 30% purity, 6.0 equiv.) was added to the mixture at 0 °C, and the mixture was stirred at 25 °C for 3 hours. The reaction mixture was quenched by addition Na2SO3 (saturated solution, 80 mL) at 25 °C, and the pH of the solution was adjusted to 5-7. The resulting mixture was extracted with DCM (20 mL × 3). The combined organic layers were washed with brine (10 mL × 3), dried over Na2SO4, and then concentrated (below 30 °C) under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate 1/0 to 2/1) to give tert-butyl (R)-4-(3-hydroxypropyl)-3-(trifluoromethyl) piperazine-1-carboxylate (180 mg, crude) as a colorless oil. 1 H NMR (400 MHz, CDCl3) δ = 4.25 - 3.88 (m, 2H), 3.78 (t, J = 5.6 Hz, 2H), 3.34 (s, 1H), 3.26 - 3.05 (m, 2H), 3.01 - 2.93 (m, 3H), 2.77 - 2.64 (m, 1H), 1.80 - 1.74 (m, 2H), 1.46 (s, 9H). Step C. Procedure for preparation of tert-butyl (R)-4-(3-(3-bromo-2- methylphenoxy)propyl)-3-(trifluoromethyl)piperazine-1-carbox ylate A mixture of tert-butyl (R)-4-(3-hydroxypropyl)-3-(trifluoromethyl)piperazine-1-carb oxylate (180 mg, 576.32 µmol, 1.0 equiv.), 3-bromo-2-methylphenol (107.79 mg, 576.32 µmol, 1.0 equiv.), and 2-(tributyl-λ5-phosphanylidene)acetonitrile (166.92 mg, 691.58 µmol, 1.2 equiv.) in toluene (4 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 120 °C for 4 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give tert-butyl (R)-4-(3-(3-bromo-2-methylphenoxy)propyl)-3-(trifluoromethyl )piperazine- 1-carboxylate (110 mg, crude) as a yellow oil. MS (ESI) m/z: 481.1 [M+H] + . 1 H NMR (400 MHz, CDCl3) δ = 7.16 (d, J = 8.0 Hz, 1H), 7.00 (t, J = 8.0 Hz, 1H), 6.77 (d, J = 8.0 Hz, 1H), 4.13 - 3.91 (m, 3H), 3.43 - 3.23 (m, 1H), 3.17 - 3.04 (m, 2H), 3.02 - 2.90 (m, 3H), 2.66 (d, J = 10.8 Hz, 1H), 2.31 (s, 3H), 2.03 - 1.93 (m, 2H), 1.58 (s, 1H), 1.46 (s, 9H). Step D. Procedure for preparation of (R)-1-(3-(3-bromo-2-methylphenoxy)propyl)-2- (trifluoromethyl)piperazine A solution of tert-butyl (R)-4-(3-(3-bromo-2-methylphenoxy)propyl)-3- (trifluoromethyl)piperazine-1-carboxylate (110 mg, 228.53 µmol, 1.0 equiv.) in HCl/dioxane (4 M, 1 mL, 17.50 equiv.) was stirred at 25 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give (R)-1-(3-(3-bromo-2-methylphenoxy)propyl)- 2-(trifluoromethyl)piperazine (110 mg, crude) as a white solid. MS (ESI) m/z: 381.0 [M+H] + . Step E. Procedure for preparation of ethyl (R)-2-(4-(3-(3-bromo-2- methylphenoxy)propyl)-3-(trifluoromethyl)piperazin-1-yl)acet ate To a solution of (R)-1-(3-(3-bromo-2-methylphenoxy)propyl)-2-(trifluoromethyl )piperazine (110 mg, 288.54 µmol, 1.0 equiv.) in CH3CN (2 mL) was added K2CO3 (199.40 mg, 1.44 mmol, 5.0 equiv.), KI (4.79 mg, 28.85 µmol, 0.1 equiv.), and ethyl 2-bromoacetate (48.19 mg, 288.54 µmol, 31.91 µL, 1.0 equiv.). The mixture was stirred at 60 °C for 4 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give ethyl (R)-2-(4-(3-(3-bromo-2-methylphenoxy)propyl)-3- (trifluoromethyl)piperazin-1-yl)acetate (120 mg, crude) as a black brown oil. MS (ESI) m/z: 467.0 [M+H] + Step F. Procedure for preparation of tert-butyl (R)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(4-(2-ethoxy-2-oxoe thyl)-2- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linate To a mixture of tert-butyl 6-(8-((3H-indol-2-yl)carbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)- 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)picolinate (157.29 mg, 256.78 µmol, 1.0 equiv.) and ethyl (R)-2-(4-(3-(3-bromo-2-methylphenoxy)propyl)-3- (trifluoromethyl)piperazin-1-yl)acetate (120 mg, 256.78 µmol, 1.0 equiv.) in dioxane (3 mL) was added KF (1.5 M, 513.57 µL, 3.0 equiv.) and Ad 2 nBuP Pd G 3 (18.70 mg, 25.68 µmol, 0.1 equiv.). After addition, the mixture was degassed and purged with N 2 three times, and then the mixture was stirred at 100 °C for 2 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~40% ethyl acetate/petroleum ether) to give compound tert-butyl (R)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquino lin- 2(1H)-yl)-3-(3-(3-(4-(2-ethoxy-2-oxoethyl)-2-(trifluoromethy l)piperazin-1-yl)propoxy)-2- methylphenyl)picolinate (200 mg, crude) as a yellow solid. MS (ESI) m/z: 873.6 [M+H] + . Step G. Procedure for preparation of (R)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)propyl)-3-(trifluoromethyl)piperazin-1-yl)acet ic acid To a solution of tert-butyl (R)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquino lin- 2(1H)-yl)-3-(3-(3-(4-(2-ethoxy-2-oxoethyl)-2-(trifluoromethy l)piperazin-1-yl)propoxy)-2- methylphenyl)picolinate (200 mg, 229.10 µmol, 1.0 equiv.) in THF (2 mL) was added LiOH•H2O (28.84 mg, 687.29 µmol, 3.0 equiv.) and H2O (0.6 mL). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove THF. The aqueous phase was adjusted to pH=4~5 with 1M HCl. The reaction mixture was filtered and diluted with ethyl acetate. The combined organic layers were dried over Na2SO4, filtered, and concentrated under reduced pressure to give (R)-2-(4-(3-(3-(6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)propyl)-3-(trif luoromethyl)piperazin-1- yl)acetic acid (180 mg, crude) as a white solid. MS (ESI) m/z: 845.6 [M+H] + . Step H. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((2R)-4-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-2-(trifluoromethyl)pipera zin-1-yl)propoxy)-2- methylphenyl)picolinate To a solution of (R)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)-3- (trifluoromethyl)piperazin-1-yl)acetic acid (90 mg, 106.52 µmol, 1.0 equiv.), 3-(6-amino-1- methyl-1H-indazol-3-yl)piperidine-2,6-dione (27.51 mg, 106.52 µmol, 1.0 equiv.) in pyridine (1.5 mL) was added EDCI (30.63 mg, 159.77 µmol, 1.5 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was diluted with water (2 mL) and extracted with ethyl acetate (2 mL × 5). The combined organic layers were washed with brine (5 mL × 2), dried over Na2SO4, filtered and concentrated under reduced pressure to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-((2R)-4-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2- oxoethyl)-2- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linate (100 mg, crude) as a white solid. MS (ESI) m/z: 1085.4 [M+H] + . Step I. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((2R)-4-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-2-(trifluoromethyl)pipera zin-1-yl)propoxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((2R)-4-(2-((3-(2,6-dioxopiperidin-3-yl)-1 -methyl-1H-indazol-6-yl)amino)- 2-oxoethyl)-2-(trifluoromethyl)piperazin-1-yl)propoxy)-2-met hylphenyl)picolinate (100 mg, 92.15 µmol, 1.0 equiv.) in DCM (1 mL) was added TFA (770.00 mg, 6.75 mmol, 0.5 mL, 73.28 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3- ((2R)-4-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)amino)-2-oxoethyl)-2- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linic acid (56.5 mg, 54.5 µmol, 59.1% yield, 99.3% purity) as a white solid. MS (ESI) m/z: 1029.3 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.98 - 12.33 (m, 2H), 10.88 (s, 1H), 9.96 - 9.63 (m, 1H), 8.13 (s, 1H), 8.08 - 7.99 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.69 - 7.59 (m, 2H), 7.51 - 7.41 (m, 3H), 7.40 - 7.30 (m, 2H), 7.14 - 7.01 (m, 2H), 6.97 (d, J = 8.8 Hz, 1H), 6.88 (d, J = 8.4 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.32 (dd, J = 5.2, 4.8 Hz, 1H), 4.01 (t, J = 6.0 Hz, 2H), 3.95 - 3.86 (m, 5H), 3.60 - 3.46 (m, 2H), 3.23 - 3.15 (m, 1H), 3.03 (t, J = 5.6 Hz, 3H), 2.94 - 2.83 (m, 3H), 2.77 - 2.57 (m, 6H), 2.40 - 2.28 (m, 1H), 2.23 - 2.10 (m, 1H), 1.99 - 1.82 (m, 5H). Example 63. Preparation of Compound 245b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((2S)-4-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino )-2-oxoethyl)-2- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linic acid Step A. Procedure for preparation of (S)-tert-butyl 4-allyl-3-(trifluoromethyl)piperazine- 1-carboxylate A mixture of (S)-tert-butyl 3-(trifluoromethyl)piperazine-1-carboxylate (400 mg, 1.57 mmol, 1 equiv.), 3-bromoprop-1-ene (209.36 mg, 1.73 mmol, 1.10 equiv.), KI (26.12 mg, 157.33 µmol, 0.10 equiv.), and K 2 CO 3 (652.30 mg, 4.72 mmol, 3.00 equiv.) in CH 3 CN (5 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 80 °C for 6 hours under N2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (25 mL× 3). The combined organic layers were washed with brine (15 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate 300/1 to 10/1) to give (S)-tert-butyl 4-allyl-3-(trifluoromethyl)piperazine-1-carboxylate (280 mg, 951.3 µmol, 60.4% yield) as a colorless oil. 1 H NMR (400 MHz,CDCl 3 ) δ = 5.91 - 5.68 (m, 1H), 5.32 - 5.16 (m, 2H), 4.17 - 3.84 (m, 2H), 3.44 - 3.30 (m, 3H), 3.26 - 3.05 (m, 2H), 2.98 - 2.86 (m, 1H), 2.68 - 2.57 (m, 1H), 1.46 (s, 9H) Step B. Procedure for preparation of (S)-tert-butyl 4-(3-hydroxypropyl)-3- (trifluoromethyl)piperazine-1-carboxylate To a mixture of (S)-tert-butyl 4-allyl-3-(trifluoromethyl)piperazine-1-carboxylate (150 mg, 509.66 µmol, 1.00 equiv.) in THF (1 mL) was added BH3•THF (1 M, 1.53 mL, 3.00 equiv.). The mixture was degassed and purged with N2 three times, and then the mixture was stirred at 0 °C for 1.5 hours under N 2 atmosphere. Then NaOH (2 M, 509.66 µL, 2.00 equiv.) was slowly added, and the mixture was stirred for 2 hours under N2 atmosphere. After stirring, H2O2 (346.72 mg, 3.06 mmol, 293.83 µL, 30% purity, 6.00 equiv.) was slowly added into the mixture. The mixture was stirred at 25 °C for 3 hours. The reaction mixture was quenched by addition of Na2SO3 (saturated solution, 50 mL) at 25°C. The pH of the solution was adjusted to 5-7, and then extracted with DCM (10 mL × 3). The combined organic layers were washed with brine (10 mL × 3), dried over Na 2 SO 4 , and then concentrated (below 30°C) under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , petroleum ether : ethyl acetate = 3:1) to give (S)-tert-butyl 4-(3-hydroxypropyl)-3-(trifluoromethyl)piperazine-1- carboxylate (40 mg, 128.0 µmol, 25.1% yield) as a colorless oil. 1 H NMR (400 MHz, CDCl 3 ) δ = 4.31 - 4.04 (m, 1H), 3.87 - 3.75 (m, 1H), 3.75 - 3.30 (m, 1H), 3.32 - 3.18 (m, 1H), 3.18 - 3.01 (m, 2H), 3.00 - 2.78 (m, 2H), 2.73 - 2.53 (m, 2H), 2.52 - 2.38 (m, 1H), 1.44 (s, 9H), 1.13 (t, J = 6.4 Hz, 3H) Step C. Procedure for preparation of (S)-tert-butyl 4-(3-(3-bromo-2- methylphenoxy)propyl)-3-(trifluoromethyl)piperazine-1-carbox ylate A mixture of (S)-tert-butyl 4-(3-hydroxypropyl)-3-(trifluoromethyl)piperazine-1-carboxyl ate (40 mg, 128.07 µmol, 1.00 equiv.), 3-bromo-2-methylphenol (35.93 mg, 192.11 µmol, 1.50 equiv.), and 2-(tributylphosphoranylidene)acetonitrile (46.37 mg, 192.11 µmol, 1.50 equiv.) in toluene (1 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 120 °C for 1 hour under N 2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (25 mL × 3). The combined organic layers were washed with brine (15 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give (S)-tert-butyl 4-(3-(3-bromo-2-methylphenoxy)propyl)-3- (trifluoromethyl)piperazine-1-carboxylate (40 mg, 83.0 µmol, 64.9% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.17 - 7.13 (m, 1H), 7.11 - 7.04 (m, 2H), 4.64 - 4.55 (m, 1H), 4.07 - 3.93 (m, 2H), 3.78 - 3.45 (m, 3H), 3.06 - 2.87 (m, 4H), 2.84 - 2.71 (m, 2H), 2.24 (d, J = 3.2 Hz, 1H), 2.22 (d, J = 5.2 Hz, 3H), 1.37 (d, J = 2.8 Hz, 9H) Step D. Procedure for preparation of (S)-1-(3-(3-bromo-2-methylphenoxy)propyl)-2- (trifluoromethyl)piperazine A mixture of (S)-tert-butyl 4-(3-(3-bromo-2-methylphenoxy)propyl)-3- (trifluoromethyl)piperazine-1-carboxylate (40 mg, 83.10 µmol, 1 equiv.) in HCl/EtOAc (5 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 16 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give (S)-1-(3-(3-bromo-2-methylphenoxy)propyl)-2-(trifluoromethyl )piperazine (40 mg, crude) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 9.69 (d, J = 2.4 Hz, 1H), 8.72 - 8.54 (m, 1H), 7.20 - 7.13 (m, 1H), 7.13 - 7.06 (m, 1H), 7.06 - 6.99 (m, 1H), 4.65 – 4.50 (m, 1H), 3.95 - 3.82 (m, 1H), 3.56 (s, 2H), 3.15 – 2.90 (m, 6H), 2.25 (d, J = 7.2 Hz, 1H), 2.21 (d, J = 3.6 Hz, 2H), 1.23 - 1.17 (m, 3H) Step E. Procedure for preparation of (S)-ethyl 2-(4-(3-(3-bromo-2- methylphenoxy)propyl)-3-(trifluoromethyl)piperazin-1-yl)acet ate A mixture of ethyl 2-bromoacetate (15.99 mg, 95.76 µmol, 10.59 µL, 1.00 equiv.), (S)-1-(3-(3- bromo-2-methylphenoxy)propyl)-2-(trifluoromethyl)piperazine (40.00 mg, 95.76 µmol, 1.00 equiv., HCl), K 2 CO 3 (66.18 mg, 478.82 µmol, 5.00 equiv.) , KI (1.59 mg, 9.58 µmol, 0.10 equiv.) in CH3CN (2 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 60 °C for 1 hour under N 2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (25 mL × 3). The combined organic layers were washed with brine (15 mL × 3), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The crude product was purified by reverse-phase HPLC to give (S)-ethyl 2-(4-(3-(3-bromo-2-methylphenoxy)propyl)-3- (trifluoromethyl)piperazin-1-yl)acetate (30 mg, 64.2 µmol, 67.0% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.14 (d, J = 7.2 Hz, 1H), 7.11 - 7.03 (m, 2H), 4.65 - 4.51 (m, 1H), 4.08 - 4.04 (m, 2H), 3.54 - 3.41 (m, 1H), 3.01 - 2.93 (m, 2H), 2.87 - 2.78 (m, 2H), 2.24 (d, J = 4.0 Hz, 2H), 2.21 (d, J = 3.2 Hz, 3H), 1.24 - 1.21 (m, 4H), 1.20 - 1.13 (m, 6H) Step F. Procedure for preparation of (S)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(4-(2-ethoxy-2-oxoe thyl)-2- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linate (S)-ethyl 2-(4-(3-(3-bromo-2-methylphenoxy)propyl)-3-(trifluoromethyl) piperazin-1- yl)acetate (30 mg, 64.20 µmol, 1.00 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3, 2-dioxaborolan-2-yl)picolinate (39.32 mg, 64.20 µmol, 1.00 equiv.), Ad 2 nBuP Pd G 3 (4.68 mg, 6.42 µmol, 0.10 equiv.), and KF (1.5 M, 128.39 µL, 3.00 equiv.) were taken up into a microwave tube in dioxane (1.5 mL). The sealed tube was heated at 100 °C for 1 hour under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , petroleum ether: ethyl acetate = 1:1) to give a compound (S)-tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-(4-(2-ethoxy-2- oxoethyl)-2-(trifluoromethyl)piperazin-1-yl)propoxy)-2-methy lphenyl)picolinate (45 mg, 51.5 µmol, 80.3% yield) as a brown oil. MS (ESI) m/z: 437.6 [M/2+H] + Step G. Procedure for preparation of (S)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)propyl)-3-(trifluoromethyl)piperazin-1-yl)acet ic acid A mixture of (S)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4-(2-ethoxy-2-oxoethyl)-2-(trifluoromethy l)piperazin-1-yl)propoxy)-2- methylphenyl)picolinate (45 mg, 51.55 µmol, 1.00 equiv.) and LiOH•H2O (6.49 mg, 154.64 µmol, 3.00 equiv.) in THF (1.5 mL) and H2O (0.5 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 40 °C for 3 hours under N 2 atmosphere. HCl (1 M) was slowly added dropwise to adjust pH to 3-4 and a white solid precipitated. The white solid was compound (S)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)-3- (trifluoromethyl)piperazin-1-yl)acetic acid (50 mg, crude). 1 H NMR (400 MHz, DMSO-d6) δ = 12.99 - 12.68 (m, 1H), 8.05 - 7.99 (m, 1H), 7.81 - 7.75 (m, 1H), 7.60 (d, J = 7.6 Hz, 1H), 7.50 - 7.42 (m, 3H), 7.42 - 7.27 (m, 3H), 7.15 - 7.05 (m, 1H), 7.04 - 6.90 (m, 2H), 6.63 - 6.53 (m, 1H), 5.03 - 4.95 (m, 2H), 4.61 (s, 1H), 3.87 (t, J = 5.6 Hz, 3H), 3.20 - 3.10 (m, 2H), 3.09 - 2.99 (m, 4H), 2.99 - 2.88 (m, 2H), 1.92 - 1.87 (m, 1H), 1.85 (s, 2H), 1.36 (s, 2H), 1.25 - 1.15 (m, 4H), 1.05 - 0.97 (m, 9H) Step H. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((2S)-4-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-2-(trifluoromethyl)pipera zin-1-yl)propoxy)-2- methylphenyl)picolinate A mixture of (S)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)-3- (trifluoromethyl)piperazin-1-yl)acetic acid (50 mg, 59.18 µmol, 1.00 equiv.), 3-(6-amino-1- methyl-1H-indazol-3-yl)piperidine-2,6-dione (16.81 mg, 65.09 µmol, 1.10 equiv.), and DIEA (22.94 mg, 177.53 µmol, 30.92 µL, 3.00 equiv.) in DMF (1 mL) was degassed and purged with N2 three times. The mixture was stirred at 25 °C for 5 minutes. After 5 minutes, HATU (27.00 mg, 71.01 µmol, 1.20 equiv.) was added, and the mixture was stirred at 25 °C for 1 hour under N2 atmosphere. The reaction mixture was slowly added dropwise into water (10 mL), and a pink solid precipitated. The pink solid was tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((2S)-4-(2-((3-(2,6 -dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-2-(trifluoromethyl)pipera zin-1-yl)propoxy)-2- methylphenyl)picolinate (60 mg, crude) MS (ESI) m/z: 543.5 [M/2+H] + Step I. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((2S)-4-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-2-(trifluoromethyl)pipera zin-1-yl)propoxy)-2- methylphenyl)picolinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-((2S)-4-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-6-yl)amino)-2- oxoethyl)-2-(trifluoromethyl)piperazin-1-yl)propoxy)-2-methy lphenyl)picolinate (60 mg, 55.29 µmol, 1.00 equiv.) in DCM (1 mL) and TFA (0.3 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 40 °C for 6 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give a compound 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((2S)-4-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)-2-(trifluoromethyl)piperazin -1-yl)propoxy)-2- methylphenyl)picolinic acid (30.5 mg, 29.3 µmol, 53.1% yield, 99.1% purity) as a yellow solid. MS (ESI) m/z: 1029.3 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 10.96 - 10.75 (m, 1H), 8.31 (s, 1H), 8.07 (s, 1H), 7.98 (d, J = 8.0 Hz, 1H), 7.78 - 7.72 (m, 1H), 7.64 (d, J = 8.0 Hz, 2H), 7.47 - 7.38 (m, 2H), 7.37 - 7.29 (m, 3H), 7.08 - 7.00 (m, 2H), 6.92 - 6.77 (m, 2H), 6.71 (d, J = 7.2 Hz, 1H), 4.97 (s, 2H), 4.62 - 4.48 (m, 1H), 4.35 - 4.29 (m, 1H), 3.92 - 3.87 (m, 5H), 3.24 (s, 2H), 3.11 (d, J = 4.4 Hz, 2H), 3.00 (s, 2H), 2.90 - 2.83 (m, 2H), 2.67 - 2.61 (m, 2H), 2.41 - 2.27 (m, 4H), 2.22 - 2.10 (m, 2H), 1.96 - 1.89 (m, 3H), 1.26 - 1.18 (m, 4H) 19 F NMR (377 MHz, DMSO-d6,) δ = -63.93 (s, 1F), -73.42 (s, 1F) Example 64. Preparation of Compound 246b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((2S)-4-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino )-2-oxoethyl)-2- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((2S)-4-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-2-oxoethyl)-2-(trifluoromethyl)pipera zin-1-yl)propoxy)-2- methylphenyl)picolinate A mixture of 3-(7-amino-1-methyl-1H-indazol-3-yl)piperidine-2,6-dione (37.60 mg, 145.57 µmol, 1.5 eq), (S)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)-3- (trifluoromethyl)piperazin-1-yl)acetic acid (100 mg, 97.05 µmol, 82% purity, 1 equiv.), and DIEA (50.17 mg, 388.19 µmol, 67.62 µL, 4 equiv.) in DMF (1 mL) was degassed and purged with N 2 three times. The mixture was stirred at 25 °C for 5 minutes. After 5 minutes, HATU (50 mg, 131.50 µmol, 1.35 equiv.) was added, and the mixture was stirred at 25 °C for 1 hour under N2 atmosphere. Water was added dropwise, and a yellow solid precipitated. The yellow solid was purified by prep-TLC (SiO 2 , DCM: MeOH = 12 : 1) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-((2S)-4-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino)-2- oxoethyl)-2- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linate (70 mg, 64.5 µmol, 66.4% yield) as a white solid. MS (ESI) m/z: 543.5 [M/2+H] + Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((2S)-4-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-2-oxoethyl)-2-(trifluoromethyl)pipera zin-1-yl)propoxy)-2- methylphenyl)picolinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-((2S)-4-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-7-yl)amino)-2- oxoethyl)-2-(trifluoromethyl)piperazin-1-yl)propoxy)-2-methy lphenyl)picolinate (70 mg, 64.50 µmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 40 °C for 4 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give a compound 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((2S)-4-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl-1H- indazol-7-yl)amino)-2-oxoethyl)-2-(trifluoromethyl)piperazin -1-yl)propoxy)-2- methylphenyl)picolinic acid (34.5 mg, 33.4 µmol, 51.8% yield, 99.4% purity) as a yellow solid. MS (ESI) m/z: 1029.4 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 12.85 (s, 2H), 10.90 (s, 1H), 9.81 - 9.57 (m, 1H), 8.13 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.65 - 7.56 (m, 2H), 7.49 - 7.43 (m, 3H), 7.40 - 7.32 (m, 2H), 7.23 - 7.18 (m, 1H), 7.12 - 7.05 (m, 2H), 6.99 - 6.91 (m, 2H), 6.66 - 6.61 (m, 1H), 4.98 (s, 2H), 4.65 - 4.50 (m, 1H), 4.42 - 4.34 (m, 1H), 4.07 - 4.02 (m, 3H), 3.92 (t, J = 5.6 Hz, 2H), 3.83 (s, 2H), 3.21 - 3.09 (m, 2H), 3.03 (t, J = 5.4 Hz, 2H), 2.95 - 2.83 (m, 2H), 2.74 - 2.61 (m, 4H), 2.43 - 2.28 (m, 2H), 2.27 - 2.25 (m, 1H), 2.26 - 2.00 (m, 1H), 1.93 - 1.86 (m, 3H), 1.24 (d, J = 3.2 Hz, 4H) 19 F NMR (376 MHz, DMSO-d 6 ) δ = -64.19 - -65.16 (m, 1F), -73.55 (s, 1F) Example 65. Preparation of Compound 248 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[2-[1-[2-[4-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]-1-piperidyl]e thyl]-4-piperidyl]ethoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[2-methyl-3-[2-[1-(2-oxoethyl)-4- piperidyl]ethoxy]phenyl]pyridine-2-carboxylic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(2-(1-(2,2-diethoxyethyl)piperidin-4-yl)ethoxy)-2-m ethylphenyl)picolinate (200 mg, 243.89 µmol, 1.0 equiv.) in HCOOH (1.5 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 85 °C for 2 hours under N 2 atmosphere. The reaction mixture was swept with nitrogen until it was dried to give 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[2-methyl-3- [2-[1-(2-oxoethyl)-4- piperidyl]ethoxy]phenyl]pyridine-2-carboxylic acid (168.2 mg, crude) as a white oil was used into the next step without further purification. MS (ESI) m/z: 708.1 [M+H + 2O +H] Step B. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[2-[1-[2-[4-[3-(2,6-dioxo-3-piperi dyl)-1-methyl-indazol-6-yl]-1- piperidyl]ethyl]-4-piperidyl]ethoxy]-2-methyl-phenyl]pyridin e-2-carboxylic acid To a solution of 3-[1-methyl-6-(4-piperidyl)indazol-3-yl]piperidine-2,6-dione (79.58 mg, 243.83 µmol, 1 equiv.), NaBH(OAc)3 (155.03 mg, 731.49 µmol, 3.0 equiv.) in DCM (5 mL) and IPA (5 mL) was added 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[2-methyl-3-[2-[1-(2-oxoethyl)-4-piperid yl]ethoxy]phenyl]pyridine-2- carboxylic acid (168.2 mg, 243.83 µmol, 1.0 equiv.) in DCM (5 mL) at 0 °C. The mixture was stirred at 25 °C for 2 hours. The reaction mixture was quenched by addition H2O (20 mL) and extracted with DCM (20 mL × 3). The combined organic layers were washed with saturated brine (20 mL × 2), dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[2-[1-[2-[4-[3-(2,6- dioxo-3-piperidyl)-1-methyl-indazol-6-yl]-1-piperidyl]ethyl] -4-piperidyl]ethoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid (19.2 mg, 18.9 µmol, 7.7% yield, 98.7% purity) as a white solid. MS (ESI) m/z: 1000.7 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 10.87 (s, 1H), 8.20 (s, 1H), 8.02 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.4 Hz, 1H), 7.64 - 7.57 (m, 2H), 7.48 - 7.31 (m, 6H), 7.11 - 6.99 (m, 2H), 6.91 - 6.85 (m, 2H), 6.66 (d, J = 7.6 Hz, 1H), 4.96 (s, 2H), 4.32 (dd, J = 5.2, 9.6 Hz, 1H), 3.98 (t, J = 4.8 Hz, 2H), 3.95 (s, 3H), 3.90 (t, J = 5.6 Hz, 2H), 3.06 - 2.98 (m, 6H), 2.96 - 2.93 (m, 2H), 2.67 - 2.60 (m, 3H), 2.20 - 2.02 (m, 6H), 1.90 (s, 3H), 1.81 - 1.73 (m, 4H), 1.72 - 1.63 (m, 4H), 1.58 - 1.44 (m, 2H), 1.27 - 1.19 (m, 2H) Example 66. Preparation of Compound 250a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(((1s,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pip erazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)propyl)cyclohexyl)oxy) -2- methylphenyl)picolinate To a solution of 3-(1-methyl-7-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (56.34 mg, 154.85 µmol, 1.2 equiv.) in DCM (2 mL) was added NaBH(OAc)3 (82.05 mg, 387.12 µmol, 3 equiv.). Then tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(2-methyl-4-(((1r,4r)-4-(3-oxopropyl)cyclohexyl)oxy)ph enyl)picolinate (94.32 mg, 129.04 µmol, 1 equiv.) was added into the mixture at 0 °C. The mixture was stirred at 25 °C for 1 hour. DCM (20 mL) and water (20 mL) were added and layers were separated. The aqueous phase was extracted with DCM (10 mL × 2). The combined organic extracts were dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(((1s,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pip erazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (120 mg, 115.1 µmol, 89.2% yield) as a yellow solid. MS (ESI) m/z: 522.0 [M/2+H] + Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)propyl)cyclohexyl)oxy) -2-methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-7- yl)piperazin-1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl)pico linate (120.00 mg, 115.13 µmol, 1 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 117.31 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(4-(((1s,4r)-4-(3-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin -1-yl)propyl)cyclohexyl)oxy)- 2-methylphenyl)picolinic acid (41.7 mg, 41.9 µmol, 36.5% yield, 99.2% purity) as a yellow solid. MS (ESI) m/z: 986.3 [M+H] + 1 H NMR (400 MHz) δ = 12.88 (s, 1H), 12.65 - 12.52 (m, 1H), 10.90 (s, 1H), 9.57 - 9.44 (m, 1H), 8.04 (d, J = 8.0 Hz, 1H), 7.80 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 7.2 Hz, 1H), 7.49 (d, J = 7.6 Hz, 1H), 7.47 - 7.43 (m, 2H), 7.40 - 7.33 (m, 2H), 7.13 - 7.05 (m, 2H), 6.98 - 6.89 (m, 2H), 6.79 (d, J = 2.4 Hz, 1H), 6.74 - 6.69 (m, 1H), 4.98 (s, 2H), 4.39 - 4.34 (m, 1H), 4.27 (s, 3H), 4.22 (s, 1H), 3.92 (t, J = 5.6 Hz, 2H), 3.62 (d, J = 11.2 Hz, 2H), 3.44 - 3.40 (m, 2H), 3.23 - 3.17 (m, 2H), 3.13 - 3.06 (m, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.71 - 2.62 (m, 2H), 2.55 (s, 2H), 2.36 - 2.31 (m, 1H), 2.21 - 2.14 (m, 1H), 2.13 - 2.07 (m, 2H), 2.03 (s, 3H), 1.86 - 1.80 (m, 2H), 1.72 (d, J = 6.4 Hz, 2H), 1.38 - 1.23 (m, 5H), 1.16 - 1.05 (m, 2H) Example 67. Preparation of Compound 252a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(((1r,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pip erazin-1-yl)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(((1r,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)-3-oxopropyl)cyclohexy l)oxy)-2- methylphenyl)picolinate To a solution of 3-[4-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1 H-isoquinolin- 2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-3-methyl-phenoxy]cycl ohexyl]propanoic acid (100 mg, 133.88 µmol, 1 equiv.) in pyridine (1 mL) was added EDCI (33.37 mg, 174.05 µmol, 1.3 equiv.) and 3-(1-methyl-7-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (58.46 mg, 160.66 µmol, 1.2 equiv., HCl). The mixture was stirred at 40 °C for 4 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=4/1 to 0/1, DCM:MeOH = 10:1) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4- (((1r,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-7-yl)piperazin-1-yl)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (140 mg, 132.5 µmol, 99.0% yield) as a yellow solid. MS (ESI) m/z: 1056.3 [M+H] + Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(((1r,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)-3-oxopropyl)cyclohexy l)oxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(((1r,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-7- yl)piperazin-1-yl)-3-oxopropyl)cyclohexyl)oxy)-2-methylpheny l)picolinate (140 mg, 132.54 µmol, 1 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 101.90 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(4-(((1r,4r)-4-(3-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin -1-yl)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (26.3 mg, 23.9 µmol, 18.0% yield, 90.8% purity) as a yellow solid. MS (ESI) m/z: 1000.3 [M+H] + 1 H NMR (400 MHz) δ = 12.86 (s, 1H), 12.69 - 12.47 (m, 1H), 10.89 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.49 - 7.42 (m, 4H), 7.39 - 7.33 (m, 2H), 7.06 - 7.00 (m, 2H), 6.96 - 6.88 (m, 2H), 6.78 (d, J = 2.0 Hz, 1H), 6.73 - 6.68 (m, 1H), 4.97 (s, 2H), 4.37 - 4.32 (m, 1H), 4.27 (s, 3H), 3.90 (t, J = 5.6 Hz, 2H), 3.59 - 3.53 (m, 2H), 3.23 - 3.13 (m, 2H), 3.02 (t, J = 5.6 Hz, 2H), 2.68 - 2.61 (m, 5H), 2.43 - 2.37 (m, 2H), 2.35 - 2.31 (m, 1H), 2.24 - 2.13 (m, 2H), 2.09 - 2.04 (m, 2H), 2.02 (s, 3H), 1.86 - 1.77 (m, 2H), 1.47 (d, J = 6.4 Hz, 2H), 1.36 - 1.25 (m, 4H), 1.15 - 1.05 (m, 2H) Example 68. Preparation of Compound 253a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pip eridin-1-yl)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of 3-((1r,4r)-4-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)cyclohexyl)propanoic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(3-ethoxy-3-oxopropyl)cyclohexyl) oxy)-2- methylphenyl)picolinate (75 mg, 96.78 µmol, 1 equiv.) in THF (0.75 mL) and H2O (0.25 mL) was added LiOH·H2O (20.31 mg, 483.89 µmol, 5 equiv.). The mixture was stirred at 25 °C for 10 hours. The reaction mixture was extracted with EtOAc (5mL). The combined organic layers were concentrated under reduced pressure to give the crude product 3-((1r,4r)-4-(3-(6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)cyclohexyl)prop anoic acid (60 mg, crude) as a white solid, which was used into the next step without further purification. MS (ESI) m/z: 747.4 [M+H] + . Step B. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperidin-1-yl)-3-oxopropyl)cyclohexy l)oxy)-2- methylphenyl)picolinate To a solution of 3-((1r,4r)-4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)cyclohexyl)propanoic acid (55 mg, 73.64 µmol, 1 equiv.) in pyridine (1 mL) was added 3-[1-methyl-7-(4-piperidyl)indazol-3-yl]piperidine-2,6-dione (36.05 mg, 110.45 µmol, 1.5 equiv.) and EDCI (42.35 mg, 220.91 µmol, 3 equiv.).The mixture was stirred at 25 °C for 2 hours. The reaction mixture was quenched by addition 1M HCl 10 (mL) at 25 °C, and then diluted with H2O 10 (mL) and extracted with DCM (10 mL). The combined organic layers were concentrated under reduced pressure to give the crude product tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1r,4r)-4-(3-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperidin -1-yl)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (70 mg, crude) as a black solid, which was used into the next step without further purification. MS (ESI) m/z: 1055.7 [M+H] + Step C. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperidin-1-yl)-3-oxopropyl)cyclohexy l)oxy)-2- methylphenyl)picolinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1r,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-7-yl)piperidin-1- yl)-3-oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (60 mg, 56.86 µmol, 1 equiv.) in TFA (0.5 mL) and DCM (1 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 25 °C for 16 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC. The compound 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1r,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-7-yl)piperidin-1-yl)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (18.8 mg, 18.74µmol, 32.9 % yield, 99.5 % purity) was obtained as a white solid. MS (ESI) m/z: 999.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.86 ( d, J = 1.2 Hz, 1H), 12.70 - 12.42 (m, 1H), 10.88 (s, 1H), 8.06 - 8.01 (m, 1H), 7.79 (d, J = 8.4 Hz, 1H), 7.62 ( d, J = 6.4 Hz, 1H), 7.55 (d, J = 8.0 Hz, 1H), 7.49 - 7.43 (m, 3H), 7.39 - 7.32 (m, 2H), 7.23 (d, J = 7.2 Hz, 1H), 7.10 - 7.04 (m, 2H), 6.94 (dd, J = 8.4, 15.6 Hz, 2H), 6.61 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.67 - 4.53 (m, 1H), 4.35 (dd, J = 5.2, 9.6 Hz, 1H), 4.24 (s, 3H), 4.22 - 4.16 (m, 1H), 4.08 - 3.99 (m, 1H), 3.92 ( t, J = 5.7 Hz, 2H), 3.67 - 3.57 (m, 1H), 3.02 ( t, J = 5.6 Hz, 2H), 2.74 - 2.61 (m, 3H), 2.41 - 2.36 (m, 3H), 2.33 (td, J = 1.8, 3.6 Hz, 1H), 2.16 ( dd, J = 5.6, 13.6 Hz, 1H), 2.11 - 2.06 (m, 2H), 1.98 - 1.90 (m, 2H), 1.87 (s, 3H), 1.82 ( d, J = 12.4 Hz, 2H), 1.72 - 1.65 (m, 1H), 1.60 - 1.52 (m, 1H), 1.50 - 1.44 (m, 2H), 1.39 - 1.28 (m, 3H), 1.15 - 1.06 (m, 2H) Example 69. Preparation of Compound 259a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip eridin-1-yl)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-(3-ethoxy-3-oxo-pro pyl)cyclohexoxy]-2-methyl- phenyl]pyridine-2-carboxylate A mixture of ethyl 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]propanoate (500 mg, 1.35 mmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (912.29 mg, 1.49 mmol, 1.1 equiv.), KF (1.5 M, 1.35 mL, 1.5 equiv.), and Ad 2 nBuP Pd G 3 (cataCXium® A Pd G 3 ) (295.81 mg, 406.18 µmol, 0.3 equiv.) in dioxane (15 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 100 °C for 1 hour under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~38 % ethyl acetate/petroleum ether) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[4-(3-ethoxy-3-oxo-propyl) cyclohexoxy]-2-methyl- phenyl]pyridine-2-carboxylate (700 mg, 496.8 µmol, 36.6% yield, 55% purity) as a yellow solid. MS (ESI) m/z: 775.5 [M+H] + Step B. Procedure for preparation of 3-[4-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl ]-2-methyl- phenoxy]cyclohexyl]propanoic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[4-(3-ethoxy-3-oxo-propyl)cyclohexoxy]-2-methyl-p henyl]pyridine-2-carboxylate (470 mg, 606.48 µmol, 1 equiv.) in THF (5 mL) was added LiOH•H2O (152.70 mg, 3.64 mmol, 6 equiv.) and H 2 O (1 mL). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 78% ethyl acetate / petroleum ether) to give 3-[4-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1 H-isoquinolin-2-yl]-2- tert-butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]cyclohexyl]p ropanoic acid (400 mg, 535.5 µmol, 88.3% yield) was obtained as a white solid. MS (ESI) m/z: 747.4 [M+H] + Step C. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo -3-piperidyl)-1-methyl- indazol-6-yl]-1-piperidyl]-3-oxo-propyl]cyclohexoxy]-2-methy l-phenyl]pyridine-2- carboxylate A mixture of 3-[4-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1 H-isoquinolin-2- yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]cycloh exyl]propanoic acid (299.60 mg, 401.12 µmol, 1 equiv.), EDCI (115.34 mg, 601.68 µmol, 1.5 equiv.) in pyridine (3 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 0 °C for 1 hour under N2 atmosphere. 3-[1-methyl-6-(4-piperidyl)indazol-3-yl]piperidine-2,6-dione (196.39 mg, 601.68 µmol, 1.5 equiv.) was added, and the mixture was stirred at 60 °C for 5 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 67% ethyl acetate / petroleum ether). The compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo -3-piperidyl)-1-methyl-indazol- 6-yl]-1-piperidyl]-3-oxo-propyl]cyclohexoxy]-2-methyl-phenyl ]pyridine-2-carboxylate (150 mg, 142.1 µmol, 35.4% yield) was obtained as a white solid. MS (ESI) m/z: 528.7 [ 1 / 2 M+H] + . Step D. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperidin-1-yl)-3-oxopropyl)cyclohexy l)oxy)-2- methylphenyl)picolinic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazo l-6-yl]-1-piperidyl]-3-oxo- propyl]cyclohexoxy]-2-methyl-phenyl]pyridine-2-carboxylate (150 mg, 142.14 µmol, 1 equiv.) in TFA (1 mL) and DCM (3 mL) was stirred at 40 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-6- yl)piperidin-1-yl)-3-oxopropyl)cyclohexyl)oxy)-2-methylpheny l)picolinic acid (28.6 mg, 25.2 µmol, 17.7% yield, 88.0% purity) as an off-white solid. MS (ESI) m/z: 999.0 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 10.89 - 10.84 (m, 1H), 8.02 (d, J = 6.8 Hz, 1H), 7.78 (d, J = 7.6 Hz, 1H), 7.64 - 7.59 (m, 2H), 7.46 - 7.42 (m, 3H), 7.40 - 7.31 (m, 2H), 7.13 - 6.99 (m, 3H), 6.92 (t, J = 7.6 Hz, 2H), 6.62 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.60 - 4.56 (m, 1H), 4.34 - 4.30 (m, 1H), 4.22 - 4.17 (m, 1H), 4.05 - 4.03 (m, 1H), 3.96 (s, 3H), 3.92 - 3.89 (m, 2H), 3.18 - 3.08 (m, 2H), 3.03 - 3.00 (m, 2H), 2.95 - 2.84 (m, 1H), 2.68 - 2.58 (m, 4H), 2.40 - 2.32 (m, 4H), 2.11 - 2.05 (m, 2H), 1.87 (s, 3H), 1.85 - 1.81 (m, 2H), 1.63 - 1.51 (m, 2H), 1.48 - 1.45 (m, 2H), 1.38 - 1.30 (m, 3H), 1.12 - 1.06 (m, 2H). Example 70. Preparation of Compound 263a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[(2R)-3-[1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-o xo-ethyl]-4-piperidyl]-2- methyl-propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(3-((2R)-3-(1- (2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)am ino)-2-oxoethyl)piperidin-4- yl)-2-methylpropoxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of tert-butyl 4-[(E)-3-ethoxy-2-methyl-3-oxo-prop-1- enyl]piperidine-1-carboxylate A mixture of ethyl 2-diethoxyphosphorylpropanoate (8.38 g, 35.17 mmol, 7.69 mL, 1.50 equiv.) in THF (50 mL) was degassed and purged with N 2 three times, and then to the mixture was added LiHMDS (1 M, 46.89 mL, 2.00 equiv.) dropwise at 0°C under N 2 atmosphere. After stirring at 0 °C for 1 hour, tert-butyl 4-formylpiperidine-1-carboxylate (5 g, 23.44 mmol, 1.00 equiv.) in THF (20 mL) was added dropwise at 0 °C. The mixture was stirred at 25 °C for 15 hours. After completion, the solution was cooled to 0 °C, and then added into a saturated NH 4 Cl solution (100 mL) slowly. The solution was extracted with ethyl acetate (100 mL × 3). The combined organic layers were washed with brine (200 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~25% ethyl acetate/petroleum ether) to give tert-butyl 4-[(E)-3-ethoxy-2-methyl-3-oxo-prop-1-enyl]piperidine-1-carb oxylate (4.3 g, 14.4 mmol, 61.6% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 6.44 - 6.36 (m, 1H), 5.78 - 5.66 (m, 1H), 4.14 - 3.97 (m, 2H), 3.92 - 3.78 (m, 2H), 2.65 (s, 2H), 1.79 - 1.71 (m, 3H), 1.54 - 1.45 (m, 2H), 1.36 - 1.25 (m, 10H), 1.19 - 1.12 (m, 3H), 1.08 - 1.02 (m, 1H) Step B. Procedure for preparation of tert-butyl 4-(3-ethoxy-2-methyl-3-oxo- propyl)piperidine-1-carboxylate A mixture of tert-butyl 4-[(E)-3-ethoxy-2-methyl-3-oxo-prop-1-enyl]piperidine-1-carb oxylate (4.3 g, 14.46 mmol, 1.00 equiv.), Pd/C (2 g, 1.45 mmol, 10% purity, 0.10 equiv.), Pd(OH) 2 (2.03 g, 1.45 mmol, 10% purity, 0.10 equiv.) in EtOH (40 mL) was degassed and purged with H2 (15 Psi) three times, and then the mixture was stirred at 25 °C for 16 hours under H2 (15 Psi) atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give crude tert-butyl 4-(3-ethoxy-2-methyl-3-oxo-propyl)piperidine-1-carboxylate (3.5 g, 11.6 mmol, 80.8% yield) as a black oil. Step C. Procedure for preparation of tert-butyl 4-(3-hydroxy-2-methyl- propyl)piperidine-1-carboxylate A mixture of tert-butyl 4-(3-ethoxy-2-methyl-3-oxo-propyl)piperidine-1-carboxylate (3.5 g, 11.69 mmol, 1.00 equiv.) in THF (50 mL) was degassed and purged with N 2 for three times, and then LiAlH4 (665.52 mg, 17.53 mmol, 1.50 equiv.) was added in portions and stirred at 25 °C for 1 hour under N2 atmosphere. To the reaction mixture was added H2O (0.7 mL) at 0 °C, then filtered and concentrated to give a crude product. The residue was purified by flash silica gel chromatography (Eluent of 0~30% ethyl acetate/petroleum ether) to give tert-butyl 4-(3- hydroxy-2-methyl-propyl)piperidine-1-carboxylate (2.6 g, 10.1 mmol, 86.4% yield) as a colorless oil 1 H NMR (400 MHz, DMSO-d 6 ) δ = 4.38 (t, J = 5.2 Hz, 1H), 3.97 - 3.85 (m, 2H), 3.29 - 3.21 (m, 1H), 3.20 - 3.12 (m, 1H), 2.78 - 2.56 (m, 2H), 1.65 - 1.54 (m, 3H), 1.51 - 1.43 (m, 1H), 1.39 (s, 9H), 1.28 - 1.21 (m, 1H), 1.00 - 0.85 (m, 3H), 0.82 (d, J = 6.8 Hz, 3H) Step D. Procedure for preparation of tert-butyl 4-[3-(3-bromo-2-methyl-phenoxy)-2- methyl-propyl]piperidine-1-carboxylate A mixture of tert-butyl 4-(3-hydroxy-2-methyl-propyl)piperidine-1-carboxylate (2.6 g, 10.10 mmol, 1.00 equiv.), 3-bromo-2-methyl-phenol (1.89 g, 10.10 mmol, 1.00 equiv.), 2-(tributyl- λ 5 -phosphanylidene)acetonitrile (2.93 g, 12.12 mmol, 1.20 equiv.) in toluene (20 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 110 °C for 16 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~5% ethyl acetate/petroleum ether) to give tert-butyl 4-[3-(3-bromo-2-methyl-phenoxy)-2-methyl- propyl]piperidine-1-carboxylate (3.2 g, 7.1 mmol, 70.5% yield, 95% purity) as a colorless oil. MS (ESI) m/z: 448.2 [M+23] + . Step E. Procedure for preparation of 4-[3-(3-bromo-2-methyl-phenoxy)-2-methyl- propyl]piperidine To a solution of tert-butyl 4-[3-(3-bromo-2-methyl-phenoxy)-2-methyl-propyl]piperidine-1 - carboxylate (3 g, 7.04 mmol, 1.00 equiv.) in HCl/EtOAc (1 mL) was stirred at 25 °C for 0.5 hour. The reaction mixture was filtered to give crude 4-[3-(3-bromo-2-methyl-phenoxy)-2- methyl-propyl]piperidine (2 g, 5.5 mmol, 78.3% yield, HCl) as a white solid. Step F. Procedure for preparation of ethyl 2-[4-[3-(3-bromo-2-methyl-phenoxy)-2- methyl-propyl]-1-piperidyl]acetate To a solution of 4-[3-(3-bromo-2-methyl-phenoxy)-2-methyl-propyl]piperidine (2 g, 5.51 mmol, 1 equiv., HCl) in CH3CN (20 mL) was added K2CO3 (2.29 g, 16.54 mmol, 3 equiv.) and ethyl 2-bromoacetate (920.80 mg, 5.51 mmol, 609.80 µL, 1 equiv.). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~30% ethyl acetate/petroleum ether) to give ethyl 2-[4-[3-(3-bromo-2-methyl-phenoxy)-2-methyl- propyl]-1-piperidyl]acetate (2.2 g, 5.3 mmol, 96.7% yield) as a colorless oil. MS (ESI) m/z: 412.2 [M+H] + . Step G. Procedure for preparation of ethyl 2-[4-[(2R)-3-(3-bromo-2-methyl-phenoxy)-2- methyl-propyl]-1-piperidyl]acetate Ethyl 2-[4-[3-(3-bromo-2-methyl-phenoxy)-2-methyl-propyl]-1-piperi dyl]acetate (1 g, 2.43 mmol, 1.00 equiv.) was separated by SFC to give ethyl 2-[4-[(2R)-3-(3-bromo-2-methyl- phenoxy)-2-methyl-propyl]-1-piperidyl]acetate (0.29 g, 703.2 µmol, 29.0% yield) as a colorless oil. Step H. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[(2R)-3-[1-(2-ethoxy-2 -oxo-ethyl)-4-piperidyl]-2- methyl-propoxy]-2-methyl-phenyl]pyridine-2-carboxylate A mixture of ethyl 2-[4-[(2R)-3-(3-bromo-2-methyl-phenoxy)-2-methyl-propyl]-1- piperidyl]acetate (150 mg, 363.76 µmol, 1.00 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tet ramethyl-1,3,2-dioxaborolan-2- yl)pyridine-2-carboxylate (222.82 mg, 363.76 µmol, 1.00 equiv.), Ad 2 nBuP Pd G 3 (264.92 mg, 363.76 µmol, 1.00 equiv.), and K 3 PO 4 (231.64 mg, 1.09 mmol, 3.00 equiv.) in dioxane (2 mL) and H 2 O (0.2 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 75 °C for 16 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, petroleum ether/ethyl acetate = 1:1) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[(2R)-3-[1-(2-ethoxy-2 -oxo-ethyl)-4-piperidyl]-2- methyl-propoxy]-2-methyl-phenyl]pyridine-2-carboxylate (150 mg, 183.3 µmol, 50.4% yield) was obtained as a yellow solid. MS (ESI) m/z: 818.5 [M+H] + . Step I. Procedure for preparation of 2-[4-[(2R)-3-[3-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-2- methyl-phenoxy]-2-methyl-propyl]-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[(2R)-3-[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]-2- methyl-propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (140 mg, 171.14 µmol, 1.00 equiv.) in THF (2 mL) was added LiOH•H2O (21.55 mg, 513.43 µmol, 3.00 equiv.) and H2O (0.5 mL). The mixture was stirred at 25 °C for 3 minutes. The reaction mixture was concentrated under reduced pressure to give crude 2-[4-[(2R)-3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H-isoquinolin-2- yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]-2-met hyl-propyl]-1-piperidyl]acetic acid (100 mg, 126.5 µmol, 73.9% yield) as a yellow solid. MS (ESI) m/z: 790.5 [M+H] + . Step J. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[(2R)-3-[1-[2-[[3-(2,6 -dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-4-piperidyl]-2-methyl-propo xy]-2-methyl- phenyl]pyridine-2-carboxylate To a solution of 2-[4-[(2R)-3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]-2-methyl-propyl]-1- piperidyl]acetic acid (80 mg, 101.27 µmol, 1.00 equiv.) and 3-(6-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (28.77 mg, 111.40 µmol, 1.10 equiv.) in DMF (1 mL) was added HATU (46.21 mg, 121.52 µmol, 1.20 equiv.) and TEA (30.74 mg, 303.81 µmol, 42.29 µL, 3.00 equiv.).The mixture was stirred at 40 °C for 1 hour. The solution was poured into water (2 mL) and filtered to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[(2R)-3-[1-[2-[[3-(2,6-dioxo-3-piperi dyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]-2-methyl-propoxy]-2-meth yl-phenyl]pyridine-2- carboxylate (80 mg, 77.6 µmol, 76.6% yield) as a pink solid. MS (ESI) m/z: 1030.5 [M+H] + . Step K. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[(2R)-3-[1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]-2-methyl-propoxy]-2-meth yl-phenyl]pyridine-2- carboxylic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)- yl)-3-(3-((2R)-3-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-6-yl)amino)-2- oxoethyl)piperidin-4-yl)-2-methylpropoxy)-2-methylphenyl)pic olinic acid] To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[(2R)-3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methy l-indazol-6-yl]amino]-2-oxo- ethyl]-4-piperidyl]-2-methyl-propoxy]-2-methyl-phenyl]pyridi ne-2-carboxylate (80 mg, 77.65 µmol, 1.00 equiv.) in DCM (1 mL) was added TFA (616.00 mg, 5.40 mmol, 400.00 µL, 69.57 equiv.).The mixture was stirred at 40 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8- (1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin- 2-yl]-3-[3-[(2R)-3-[1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-4-piperidyl]-2-methyl- propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid [rel-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-((2R)-3-( 1-(2-((3-(2,6-dioxopiperidin-3- yl)-1-methyl-1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-4-y l)-2-methylpropoxy)-2- methylphenyl)picolinic acid] (28.5 mg, 27.7 µmol, 35.7% yield, 99.1% purity) as a yellow solid. MS (ESI) m/z: 488.0 [M/2+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.36 - 11.83 (m, 1H), 10.88 (s, 1H), 9.83 (s, 1H), 8.16 (s, 1H), 8.08 - 7.97 (m, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 8.4 Hz, 2H), 7.49 - 7.41 (m, 3H), 7.40 - 7.31 (m, 2H), 7.21 (dd, J = 1.6, 8.8 Hz, 1H), 7.12 - 7.05 (m, 1H), 6.95 (d, J = 8.8 Hz, 1H), 6.87 (d, J = 8.4 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.32 (dd, J = 5.2, 9.6 Hz, 1H), 3.95 - 3.86 (m, 5H), 3.83 - 3.73 (m, 2H), 3.13 (s, 2H), 3.02 (t, J = 5.2 Hz, 2H), 2.91 - 2.83 (m, 2H), 2.67 - 2.59 (m, 2H), 2.20 - 2.14 (m, 2H), 2.02 (s, 1H), 1.95 - 1.87 (m, 3H), 1.74 - 1.61 (m, 2H), 1.49 - 1.37 (m, 2H), 1.36 - 1.09 (m, 4H), 1.04 - 0.97 (m, 3H) Example 71. Preparation of Compound 267 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-4-methyl-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 4-[(E)-3-ethoxy-3-oxo-prop-1-enyl]-4- methyl-piperidine-1-carboxylate To a solution of ethyl 2-diethoxyphosphorylacetate (1.97 g, 8.80 mmol, 1.75 mL, 2.0 equiv.) in THF (10 mL) was added LiHMDS (1 M, 13.20 mL, 3.0 equiv.) at 0 °C, and then the reaction mixture was stirred at 0 °C for 1 hour. After that, tert-butyl 4-formyl-4-methyl-piperidine-1- carboxylate (1 g, 4.40 mmol, 1.0 equiv.) was added into above mixture, and the resulting mixture was stirred at 25 °C for 11 hours. The reaction mixture was quenched by adding saturated ammonium chloride solution (10 mL) at 25 °C. It was then diluted with EtOAc (10 mL) and extracted with EtOAc 30 mL (10 mL × 3). The combined organic layers were washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to tert-butyl 4-[(E)-3-ethoxy-3-oxo-prop-1-enyl]-4-methyl-piperidine-1- carboxylate (1.08 g, crude) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 6.85 (d, J = 16.0 Hz, 1H), 5.83 (d, J = 16.0 Hz, 1H), 4.15 - 4.08 (m, 2H), 3.37 (d, J = 4.0 Hz, 2H), 3.26 - 3.19 (m, 2H), 1.62 - 1.53 (m, 2H), 1.38 (s, 9H), 1.36 - 1.32 (m, 2H), 1.21 (t, J = 7.2 Hz, 3H), 1.05 (s, 3H). Step B. Procedure for preparation of tert-butyl 4-(3-ethoxy-3-oxo-propyl)-4-methyl- piperidine-1-carboxylate A mixture of tert-butyl 4-[(E)-3-ethoxy-3-oxo-prop-1-enyl]-4-methyl-piperidine-1- carboxylate (1.08 g, 3.63 mmol, 1.0 equiv.) Pd/C (500 mg, 10% purity, 1.0 equiv.) in MeOH (10 mL) was degassed and purged with H2 three times, and then the mixture was stirred at 25 °C for 12 hours under H 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give tert-butyl 4-(3-ethoxy-3-oxo-propyl)-4-methyl-piperidine-1- carboxylate (1.06 g, 3.5 mmol, 97.4% yield) as a yellow oil. 1 H NMR (400 MHz, CDCl 3 ) δ = 4.18 - 4.09 (m, 2H), 3.58 (d, J = 12.8 Hz, 3H), 3.25 - 3.15 (m, 3H), 2.41 (s, 1H), 2.32 - 2.23 (m, 2H), 1.66 - 1.60 (m, 3H), 1.46 (s, 9H), 1.27 (t, J = 7.2 Hz, 3H), 0.93 (s, 3H). Step C. Procedure for preparation of tert-butyl 4-(3-hydroxypropyl)-4-methyl- piperidine-1-carboxylate To a solution of tert-butyl 4-(3-ethoxy-3-oxo-propyl)-4-methyl-piperidine-1-carboxylate (1 g, 3.34 mmol, 1.0 equiv.) in THF (10 mL) was added LiAlH 4 (152.12 mg, 4.01 mmol, 1.2 equiv.). The mixture was stirred at 0 °C for 1 hour. The reaction mixture was quenched with saturated sodium sulfate solution (0.456 mL) at 0 °C, filtered, and concentrated to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate 10/1 to 2/1) to give tert-butyl 4-(3-hydroxypropyl)-4-methyl-piperidine-1-carboxylate (500 mg, 1.9 mmol, 58.1% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 4.36 (t, J = 5.2 Hz, 1H), 3.45 - 3.39 (m, 2H), 3.36 (d, J = 5.6 Hz, 2H), 3.13 (d, J = 5.6 Hz, 2H), 1.38 (s, 9H), 1.36 - 1.31 (m, 2H), 1.26 - 1.20 (m, 6H), 0.88 - 0.86 (m, 3H). Step D. Procedure for preparation of tert-butyl 4-[3-(3-bromo-2-methyl- phenoxy)propyl]-4-methyl-piperidine-1-carboxylate To a solution of tert-butyl 4-(3-hydroxypropyl)-4-methyl-piperidine-1-carboxylate (250 mg, 971.37 µmol, 1.0 equiv.) in toluene (3 mL) and 3-bromo-2-methyl-phenol (218.01 mg, 1.17 mmol, 1.2 equiv.) was added 2-(tributyl-λ5-phosphanylidene)acetonitrile (351.67 mg, 1.46 mmol, 1.5 equiv.). The mixture was stirred at 120 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate 1/0 to 20/1) to give tert-butyl 4-[3-(3- bromo-2-methyl-phenoxy)propyl]-4-methyl-piperidine-1-carboxy late (200 mg, 469.0 µmol, 48.2% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.16 - 7.13 (m, 1H), 7.12 - 7.06 (m, 1H), 6.95 (d, J = 8.0 Hz, 1H), 3.96 (t, J = 6.4 Hz, 2H), 3.45 - 3.40 (m, 2H), 3.22 - 3.12 (m, 2H), 2.23 (s, 3H), 1.74 - 1.64 (m, 2H), 1.41 - 1.36 (m, 11H), 1.31 - 1.24 (m, 4H), 0.92 (s, 3H). Step E. Procedure for preparation of 4-[3-(3-bromo-2-methyl-phenoxy)propyl]-4-methyl- piperidine A solution of tert-butyl 4-[3-(3-bromo-2-methyl-phenoxy)propyl]-4-methyl-piperidine-1 - carboxylate (200 mg, 469.06 µmol, 1.0 equiv.) in HCl/dioxane (2 mL) was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give compound 4-[3- (3-bromo-2-methyl-phenoxy)propyl]-4-methyl-piperidine (240 mg, crude) as a white solid. Step F. Procedure for preparation of ethyl 2-[4-[3-(3-bromo-2-methyl-phenoxy)propyl]- 4-methyl-1-piperidyl]acetate To a solution of 4-[3-(3-bromo-2-methyl-phenoxy)propyl]-4-methyl-piperidine (240 mg, 735.59 µmol, 1.0 equiv.) and ethyl 2-bromoacetate (98.27 mg, 588.47 µmol, 65.08 µL, 1.0 equiv.) in CH 3 CN (2 mL) was added K 2 CO 3 (304.99 mg, 2.21 mmol, 3.0 equiv.). The mixture was stirred at 60 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give ethyl 2-[4-[3-(3-bromo-2-methyl-phenoxy)propyl]-4-methyl-1- piperidyl]acetate (300 mg, 727.5 µmol, 98.9% yield) as a yellow oil. MS (ESI) m/z: 412.2 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 7.17 - 7.13 (m, 1H), 7.12 - 7.06 (m, 1H), 6.95 (d, J = 7.6 Hz, 1H), 4.11 - 4.04 (m, 2H), 3.96 (t, J = 6.4 Hz, 2H), 3.18 (s, 2H), 2.53 (s, 1H), 2.48 - 2.47 (m, 1H), 2.46 - 2.39 (m, 2H), 2.24 (s, 3H), 1.75 - 1.60 (m, 2H), 1.40 - 1.29 (m, 6H), 1.20 - 1.16 (m, 3H), 0.88 (s, 3H). Step G. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-(2-ethoxy-2-oxo- ethyl)-4-methyl-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate Ethyl 2-[4-[3-(3-bromo-2-methyl-phenoxy)propyl]-4-methyl-1-piperid yl]acetate (300 mg, 727.52 µmol, 1.0 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (534.77 mg, 873.02 µmol, 1.2 equiv.), [2-(2-aminophenyl)phenyl]palladium(1+);bis(1- adamantyl)-butyl-phosphane;methanesulfonate (52.98 mg, 72.75 µmol, 0.1 equiv.), and KF (1.5 M, 1.46 mL, 3 equiv.) were taken up into a microwave tube in dioxane (3 mL). The sealed tube was heated at 100 °C for 1 hour under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate 10/1 to 1/1) to give tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[3-[1-(2-ethoxy-2-oxo- ethyl)-4-methyl-4-piperidyl]propoxy]-2-methyl-phenyl]pyridin e-2-carboxylate (320 mg, 391.1 µmol, 53.7% yield) as a yellow solid. MS (ESI) m/z: 818.5 [M+H] + Step H. Procedure for preparation of 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyr idyl]-2-methyl- phenoxy]propyl]-4-methyl-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[3-[1-(2-ethoxy-2-oxo-ethyl)-4-methyl-4-piperidyl ]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (120 mg, 146.69 µmol, 1.0 equiv.) in THF (1 mL) and H 2 O (1 mL) was added LiOH·H 2 O (13 mg, 1.47 mmol, 10 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was adjusted to pH = ~4 with HCl (1 M) and extracted with DCM (2 mL). The combined organic layers were concentrated under reduced pressure to give 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H-isoquinolin-2-yl]-2-tert- butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]propyl]-4-methyl- 1-piperidyl]acetic acid (100 mg, 126.5 µmol, 86.2% yield) as a white solid. Step I. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-4-methyl-4-piperidyl]propoxy]-2-met hyl-phenyl]pyridine-2- carboxylate To a solution of 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]propyl]-4-methyl-1- piperidyl]acetic acid (120 mg, 151.90 µmol, 1.0 equiv.) and 3-(6-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (47.08 mg, 182.28 µmol, 1.2 equiv.) in pyridine (1 mL) was added EDCI (43.68 mg, 227.85 µmol, 1.5 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was quenched by adding water (5 mL), and then filtered and concentrated under reduced pressure to give compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-diox o-3-piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-4-methyl-4-piperidyl]propoxy]-2-met hyl-phenyl]pyridine-2- carboxylate (100 mg, 97.0 µmol, 63.9% yield) as a yellow solid. MS (ESI) m/z: 1030.4 [M+H] + Step J. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-methyl-4-piperidyl]propoxy]-2-methy l-phenyl]pyridine-2- carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-ind azol-6-yl]amino]-2-oxo-ethyl]-4- methyl-4-piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carbo xylate (100 mg, 97.06 µmol, 1.0 equiv.) in DCM (1 mL) and TFA (1 mL). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)- 1-methyl-indazol-6-yl]amino]-2- oxo-ethyl]-4-methyl-4-piperidyl]propoxy]-2-methyl-phenyl]pyr idine-2-carboxylic acid (48.9 mg, 49.5 µmol, 51.0% yield, 98.6% purity) as a white solid. MS (ESI) m/z: 974.4 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.96 - 12.74 (m, 1H), 10.87 (s, 1H), 8.02 (d, J = 8.0 Hz, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.70 - 7.59 (m, 2H), 7.50 - 7.42 (m, 3H), 7.40 - 7.32 (m, 2H), 7.19 - 7.14 (m, 1H), 7.10 (t, J = 8.0 Hz, 1H), 6.97 (d, J = 8.8 Hz, 1H), 6.89 (d, J = 8.4 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.38 - 4.28 (m, 1H), 3.97 (d, J = 5.2 Hz, 2H), 3.95 - 3.88 (m, 5H), 3.23 - 3.13 (m, 2H), 3.03 (t, J = 6.0 Hz, 2H), 2.72 - 2.55 (m, 5H), 2.37 - 2.30 (m, 1H), 2.23 - 2.12 (m, 1H), 1.92 (s, 3H), 1.82 - 1.37 (m, 9H), 0.97 (s, 3H). Example 72. Preparation of Compound 272a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-((2S)-8-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino )-2-oxoethyl)-8- azaspiro[4.5]decan-2-yl)ethoxy)-2-methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(3-(2-((2S)-8- (2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)am ino)-2-oxoethyl)-8- azaspiro[4.5]decan-2-yl)ethoxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of tert-butyl (E)-2-(2-ethoxy-2-oxoethylidene)-8- azaspiro[4.5]decane-8-carboxylate To a solution of NaH (331.54 mg, 8.29 mmol, 60% purity, 2.1 equiv.) in THF (10 mL) was added dropwise ethyl 2-(diethoxyphosphoryl)acetate (1.77 g, 7.89 mmol, 1.57 mL, 2 equiv.) in THF (5 mL) at 0 °C for 15 min under N2 atmosphere. Then tert-butyl 2-oxo-8- azaspiro[4.5]decane-8-carboxylate (1 g, 3.95 mmol, 1 equiv.) was added at 0 °C under N2 atmosphere and the mixture was stirred for 2 hours. The reaction mixture was quenched by addition NH 4 Cl (15 mL) at 0 °C, and then diluted with H 2 O (10 mL) and extracted with ethyl acetate 30 mL (10 mL × 3). The combined organic layers were washed with NaHCO330 mL (10 mL × 3), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 20% ethyl acetate / petroleum ether) to give tert-butyl (E)-2-(2-ethoxy-2-oxoethylidene)-8- azaspiro[4.5]decane-8-carboxylate (0.8 g, 2.4 mmol, 62.6% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 5.76 (s, 1H), 4.05 (q, J = 7.2 Hz, 2H), 3.32 - 3.15 (m, 5H), 2.76 - 2.72 (m, 1H), 2.64 - 2.51 (m, 1H), 2.37 (s, 1H), 1.74 - 1.52 (m, 2H), 1.42 - 1.29 (m, 13H), 1.22 - 1.14 (m, 3H) Step B. Procedure for preparation of tert-butyl 2-(2-ethoxy-2-oxoethyl)-8- azaspiro[4.5]decane-8-carboxylate A mixture of tert-butyl (E)-2-(2-ethoxy-2-oxoethylidene)-8-azaspiro[4.5]decane-8- carboxylate (0.8 g, 2.47 mmol, 1 equiv.), Pd/C (0.2 g, 10% purity) in EtOH (10 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 2 hours under H2 (15 Psi) atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was used for next step without other purification. The compound tert-butyl 2-(2-ethoxy-2-oxoethyl)-8-azaspiro[4.5]decane-8-carboxylate (800 mg, 2.1 mmol, 88.5% yield, 89% purity) was obtained as a colorless oil. MS (ESI) m/z: 348.2 [M+22] + . Step C. Procedure for preparation of tert-butyl 2-(2-hydroxyethyl)-8- azaspiro[4.5]decane-8-carboxylate To a solution of tert-butyl 2-(2-ethoxy-2-oxoethyl)-8-azaspiro[4.5]decane-8-carboxylate (800 mg, 2.46 mmol, 1 equiv.) in THF (10 mL) was added LAH (111.96 mg, 2.95 mmol, 1.2 equiv.) under 0 °C. The mixture was stirred at 0 °C for 2 hours. The reaction mixture was quenched by addition Na 2 SO 4 •10H 2 O (300mg) under 0 °C and N 2 atmosphere, and then filtered and concentrated under reduced pressure to give a residue. The residue was used for next step without other purification. The compound tert-butyl 2-(2-hydroxyethyl)-8- azaspiro[4.5]decane-8-carboxylate (800 mg, 2.1 mmol, 86.2% yield, 75% purity) was obtained as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 4.33 (d, J = 3.2 Hz, 1H), 3.37 (t, J = 6.8 Hz, 2H), 3.28 - 3.23 (m, 3H), 1.98 - 1.90 (m, 1H), 1.78 - 1.67 (m, 2H), 1.59 - 1.39 (m, 4H), 1.38 (s, 9H), 1.36 - 1.26 (m, 5H), 1.20 - 1.12 (m, 1H), 0.91 (dd, J = 10.0, 12.8 Hz, 1H) Step D. Procedure for preparation of tert-butyl 2-(2-(3-bromo-2-methylphenoxy)ethyl)-8- azaspiro[4.5]decane-8-carboxylate A mixture of tert-butyl 2-(2-hydroxyethyl)-8-azaspiro[4.5]decane-8-carboxylate (750.06 mg, 2.65 mmol, 1.1 equiv.), 3-bromo-2-methyl-phenol (450 mg, 2.41 mmol, 1 equiv.), 2-(tributyl- λ 5 -phosphanylidene)acetonitrile (871.04 mg, 3.61 mmol, 1.5 equiv.) in toluene (8 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 120 °C for 10 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 20% ethyl acetate / petroleum ether) to give tert-butyl 2-(2-(3-bromo-2- methylphenoxy)ethyl)-8-azaspiro[4.5]decane-8-carboxylate (800 mg, 1.3 mmol, 55.1% yield, 75% purity) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.17 - 7.12 (m, 1H), 7.11 - 7.05 (m, 1H), 6.99 - 6.95 (m, 1H), 3.98 (t, J = 6.4 Hz, 2H), 3.26 (d, J = 4.4 Hz, 4H), 2.23 (s, 3H), 2.14 - 2.02 (m, 1H), 1.83 - 1.72 (m, 4H), 1.43 - 1.28 (m, 16H), 1.10 - 0.99 (m, 1H) Step E. Procedure for preparation of 2-(2-(3-bromo-2-methylphenoxy)ethyl)-8- azaspiro[4.5]decane A solution of tert-butyl 2-(2-(3-bromo-2-methylphenoxy)ethyl)-8-azaspiro[4.5]decane-8 - carboxylate (800 mg, 1.33 mmol, 75% purity, 1 equiv.) in HCl / dioxane (4 M, 8 mL) was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was used for next step without other purification. The compound 2-(2-(3-bromo-2-methylphenoxy)ethyl)-8-azaspiro[4.5]decane (300 mg, 771.7 µmol, 58.1% yield, HCl) was obtained as a white solid. MS (ESI) m/z: 352.0 [M+H] + . Step F. Procedure for preparation of ethyl 2-[3-[2-(3-bromo-2-methyl-phenoxy)ethyl]-8- azaspiro[4.5]decan-8-yl]acetate A mixture of 2-(2-(3-bromo-2-methylphenoxy)ethyl)-8-azaspiro[4.5]decane (300 mg, 851.53 µmol, 1 equiv.), ethyl 2-bromoacetate (142.21 mg, 851.53 µmol, 94.18 µL, 1 equiv.), K2CO3 (353.06 mg, 2.55 mmol, 3 equiv.), KI (70.68 mg, 425.76 µmol, 0.5 equiv.) in CH 3 CN (5 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 60 °C for 2 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 10% ethyl acetate / petroleum ether). The compound ethyl 2-[3-[2-(3-bromo-2-methyl- phenoxy)ethyl]-8-azaspiro[4.5]decan-8-yl]acetate (200 mg, 452.8 µmol, 53.1% yield, 99.2% purity) was obtained as a colorless oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.16 - 7.11 (m, 1H), 7.11 - 7.04 (m, 1H), 6.95 (d, J = 8.0 Hz, 1H), 4.06 (q, J = 7.2 Hz, 2H), 3.96 (t, J = 6.4 Hz, 2H), 3.19 - 3.15 (m, 2H), 2.47 - 2.37 (m, 4H), 2.22 (s, 3H), 2.10 - 2.01 (m, 1H), 1.82 - 1.69 (m, 4H), 1.46 - 1.33 (m, 6H), 1.27 - 1.21 (m, 1H), 1.17 (t, J = 7.2 Hz, 3H), 0.98 (dd, J = 10.2, 12.4 Hz, 1H) Step G. Procedure for preparation of ethyl (S)-2-(2-(2-(3-bromo-2-methylphenoxy)ethyl)- 8-azaspiro[4.5]decan-8-yl)acetate The compound ethyl 2-[3-[2-(3-bromo-2-methyl-phenoxy)ethyl]-8-azaspiro[4.5]deca n-8- yl]acetate (200 mg, 452.88 µmol, 53.18% yield, 99.27% purity) was purified by SFC to give ethyl (R)-2-(2-(2-(3-bromo-2-methylphenoxy)ethyl)-8-azaspiro[4.5]d ecan-8-yl)acetate (50 mg, 114.05 µmol, 25.00% yield) as a colorless oil, and ethyl (S)-2-(2-(2-(3-bromo-2- methylphenoxy)ethyl)-8-azaspiro[4.5]decan-8-yl)acetate (66 mg, 150.5 µmol, 33.0% yield) as a colorless oil. Step H. Procedure for preparation of tert-butyl (S)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(8-(2-ethoxy-2-oxoe thyl)-8-azaspiro[4.5]decan- 2-yl)ethoxy)-2-methylphenyl)picolinate A mixture of ethyl (S)-2-(2-(2-(3-bromo-2-methylphenoxy)ethyl)-8-azaspiro[4.5]d ecan-8- yl)acetate (66 mg, 150.55 µmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3, 2-dioxaborolan-2-yl)picolinate (129.10 mg, 210.77 µmol, 1.4 equiv.), [2-(2-aminophenyl)phenyl]palladium(1 + );bis(1- adamantyl)-butyl-phosphane;methanesulfonate (21.93 mg, 30.11 µmol, 0.2 equiv.), K2CO3 (1.5 M, 150.55 µL, 1.5 equiv.) in dioxane (0.7 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 100 °C for 1 hour under M.W. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 50% ethyl acetate / petroleum ether) to give tert-butyl (S)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquino lin-2(1H)-yl)- 3-(3-(2-(8-(2-ethoxy-2-oxoethyl)-8-azaspiro[4.5]decan-2-yl)e thoxy)-2- methylphenyl)picolinate (120 mg, 142.1 µmol, 94.4% yield) as a colorless oil. MS (ESI) m/z: 844.4 [M+H] + . Step I. Procedure for preparation of (S)-2-(2-(2-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)ethyl)-8-azaspiro[4.5]decan-8-yl)acetic acid A mixture of tert-butyl (S)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquino lin- 2(1H)-yl)-3-(3-(2-(8-(2-ethoxy-2-oxoethyl)-8-azaspiro[4.5]de can-2-yl)ethoxy)-2- methylphenyl)picolinate (120 mg, 142.17 µmol, 1 equiv.), LiOH•H2O (17.90 mg, 426.51 µmol, 3 equiv.) in THF (3 mL) and H 2 O (1 mL) was stirred at 25 °C for 10 hours. The mixture was concentrated, and the pH was adjusted to 3, then triturated and filtered to obtain a residue. The residue was used for next step without other purification. The compound (S)-2-(2-(2-(3- (6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)ethyl)-8-azaspi ro[4.5]decan-8-yl)acetic acid (100 mg, 122.5 µmol, 86.2% yield) was obtained as a white solid. MS (ESI) m/z: 816.4 [M+H] + . Step J. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[(3S)-8-[2-[[3-(2,6 -dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-8-azaspiro[4.5]decan-3-yl]e thoxy]-2-methyl- phenyl]pyridine-2-carboxylate A mixture of (S)-2-(2-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)ethyl)-8- azaspiro[4.5]decan-8-yl)acetic acid (100 mg, 122.55 µmol, 1 equiv.), 3-(6-amino-1-methyl- 1H-indazol-3-yl)piperidine-2,6-dione (37.98 mg, 147.06 µmol, 1.2 equiv.), EDCI (35.24 mg, 183.82 µmol, 1.5 equiv.) in pyridine (1 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 2 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was used for next step without other purification. The compound tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[(3S)- 8-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]-8-azasp iro[4.5]decan-3-yl]ethoxy]-2- methyl-phenyl]pyridine-2-carboxylate (100 mg, 94.7 µmol, 77.2% yield) was obtained as a colorless oil. MS (ESI) m/z: 1056.4 [M+H] + . Step K. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((2S)-8-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-8-azaspiro[4.5]decan-2-yl )ethoxy)-2- methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((2S)-8-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-8-azaspiro[4.5]decan-2-yl )ethoxy)-2- methylphenyl)picolinic acid] A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[2-[(3S)-8-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo- ethyl]-8-azaspiro[4.5]decan-3-yl]ethoxy]-2-methyl-phenyl]pyr idine-2-carboxylate (100 mg, 94.67 µmol, 1 equiv.) in TFA (0.5 mL) and DCM (0.5 mL) was stirred at 25 °C for 10 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((2R)-8-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)-8-azaspiro[4.5]decan-2-yl)et hoxy)-2- methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((2S)-8-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)-8-azaspiro[4.5]decan-2-yl)et hoxy)-2- methylphenyl)picolinic acid] (14.8 mg, 14.5 µmol, 15.3% yield, 97.9% purity) as a yellow solid. MS (ESI) m/z: 1000.4 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.98 - 12.71 (m, 1H), 10.87 (s, 1H), 9.95 - 9.69 (m, 1H), 8.06 - 8.00 (m, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.66 - 7.59 (m, 2H), 7.49 - 7.21 (m, 5H), 7.22 (s, 1H), 7.09 (t, J = 8.0 Hz, 1H), 6.96 (d, J = 9.2 Hz, 1H), 6.89 (d, J = 8.0 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.35 - 4.28 (m, 1H), 4.00 - 3.88 (m, 8H), 3.17 - 2.99 (m, 4H), 2.67 - 2.60 (m, 4H), 2.39 - 2.29 (m, 2H), 2.21 - 2.08 (m, 2H), 1.89 (s, 3H), 1.85 - 1.75 (m, 4H), 1.60 - 1.43 (m, 6H), 1.39 - 1.20 (m, 2H) Example 73. Preparation of Compound 273b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1R,4r)-4-(3- ((2S)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)-2-methylpiperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl (2S)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1- methyl-indazol-6-yl]-2-methyl-piperazine-1-carboxylate A mixture of 6-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (1.5 g, 3.00 mmol, 1.0 equiv.), tert-butyl (2S)-2-methylpiperazine-1-carboxylate (1.80 g, 9.00 mmol, 3.0 equiv.), RuPhos (279.77 mg, 600.00 µmol, 0.2 equiv.), Cs 2 CO 3 (2.93 g, 9.00 mmol, 3.0 equiv.), and Pd 2 (dba) 3 (274.50 mg, 300.00 µmol, 0.1 equiv.) in toluene (15 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 110 °C for 16 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to remove toluene. The resulting residue was purified by flash silica gel chromatography (Eluent of 16% ethyl acetate/petroleum ether) to give tert-butyl (2S)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl- indazol-6-yl]-2-methyl-piperazine-1-carboxylate (1.2 g, 1.8 mmol, 61.1% yield, 94.7% purity) was obtained as brown oil. MS (ESI) m/z: 620.6 [M+H] + . Step B. Procedure for preparation of tert-butyl (2S)-4-[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]-2-methyl-piperazine-1-carboxylate A mixture of tert-butyl (2S)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-yl] -2- methyl-piperazine-1-carboxylate (1.20 g, 1.94 mmol, 1 equiv.), Pd/C (300 mg, 1.94 mmol, 10% purity, 1.00 equiv.), Pd(OH) 2 (300 mg, 2.14 mmol, 1.10 equiv.), and AcOH (116.28 mg, 1.94 mmol, 110.74 uL, 1.0 equiv.) in THF (10 mL) and EtOH (10 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 16 hours under N2 (15 Psi) atmosphere. The mixture was filtered, and filter cake was washed by THF (20 mL). The filtrate was concentrated under reduced pressure to give tert-butyl (2S)-4-[3-(2,6-dioxo-3-piperidyl)- 1-methyl-indazol-6-yl]-2-methyl-piperazine-1-carboxylate (900 mg, 1.4 mmol, 74.5% yield, 70.8% purity) as brown oil. MS (ESI) m/z: 442.2 [M+H] + . Step C. Procedure for preparation of 3-[1-methyl-6-[(3S)-3-methylpiperazin-1- yl]indazol-3-yl]piperidine-2,6-dione A mixture of tert-butyl (2S)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]-2-m ethyl- piperazine-1-carboxylate (900.00 mg, 2.04 mmol, 1 equiv.) in a solvent of DCM (5 mL) and HCl/dioxane (10 mL) was stirred at 25 °C for 2 hours. The reaction mixture was treated with EtOAc (20 mL) and then filtered. The filtrate was concentrated under reduced pressure to give 3-[1-methyl-6-[(3S)-3-methylpiperazin-1-yl]indazol-3-yl]pipe ridine-2,6-dione (600 mg, 1.7 mmol, 86.2% yield) as a greyish-green solid. MS (ESI) m/z: 342.1 [M+H] + . Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[(2S)-4-[3-(2,6- dioxo-3-piperidyl)-1-methyl- indazol-6-yl]-2-methyl-piperazin-1-yl]propyl]cyclohexoxy]-2- methyl-phenyl]pyridine-2- carboxylate A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[2-methyl-3-[4-(3-oxopropyl)cyclohexoxy]phenyl]pyridin e-2-carboxylate (150 mg, 205.22 µmol, 1.0 equiv.) and 3-[1-methyl-6-[(3S)-3-methylpiperazin-1-yl]indazol-3- yl]piperidine-2,6-dione (84.08 mg, 246.27 µmol, 1.2 equiv.), NaBH(OAc)3 (130.49 mg, 615.67 µmol, 3.0 equiv.) in DCM (3 mL), and then the mixture was stirred at 25 °C for 3.5 hours. The reaction mixture was diluted with water (20 mL) and extracted with ethyl acetate (20 mL × 3). The combined organic layers were washed with brine (10 mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep- TLC (SiO 2 , DCM : MeOH = 10:1) to give compound tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[(2 S)-4-[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol-6-yl]-2-methyl-piperazin-1-yl]pr opyl]cyclohexoxy]-2-methyl- phenyl]pyridine-2-carboxylate (60 mg, 55.8 µmol, 27.2% yield, 98.3% purity) as a brown solid. MS (ESI) m/z: 1057.6 [M+H] + . Step E. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-(3-((2S)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-2-methylpiperazin-1-yl)propyl)cycloh exyl)oxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[4-[3-[(2S)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl -indazol-6-yl]-2-methyl- piperazin-1-yl]propyl]cyclohexoxy]-2-methyl-phenyl]pyridine- 2-carboxylate (50 mg, 47.33 µmol, 1.0 equiv.) in DCM (1 mL) was added TFA (1 mL).The mixture was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to remove DCM and TFA to give a residue. The crude product was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1R,4r)-4-(3-((2S)- 4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)-2-m ethylpiperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (31.1 mg, 27.9 µmol, 58.9% yield, 89.5% purity) as a brown solid. MS (ESI) m/z: 1000.5 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.95 - 12.79 (m, 1H), 10.90 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 7.2 Hz, 1H), 7.62 (d, J = 6.0 Hz, 1H), 7.51 - 7.42 (m, 4H), 7.40 - 7.29 (m, 2H), 7.12 - 7.04 (m, 3H), 6.95 (dd, J = 8.4, 13.8 Hz, 2H), 6.66 - 6.58 (m, 1H), 4.98 (s, 2H), 4.36 (dd, J = 5.2, 9.6 Hz, 1H), 4.26 (s, 4H), 3.94 - 3.88 (m, 2H), 3.74 - 3.56 (m, 2H), 3.03 (t, J = 5.6 Hz, 4H), 2.93 - 2.84 (m, 1H), 2.68 - 2.61 (m, 2H), 2.54 (s, 6H), 2.37 - 2.29 (m, 2H), 2.19 - 2.05 (m, 4H), 1.91 - 1.79 (m, 6H), 1.38 - 1.26 (m, 6H), 1.15 - 1.06 (m, 2H). Example 74. Preparation of Compound 274a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1S,4r)-4-(3- ((3R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-y l)-3-methylpiperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl (3R)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1- methyl-indazol-7-yl]-3-methyl-piperazine-1-carboxylate A mixture of 7-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (1.5 g, 3.00 mmol, 1 equiv.), tert-butyl (3R)-3-methylpiperazine-1-carboxylate (900.55 mg, 4.50 mmol, 1.5 equiv.), Cs2CO3 (2.93 g, 8.99 mmol, 3 equiv.) and 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5-dichloro- 2H-imidazol-1-ium-2-ide;3-chloropyridine;dichloropalladium (145.80 mg, 149.88 µmol, 0.05 equiv.) in 2-methyl-2-butanol (15 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 100 °C for 16 hours under N2 atmosphere. The reaction mixture was filtered and diluted with H2O (30 mL) and extracted with EtOAc (30 mL×3). The combined organic layers were washed with brine (50 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~30% ethyl acetate/petroleum ether) to afford tert-butyl (3R)-4- [3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-7-yl]-3-meth yl-piperazine-1-carboxylate (260 mg, 385.9 µmol, 12.8% yield, 92% purity) as a light yellow oil. MS (ESI) m/z: 620.4 [M+H] + . Step B. Procedure for preparation of tert-butyl (3R)-4-[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-7-yl]-3-methyl-piperazine-1-carboxylate To a solution of tert-butyl (3R)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-7-yl] -3- methyl-piperazine-1-carboxylate (300 mg, 484.07 µmol, 1 equiv.) in THF (5 mL) and EtOH (5 mL) was added Pd/C (100 mg, 484.07 µmol, 10% purity), Pd(OH)2 (100 mg, 71.20 µmol, 10% purity) and AcOH (87.21 mg, 1.45 mmol, 83.05 µL, 3 equiv.) under N2 atmosphere. The suspension was degassed and purged with H 2 three times. The mixture was stirred under H 2 (15 Psi) at 25 °C for 16 hours. The reaction mixture was filtered was washed with THF (30 mL). The filtrate was concentrated to afford tert-butyl (3R)-4-[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-7-yl]-3-methyl-piperazine-1-carboxylate (300 mg, crude) as a black brown oil, which was used in the next step without further purification. Step C. Procedure for preparation of 3-[1-methyl-7-[(2R)-2-methylpiperazin-1- yl]indazol-3-yl]piperidine-2,6-dione To a solution of tert-butyl (3R)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]-3-m ethyl- piperazine-1-carboxylate (300 mg, 679.47 µmol, 1 equiv.) in dioxane (5 mL) was added HCl/EtOAc (4 M, 169.87 µL, 1 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was filtered to afford 3-[1-methyl-7-[(2R)-2-methylpiperazin-1-yl]indazol-3- yl]piperidine-2,6-dione (150 mg, crude) as a light yellow solid, which was used in the next step without further purification. Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[(3R)-4-[3-(2,6- dioxo-3-piperidyl)-1-methyl- indazol-7-yl]-3-methyl-piperazin-1-yl]propyl]cyclohexoxy]-2- methyl-phenyl]pyridine-2- carboxylate To a solution of 3-[1-methyl-7-[(2R)-2-methylpiperazin-1-yl]indazol-3-yl]pipe ridine-2,6- dione (77.07 mg, 225.75 µmol, 1.1 equiv.) in DCM (2 mL) was added tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[2-methyl-3-[4-(3- oxopropyl)cyclohexoxy]phenyl]pyridine-2-carboxylate (150 mg, 205.22 µmol, 1 equiv.). The mixture was stirred at 25 °C for 1 hour, and then treated with NaBH(OAc) 3 (130.49 mg, 615.67 µmol, 3 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was filtered and concentrated to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH = 15:1) to afford tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[4-[3-[(3R)-4-[3-(2,6-dioxo-3-piperid yl)-1-methyl-indazol-7-yl]-3- methyl-piperazin-1-yl]propyl]cyclohexoxy]-2-methyl-phenyl]py ridine-2-carboxylate (140 mg, 107.3 µmol, 52.3% yield, 81% purity) as a light yellow solid MS (ESI) m/z: 529.1 [M/2+H] + . Step E. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-(3-((3R)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)-3-methylpiperazin-1-yl)propyl)cycloh exyl)oxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[4-[3-[(3R)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl -indazol-7-yl]-3-methyl- piperazin-1-yl]propyl]cyclohexoxy]-2-methyl-phenyl]pyridine- 2-carboxylate (140 mg, 132.54 µmol, 1 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 101.91 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to afford 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1S,4r)-4-(3-((3R)- 4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)-3-m ethylpiperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (42.9 mg, 38.9 µmol, 29.3% yield, 90.6% purity) as a white solid. MS (ESI) m/z: 501.1[M/2+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.88 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.50 - 7.42 (m, 4H), 7.39 - 7.32 (m, 2H), 7.27 - 7.20 (m, 1H), 7.11 - 7.04 (m, 2H), 6.98 - 6.90 (m, 2H), 6.61 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.36 - 4.32 (m, 1H), 4.27 (s, 3H), 4.23 - 4.16 (m, 1H), 3.94 - 3.81 (m, 2H), 3.05 - 3.00 (m, 3H), 2.90 - 2.84 (m, 1H), 2.82 - 2.73 (m, 2H), 2.68 - 2.61 (m, 3H), 2.39 - 2.28 (m, 4H), 2.21 - 2.15 (m, 1H), 2.11 - 2.05 (m, 2H), 1.95 - 1.90 (m, 1H), 1.87 (s, 3H), 1.84 - 1.77 (m, 2H), 1.53 - 1.46 (m, 2H), 1.41 -1.33 (m, 2H), 1.29 - 1.20 (m, 3H), 1.12 - 1.02 (m, 2H), 0.87 - 0.77 (m, 3H) Example 75. Preparation of Compound 274b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1R,4r)-4-(3- ((3S)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-y l)-3-methylpiperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl (3S)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1- methyl-indazol-7-yl]-3-methyl-piperazine-1-carboxylate A mixture of 7-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (1.7 g, 3.40 mmol,1.0 equiv.), tert-butyl (3S)-3-methylpiperazine-1-carboxylate (1.02 g, 5.10 mmol, 1.5 equiv.), Cs 2 CO 3 (3.32 g, 10.19 mmol, 3.0 equiv.), and 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5- dichloro-2H-imidazol-1-ium-2-ide;3-chloropyridine; dichloro palladium (165.24 mg, 169.87 µmol, 0.05 equiv.) in 2-methyl-2-butanol (15 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 100 °C for 16 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was diluted with H2O (100 mL) and extracted with ethyl acetate (100 mL × 3). The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give tert-butyl (3S)-4-[3-(2,6- dibenzyloxy-3-pyridyl)-1-methyl-indazol-7-yl]-3-methyl-piper azine-1-carboxylate (300 mg, 371.8 µmol, 10.9% yield, 76.8% purity) as yellow oil. MS (ESI) m/z: 620.4 [M+H] + Step B. Procedure for preparation of tert-butyl (3S)-4-[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-7-yl]-3-methyl-piperazine-1-carboxylate To a solution of tert-butyl (3S)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-7-yl] -3- methyl-piperazine-1-carboxylate (300 mg, 484.07 µmol, 1.0 equiv.) in MeOH (10 mL) was added Pd/C (50 mg, 48.41 µmol, 10% purity, 0.1 equiv.) and Pd(OH) 2 (67.98 mg, 48.41 µmol, 10% purity, 0.1 equiv.) under N 2 . The suspension was degassed under vacuum and purged with H2 several times. The mixture was stirred under H2 (15 psi) at 25 °C for 16 hours. After the completion of the reaction, the reaction solution was filtered through celite under a moderate N 2 atmosphere and concentrated to give tert-butyl (3S)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl- indazol-7-yl]-3-methyl-piperazine-1-carboxylate (132 mg, 298.9 µmol, 61.7% yield) as a yellow solid. MS (ESI) m/z: 442.3 [M+H] + Step C. Procedure for preparation of 3-[1-methyl-7-[(2S)-2-methylpiperazin-1- yl]indazol-3-yl]piperidine-2,6-dione To a solution of tert-butyl (3S)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]-3-m ethyl- piperazine-1-carboxylate (200 mg, 452.98 µmol, 1.0 equiv.) in HCl/dioxane (10 mL) and dioxane (10 mL). The mixture was stirred at 25 °C for 2 hours. The mixture was filtered to give 3-[1-methyl-7-[(2S)-2-methylpiperazin-1-yl]indazol-3-yl]pipe ridine-2,6-dione (132 mg, 349.4 µmol, 77.1% yield, 90.3% purity) as yellow oil which was used in the next step without further purification. MS (ESI) m/z: 342.2 [M+H] + Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[(3S)-4-[3-(2,6- dioxo-3-piperidyl)-1-methyl- indazol-7-yl]-3-methyl-piperazin-1-yl]propyl]cyclohexoxy]-2- methyl-phenyl]pyridine-2- carboxylate To a solution of 3-[1-methyl-7-[(2S)-2-methylpiperazin-1-yl]indazol-3-yl]pipe ridine-2,6- dione (90 mg, 263.62 µmol, 1.0 equiv.) in DCM (5 mL) was added dropwise NaBH(OAc) 3 (167.61 mg, 790.85 µmol, 3.0 equiv.) at 25 °C. After addition, the mixture was stirred at this temperature for 0.5 hour, and then tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[2-methyl-3-[4-(3-oxopropyl)c yclohexoxy]phenyl]pyridine- 2-carboxylate (192.68 mg, 263.62 µmol, 1.0 equiv.) was added dropwise at 0°C. The resulting mixture was stirred at 25°C for 16 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was diluted with H2O (50 mL) and extracted with ethyl acetate (50 mL × 3). The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[(3 S)-4-[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol-7-yl]-3-methyl-piperazin-1-yl]pr opyl]cyclohexoxy]-2-methyl- phenyl]pyridine-2-carboxylate (128 mg, 119.9 µmol, 45.5% yield, 99% purity) as a brown solid. MS (ESI) m/z: 529.1 [M/2+H] + Step E. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-(3-((3S)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)-3-methylpiperazin-1-yl)propyl)cycloh exyl)oxy)-2- methylphenyl)picolinic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[4-[3-[(3S)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-i ndazol-7-yl]-3-methyl-piperazin- 1-yl]propyl]cyclohexoxy]-2-methyl-phenyl]pyridine-2-carboxyl ate (95 mg, 89.93 µmol, 1.0 equiv.) in TFA (3 mL) and DCM (6 mL) was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep- HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1R,4r)-4-(3-((3S)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl -1H-indazol-7-yl)-3- methylpiperazin-1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl)p icolinic acid (56.45 mg, 56.44 µmol, 62.75% yield) as a yellow solid. MS (ESI) m/z: 1001.3 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 10.88 (s, 1H), 8.15 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.49 - 7.42 (m, 4H), 7.39 - 7.33 (m, 2H), 7.24 (d, J = 5.6 Hz, 1H), 7.07 ( t, J = 7.6 Hz, 2H), 6.94 (dd, J = 8.8, 12.8 Hz, 2H), 6.62 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.34 (dd, J = 5.2, 10.6 Hz, 1H), 4.27 (s, 3H), 4.23 - 4.17 (m, 1H), 3.91 (t, J = 6.0 Hz, 2H), 3.02 (t, J = 5.6 Hz, 2H), 2.88 - 2.84 (m, 1H), 2.81 - 2.73 (m, 2H), 2.70 - 2.61 (m, 4H), 2.33 (d, J = 1.6 Hz, 3H), 2.29 - 2.23 (m, 1H), 2.17 (dd, J = 5.2, 13.2 Hz, 1H), 2.11 - 2.05 (m, 2H), 1.94 - 1.85 (m, 4H), 1.80 (d, J = 10.4 Hz, 2H), 1.53 - 1.46 (m, 2H), 1.40 - 1.33 (m, 2H), 1.29 - 1.21 (m, 3H), 1.13 - 1.04 (m, 2H), 0.86 - 0.77 (m, 3H). Example 76. Preparation of Compound 275a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1S,4r)-4-(3- ((2R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-y l)-2-methylpiperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl (2R)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1- methyl-indazol-7-yl]-2-methyl-piperazine-1-carboxylate A mixture of 7-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (1.5 g, 3.00 mmol, 1 equiv.), tert-butyl (2R)-2-methylpiperazine-1-carboxylate (1.80 g, 8.99 mmol, 3 equiv.), RuPhos (279.77 mg, 599.54 µmol, 0.2 equiv.), Cs2CO3 (2.93 g, 8.99 mmol, 3 equiv.), and Pd 2 (dba) 3 (274.50 mg, 299.77 µmol, 0.1 equiv.) in toluene (15 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 110 °C for 16 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~15% ethyl acetate/petroleum ether) to afford tert-butyl (2R)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl- indazol-7-yl]-2-methyl-piperazine-1-carboxylate (1 g, 1.4 mmol, 49.5% yield, 92% purity) as a yellow oil. MS (ESI) m/z: 620.4 [M+H] + . Step B. Procedure for preparation of tert-butyl (2R)-4-[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-7-yl]-2-methyl-piperazine-1-carboxylate To a solution of tert-butyl (2R)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-7-yl] -2- methyl-piperazine-1-carboxylate (1.14 g, 1.84 mmol, 1 equiv.) in THF (10 mL) and EtOH (10 mL) was added Pd/C (300 mg, 1.84 mmol, 10% purity, 1.00 equiv.), Pd(OH)2 (300 mg, 213.61 µmol, 10% purity, 1.16e-1 equiv.), and AcOH (110.46 mg, 1.84 mmol, 105.20 µL, 1 equiv.) under N2 atmosphere. The suspension was degassed and purged with H2 three times. The mixture was stirred under H2 (15 Psi) at 25 °C for 16 hours. The reaction mixture was filtered, and the filter cake was washed with THF (50 mL). The filtrate was concentrated to afford tert- butyl (2R)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]-2-m ethyl-piperazine-1- carboxylate (900 mg, crude) as a black, brown oil, which was used in the next step without further purification. Step C. Procedure for preparation of 3-[1-methyl-7-[(3R)-3-methylpiperazin-1- yl]indazol-3-yl]piperidine-2,6-dione To a solution of tert-butyl (2R)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]-2-m ethyl- piperazine-1-carboxylate (900 mg, 2.04 mmol, 1 equiv.) in dioxane (10 mL) was added HCl/EtOAc (4 M, 509.60 uL, 1 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was filtered to afford 3-[1-methyl-7-[(3R)-3-methylpiperazin-1-yl]indazol-3- yl]piperidine-2,6-dione (550 mg, crude) as a light-yellow solid, was used in the next step without further purification. Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[(2R)-4-[3-(2,6- dioxo-3-piperidyl)-1-methyl- indazol-7-yl]-2-methyl-piperazin-1-yl]propyl]cyclohexoxy]-2- methyl-phenyl]pyridine-2- carboxylate To a solution of 3-[1-methyl-7-[(3R)-3-methylpiperazin-1-yl]indazol-3-yl]pipe ridine-2,6- dione (84.08 mg, 246.27 µmol, 1.2 equiv.) in DCM (2 mL) was added tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[2-methyl-3-[4-(3- oxopropyl)cyclohexoxy]phenyl]pyridine-2-carboxylate (150 mg, 205.22 µmol, 1 equiv.). The mixture was stirred at 25 °C for 1 hour and then treated with NaBH(OAc)3 (130.49 mg, 615.67 µmol, 3 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep- TLC (SiO2, DCM: MeOH = 15:1) to afford tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[(2R)-4-[3-(2,6- dioxo-3-piperidyl)-1-methyl- indazol-7-yl]-2-methyl-piperazin-1-yl]propyl]cyclohexoxy]-2- methyl-phenyl]pyridine-2- carboxylate (30 mg, 24.4 µmol, 11.9% yield, 86% purity) as a light yellow solid. MS (ESI) m/z: 1056.5 [M+H] + . Step E. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-(3-((2R)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)-2-methylpiperazin-1-yl)propyl)cycloh exyl)oxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[4-[3-[(2R)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl -indazol-7-yl]-2-methyl- piperazin-1-yl]propyl]cyclohexoxy]-2-methyl-phenyl]pyridine- 2-carboxylate (30 mg, 28.40 µmol, 1 equiv.) in DCM (0.5 mL) was added TFA (770.00 mg, 6.75 mmol, 0.5 mL, 237.78 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to afford 6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2( 1H)-yl)-3-(3-(((1S,4r)-4-(3- ((2R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-y l)-2-methylpiperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (18.9 mg, 18.8 µmol, 66.3% yield, 99.6% purity) as a yellow solid. MS (ESI) m/z: 1000.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.88 (s, 1H), 8.02 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 12.6 Hz, 1H), 7.61 (d, J = 7.2 Hz, 1H), 7.51 - 7.30 (m, 6H), 7.10 - 6.98 (m, 3H), 6.97 - 6.87 (m, 2H), 6.62 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.37 - 4.30 (m, 1H), 4.24 (s, 3H), 4.21 - 4.13 (m, 1H), 3.96 - 3.85 (m, 2H), 3.16 - 3.07 (m, 2H), 3.06 - 2.98 (m, 3H), 2.97 - 2.90 (m, 1H), 2.88 - 2.73 (m, 2H), 2.70 - 2.60 (m, 3H), 2.36-2.28 (m, 2H), 2.20 - 2.13 (m, 1H), 2.11 - 2.04 (m, 2H), 1.87 (s, 3H), 1.80 (d, J = 11.6 Hz, 2H), 1.54 - 1.16 (m, 8H), 1.13 - 1.02 (m, 5H) Example 77. Preparation of Compound 276a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4-[(1R)-3-[1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-o xo-ethyl]-4-piperidyl]-1- methyl-propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(4-(((2R)-4- (1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl )amino)-2-oxoethyl)piperidin-4- yl)butan-2-yl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of tert-butyl 4-[3-[methoxy(methyl)amino]-3-oxo- propyl]piperidine-1-carboxylate To a solution of 3-(1-tert-butoxycarbonyl-4-piperidyl)propanoic acid (2 g, 7.77 mmol, 1 equiv.) in DMF (25 mL) was added HATU (4.43 g, 11.66 mmol, 1.5 equiv.), DIPEA (5.02 g, 38.86 mmol, 6.77 mL, 5 equiv.), and N-methoxymethanamine (1.14 g, 11.66 mmol, 1.5 equiv., HCl). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give the compound tert-butyl 4-[3-[methoxy(methyl)amino]-3-oxo-propyl]piperidine-1- carboxylate (1.7 g, 5.6 mmol, 72.8% yield) as a yellow oil. 1 H NMR (400 MHz, CDCl3) δ = 4.08 (d, J = 13.2 Hz, 2H), 3.69 (s, 3H), 3.19 (s, 3H), 2.77 - 2.62 (m, 2H), 2.45 (t, J = 7.6 Hz, 2H), 1.68 (d, J = 12.4 Hz, 2H), 1.64 - 1.56 (m, 2H), 1.46 (s, 10H), 1.18 - 1.05 (m, 2H). Step B. Procedure for preparation of tert-butyl 4-(3-oxobutyl)piperidine-1-carboxylate A mixture of tert-butyl 4-[3-[methoxy(methyl)amino]-3-oxo-propyl]piperidine-1-carbox ylate (1.7 g, 5.66 mmol, 1 equiv.) in THF (10 mL) was degassed and purged with N 2 three times, and then MeMgBr (3 M, 11.32 mL, 6 equiv.) was added dropwise to the mixture and stirred at 0 °C for 1 hour under N2 atmosphere. The reaction mixture was quenched by addition of saturated ammonium chloride (10 mL) at 0 °C, and then diluted with water (10 mL) and extracted with CH2Cl2 (10 mL × 3). The combined organic layers were washed with brine (10 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give compound tert-butyl 4-(3-oxobutyl)piperidine-1-carboxylate (1.3 g, 5.0 mmol, 89.9% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ = 3.90 (d, J = 12.0 Hz, 2H), 2.69 - 2.59 (m, 2H), 2.44 (t, J = 7.2 Hz, 2H), 2.07 (s, 3H), 1.59 (d, J = 12.4 Hz, 3H), 1.38 (s, 9H), 1.35 - 1.31 (m, 1H), 0.97 - 0.87 (m, 2H). Step C. Procedure for preparation of tert-butyl 4-(3-hydroxybutyl)piperidine-1- carboxylate To a solution of tert-butyl 4-(3-oxobutyl)piperidine-1-carboxylate (1.3 g, 5.09 mmol, 1 equiv.) in EtOH (10 mL) was added NaBH4 (231.13 mg, 6.11 mmol, 1.2 equiv.). The mixture was stirred at 0 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to remove EtOH, and then diluted with EtOAc (10 mL) and NH 4 Cl (10 ml) and extracted with EtOAc (10 mL × 3). The combined organic layers were washed with brine (10 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give compound tert-butyl 4-(3-hydroxybutyl)piperidine-1-carboxylate (1.3 g, 4.5 mmol, 89.3% yield, 90% purity) as a yellow oil. 1 H NMR (400 MHz, CDCl3) δ = 4.19 - 3.96 (m, 2H), 3.85 - 3.71 (m, 1H), 2.68 (t, J = 12.4 Hz, 2H), 1.66 (d, J = 12.8 Hz, 2H), 1.53 - 1.48 (m, 1H), 1.46 (s, 9H), 1.42 - 1.23 (m, 4H), 1.23 - 1.17 (m, 3H), 1.17 - 1.03 (m, 2H). Step D. Procedure for preparation of tert-butyl 4-[3-(4-bromo-3-methyl- phenoxy)butyl]piperidine-1-carboxylate A mixture of 4-bromo-3-methyl-phenol (1.05 g, 5.60 mmol, 1.2 equiv.), tert-butyl 4-(3- hydroxybutyl)piperidine-1-carboxylate (1.2 g, 4.66 mmol, 1 equiv.), and 2-(tributyl-λ5- phosphanylidene)acetonitrile (1.35 g, 5.60 mmol, 1.2 equiv.) in toluene (10 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 120 °C for 4 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give compound tert-butyl 4-[3-(4- bromo-3-methyl-phenoxy)butyl]piperidine-1-carboxylate (1 g, 2.3 mmol, 49.4% yield, 98.3% purity) as a yellow oil. MS (ESI) m/z: 369.9 [M-56+H] + 1 H NMR (400 MHz, CDCl 3 ) δ = 7.38 (d, J = 8.8 Hz, 1H), 6.77 (d, J = 2.4 Hz, 1H), 6.63 - 6.55 (m, 1H), 4.33 - 4.22 (m, 1H), 4.08 (d, J = 13.2 Hz, 2H), 2.67 (t, J = 11.6 Hz, 2H), 2.36 (s, 3H), 1.70 - 1.60 (m, 3H), 1.46 (s, 9H), 1.43 - 1.19 (m, 7H), 1.15 - 1.03 (m, 2H). Step E. Procedure for preparation of tert-butyl 4-[(3R)-3-(4-bromo-3-methyl- phenoxy)butyl]piperidine-1-carboxylate Tert-butyl 4-[3-(4-bromo-3-methyl-phenoxy)butyl]piperidine-1-carboxylat e was further separated by SFC to give tert-butyl 4-[(3R)-3-(4-bromo-3-methyl-phenoxy)butyl]piperidine-1- carboxylate (400 mg, 920.7 µmol, 39.2% yield, 98.1% purity) as a yellow oil and tert-butyl 4- [(3S)-3-(4-bromo-3-methyl-phenoxy)butyl]piperidine-1-carboxy late (350 mg, 820.4 µmol, 34.9% yield, 99.9% purity) as a yellow oil. Step F. Procedure for preparation of 4-[(3R)-3-(4-bromo-3-methyl- phenoxy)butyl]piperidine To a solution of tert-butyl 4-[(3R)-3-(4-bromo-3-methyl-phenoxy)butyl]piperidine-1- carboxylate (400 mg, 938.12 µmol, 1 equiv.) was added HCl/EtOAc (4 M, 234.53 µL, 1 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give 4-[(3R)-3-(4-bromo-3-methyl-phenoxy)butyl]piperidine (400 mg, crude) as a yellow oil. Step G. Procedure for preparation of ethyl 2-[4-[(3R)-3-(4-bromo-3-methyl- phenoxy)butyl]-1-piperidyl]acetate To a solution of 4-[(3R)-3-(4-bromo-3-methyl-phenoxy)butyl]piperidine (400 mg, 1.23 mmol, 1 equiv.), ethyl 2-bromoacetate (204.74 mg, 1.23 mmol, 135.59 µL, 1 equiv.) in CH3CN (5 mL) was added K2CO3 (508.31 mg, 3.68 mmol, 3 equiv.). The mixture was stirred at 60 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give ethyl 2-[4-[(3R)-3-(4-bromo-3-methyl-phenoxy)butyl]-1-piperidyl]ac etate (350 mg, crude) was obtained as a yellow oil. MS (ESI) m/z: 412.9 [M+H] + Step H. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[(1R)-3-[1-(2-ethoxy-2 -oxo-ethyl)-4-piperidyl]-1- methyl-propoxy]-2-methyl-phenyl]pyridine-2-carboxylate A mixture of ethyl 2-[4-[(3R)-3-(4-bromo-3-methyl-phenoxy)butyl]-1-piperidyl]ac etate (150 mg, 363.76 µmol, 1 equiv.) and tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborol an-2-yl)pyridine-2-carboxylate (222.82 mg, 363.76 µmol, 1 equiv.) in dioxane (3 mL) was added KF (1.5 M, 727.52 µL, 3 equiv.) and Ad2nBuP Pd G3 (cataCXium® A Pd G3) (26.49 mg, 36.38 µmol, 0.1 equiv.). After addition, the mixture was degassed and purged with N 2 for three times, and then the mixture was stirred at 100 °C for 2 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~45% ethyl acetate/petroleum ether) to give tert-butyl 6-[8- (1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin- 2-yl]-3-[4-[(1R)-3-[1-(2- ethoxy-2-oxo-ethyl)-4-piperidyl]-1-methyl-propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (150 mg, 171.0 µmol, 47.0% yield, 93.2% purity) as a yellow solid. MS (ESI) m/z: 818.3 [M+H] + Step I. Procedure for preparation of 2-[4-[(3R)-3-[4-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-3- methyl-phenoxy]butyl]-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[4-[(1R)-3-[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]-1- methyl-propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (150 mg, 183.37 µmol, 1 equiv.) in THF (1 mL) and H 2 O (1 mL) was added LiOH·H 2 O (21.96 mg, 916.83 µmol, 5 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was quenched by HCl (1M, 5 mL) and then filtered and concentrated under reduced pressure to give 2-[4-[(3R)-3-[4-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-3-methyl- phenoxy]butyl]-1-piperidyl]acetic acid (140 mg, 177.2 µmol, 96.6% yield) was obtained as a white solid. MS (ESI) m/z: 790.4 [M+H] + Step J. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[(1R)-3-[1-[2-[[3-(2,6 -dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-4-piperidyl]-1-methyl-propo xy]-2-methyl- phenyl]pyridine-2-carboxylate To a solution of 2-[4-[(3R)-3-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-3-methyl- phenoxy]butyl]-1- piperidyl]acetic acid (140 mg, 177.22 µmol, 1 equiv.) and 3-(6-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (54.93 mg, 212.66 µmol, 1.2 equiv.) in pyridine (1 mL) was added EDCI (50.96 mg, 265.83 µmol, 1.5 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was quenched by addition of water (10 mL), and then filtered and concentrated under reduced pressure to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[4-[(1R)-3-[1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]-1-methyl-propoxy]-2-meth yl-phenyl]pyridine-2- carboxylate (150 mg, 145.6 µmol, 82.1% yield) as a red solid. MS (ESI) m/z: 1030.4 [M+H] + Step K. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[4-[(1R)-3-[1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]-1-methyl-propoxy]-2-meth yl-phenyl]pyridine-2- carboxylic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)- yl)-3-(4-(((2R)-4-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-meth yl-1H-indazol-6-yl)amino)-2- oxoethyl)piperidin-4-yl)butan-2-yl)oxy)-2-methylphenyl)picol inic acid] To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[4-[(1R)-3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methy l-indazol-6-yl]amino]-2-oxo- ethyl]-4-piperidyl]-1-methyl-propoxy]-2-methyl-phenyl]pyridi ne-2-carboxylate (150 mg, 145.60 µmol, 1 equiv.) in DCM (2 mL) was added TFA (1.15 g, 10.13 mmol, 750.00 µL, 69.57 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4-[(1R)- 3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]am ino]-2-oxo-ethyl]-4-piperidyl]- 1-methyl-propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid [rel-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(4-(((2R)-4- (1-(2-((3-(2,6-dioxopiperidin- 3-yl)-1-methyl-1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-4 -yl)butan-2-yl)oxy)-2- methylphenyl)picolinic acid] (91.9 mg, 93. µmol, 64.2% yield, 99.1% purity) as a white solid. MS (ESI) m/z: 974.3 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 10.87 (s, 1H), 9.82 (s, 1H), 8.15 (s, 1H), 8.09 - 7.99 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 8.8 Hz, 2H), 7.50 - 7.42 (m, 3H), 7.40 - 7.31 (m, 2H), 7.25 - 7.19 (m, 1H), 6.92 (t, J = 9.2 Hz, 2H), 6.76 (d, J = 2.0 Hz, 1H), 6.73 - 6.64 (m, 1H), 4.97 (s, 2H), 4.43 - 4.36 (m, 1H), 4.35 - 4.29 (m, 1H), 3.97 - 3.87 (m, 5H), 3.13 (s, 2H), 3.02 (t, J = 5.2 Hz, 2H), 2.87 (d, J = 10.4 Hz, 2H), 2.67 - 2.59 (m, 2H), 2.37 - 2.29 (m, 1H), 2.21 - 2.09 (m, 3H), 2.02 (s, 3H), 1.72 - 1.62 (m, 3H), 1.61 - 1.51 (m, 1H), 1.43 - 1.34 (m, 1H), 1.33 - 1.20 (m, 7H). Example 78. Preparation of Compound 276b 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4-[(1S)-3-[1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-o xo-ethyl]-4-piperidyl]-1- methyl-propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(4-(((2S)-4- (1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl )amino)-2-oxoethyl)piperidin-4- yl)butan-2-yl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of 4-[(3S)-3-(4-bromo-3-methyl- phenoxy)butyl]piperidine A solution of tert-butyl 4-[(3S)-3-(4-bromo-3-methyl-phenoxy)butyl]piperidine-1-carbo xylate (350.00 mg, 820.85 µmol, 1 equiv.) in HCl/EtOAc (4 mL) was stirred at 25 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give 4-[(3S)-3-(4- bromo-3-methyl-phenoxy)butyl]piperidine (350 mg, crude) as a yellow oil. MS (ESI) m/z: 326.0 [M+H] + Step B. Procedure for preparation of ethyl 2-[4-[(3S)-3-(4-bromo-3-methyl- phenoxy)butyl]-1-piperidyl]acetate To a solution of 4-[(3S)-3-(4-bromo-3-methyl-phenoxy)butyl]piperidine (350 mg, 1.07 mmol, 1 equiv.), ethyl 2-bromoacetate (179.15 mg, 1.07 mmol, 118.64 µL, 1 equiv.) in CH3CN (4 mL) was added K2CO3 (444.79 mg, 3.22 mmol, 3 equiv.). The mixture was stirred at 60 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give ethyl 2-[4-[(3S)-3-(4-bromo-3-methyl-phenoxy)butyl]-1-piperidyl]ac etate (340 mg, crude) as a yellow oil. MS (ESI) m/z: 412.0 [M+H] + Step C. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[(1S)-3-[1-(2-ethoxy-2 -oxo-ethyl)-4-piperidyl]-1- methyl-propoxy]-2-methyl-phenyl]pyridine-2-carboxylate A mixture of ethyl 2-[4-[(3S)-3-(4-bromo-3-methyl-phenoxy)butyl]-1-piperidyl]ac etate (150.00 mg, 363.76 µmol, 1 equiv.) and tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)pyridine-2- carboxylate (222.82 mg, 363.76 µmol, 1 equiv.) in dioxane (3 mL) was added KF (1.5 M, 727.52 µL, 3 equiv.) and Ad2nBuP Pd G3(cataCXium® A Pd G3) (26.49 mg, 36.38 µmol, 0.1 equiv.). After addition, the mixture was degassed and purged with N 2 three times, and then the mixture was stirred at 100 °C for 2 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~40% ethyl acetate/petroleum ether) to give compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[4-[(1S)-3-[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]-1-me thyl-propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (180 mg, 210.7 µmol, 57.9% yield, 95.7% purity) as a white solid. MS (ESI) m/z: 818.8 [M+H] + Step D. Procedure for preparation of 2-[4-[(3S)-3-[4-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-3- methyl-phenoxy]butyl]-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[4-[(1S)-3-[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]-1- methyl-propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (150 mg, 183.37 µmol, 1 equiv.) in THF (1 mL) and H 2 O (1 mL) was added LiOH (21.96 mg, 916.83 µmol, 5 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was quenched by HCl 1M (5 mL), and then filtered and concentrated under reduced pressure to give 2-[4-[(3S)-3-[4-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-3-methyl- phenoxy]butyl]-1-piperidyl]acetic acid (130 mg, 164.5 µmol, 89.7% yield) as a white solid. MS (ESI) m/z: 790.3 [M+H] + Step E. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[(1S)-3-[1-[2-[[3-(2,6 -dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-4-piperidyl]-1-methyl-propo xy]-2-methyl- phenyl]pyridine-2-carboxylate To a solution of 2-[4-[(3S)-3-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-3-methyl- phenoxy]butyl]-1- piperidyl]acetic acid (130.00 mg, 164.56 µmol, 1 equiv.) and 3-(6-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (51.00 mg, 197.47 µmol, 1.2 equiv.) in pyridine (1 mL) was added EDCI (47.32 mg, 246.84 µmol, 1.5 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was quenched by addition of water (10 mL), and then filtered and concentrated under reduced pressure to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[4-[(1S)-3-[1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]-1-methyl-propoxy]-2-meth yl-phenyl]pyridine-2- carboxylate (150 mg, 145.6 µmol, 88.4% yield) as a red solid.. MS (ESI) m/z: 1030.4 [M+H] + Step F. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[4-[(1S)-3-[1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]-1-methyl-propoxy]-2-meth yl-phenyl]pyridine-2- carboxylic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)- yl)-3-(4-(((2S)-4-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-meth yl-1H-indazol-6-yl)amino)-2- oxoethyl)piperidin-4-yl)butan-2-yl)oxy)-2-methylphenyl)picol inic acid] To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[4-[(1S)-3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methy l-indazol-6-yl]amino]-2-oxo- ethyl]-4-piperidyl]-1-methyl-propoxy]-2-methyl-phenyl]pyridi ne-2-carboxylate (150.00 mg, 145.60 µmol, 1 equiv.) in DCM (2 mL) was added TFA (1.15 g, 10.13 mmol, 750.00 µL, 69.57 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4-[(1S)-3- [1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amin o]-2-oxo-ethyl]-4-piperidyl]-1- methyl-propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid [rel-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(4-(((2S)-4- (1-(2-((3-(2,6-dioxopiperidin- 3-yl)-1-methyl-1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-4 -yl)butan-2-yl)oxy)-2- methylphenyl)picolinic acid] (70.2 mg, 71.2 µmol, 48.9% yield, 98.8% purity) as a yellow solid. MS (ESI) m/z: 974.4 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.87 (s, 1H), 9.82 (s, 1H), 8.15 (s, 1H), 8.07 - 8.00 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 8.4 Hz, 2H), 7.50 - 7.41 (m, 3H), 7.40 - 7.31 (m, 2H), 7.25 - 7.18 (m, 1H), 6.96 - 6.88 (m, 2H), 6.76 (d, J = 2.0 Hz, 1H), 6.72 - 6.65 (m, 1H), 4.97 (s, 2H), 4.43 - 4.36 (m, 1H), 4.35 - 4.29 (m, 1H), 3.95 - 3.87 (m, 5H), 3.13 (s, 2H), 3.02 (t, J = 5.6 Hz, 2H), 2.87 (d, J = 10.4 Hz, 2H), 2.68 - 2.60 (m, 2H), 2.37 - 2.30 (m, 1H), 2.20 - 2.10 (m, 3H), 2.02 (s, 3H), 1.66 (d, J = 8.0 Hz, 3H), 1.61 - 1.52 (m, 1H), 1.43 - 1.35 (m, 1H), 1.32 - 1.21 (m, 7H). Example 79. Preparation of Compound 279 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[1-[[1-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]-4-piperidyl]m ethyl]-4-piperidyl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 3-(2,6-dibenzyloxy-3-pyridyl)-7-[4- (dimethoxymethyl)-1-piperidyl]-1-methyl-indazole A mixture of 7-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (1 g, 2.00 mmol, 1 equiv.), 4-(dimethoxymethyl)piperidine (477.31 mg, 3.00 mmol, 1.5 equiv.), 1,3-bis[2,6-bis(1- propylbutyl)phenyl]-4,5-dichloro-2H-imidazol-1-ium-2-ide;3- chloropyridine;dichloropalladium (194.40 mg, 199.85 µmol, 0.1 equiv.), and Cs2CO3 (1.95 g, 6.00 mmol, 3 equiv.) in 2-methyl-2-butanol (15 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 110 °C for 3 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to remove solvent. The residue was purified by flash silica gel chromatography (Eluent of 0~20% ethyl acetate/petroleum ether). The compound 3-(2,6-dibenzyloxy-3-pyridyl)-7-[4-(dimethoxymethyl)-1-piper idyl]-1- methyl-indazole (360 mg, 622.0 µmol, 31.1% yield) was obtained as a yellow solid. MS (ESI) m/z: 579.5 [M+H] +. Step B. Procedure for preparation of 3-[7-[4-(dimethoxymethyl)-1-piperidyl]-1-methyl- indazol-3-yl]piperidine-2,6-dione A mixture of 3-(2,6-dibenzyloxy-3-pyridyl)-7-[4-(dimethoxymethyl)-1-piper idyl]-1-methyl- indazole (360 mg, 622.08 µmol, 1 equiv.), Pd/C (150 mg, 622.08 µmol, 10% purity, 1 equiv.), Pd(OH) 2 (150 mg, 106.81 µmol, 10% purity, 0.172 equiv.) and AcOH (112.07 mg, 1.87 mmol, 106.74 µL, 3 equiv.) in EtOH (2.5 mL) and THF (2.5 mL) was degassed and purged with H2 three times, and then the mixture was stirred at 25 °C for 12 hours under H 2 (15 Psi) atmosphere. The mixture was filtered with THF (30 mL), and the filtrate was concentrated under reduced pressure to give 3-[7-[4-(dimethoxymethyl)-1-piperidyl]-1-methyl-indazol-3-yl ]piperidine- 2,6-dione (330 mg, crude) as a black solid, which was used in the next step without further purification. MS (ESI) m/z: 401.3 [M+H] +. Step C. Procedure for preparation of 1-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7- yl]piperidine-4-carbaldehyde A mixture of 3-[7-[4-(dimethoxymethyl)-1-piperidyl]-1-methyl-indazol-3-yl ]piperidine-2,6- dione (330 mg, 824.03 µmol, 1 equiv.) in HCOOH (3.5 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 80 °C for 2 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give 1-[3-(2,6-dioxo-3-piperidyl)- 1-methyl-indazol-7-yl]piperidine-4-carbaldehyde (220 mg, crude) as a brown solid. MS (ESI) m/z: 355.2 [M+H] +. Step D. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[1-[[1-[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-7-yl]-4- piperidyl]methyl]-4-piperidyl]propoxy]-2-methyl-phenyl]pyrid ine-2-carboxylic acid A solution of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[2- methyl-3-[3-(4-piperidyl)propoxy]phenyl]pyridine-2-carboxyli c acid (150 mg, 226.65 µmol, 1 equiv.) and 1-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]piperidin e-4-carbaldehyde (240.98 mg, 679.95 µmol, 3 equiv.) in DCM (15 mL) was stirred at 25 °C for 11 h. To the mixture was then added NaBH(OAc) 3 (144.11 mg, 679.95 µmol, 3 equiv.). The mixture was stirred at 25 °C for 1 h . The reaction mixture was quenched by addition H 2 O (30 mL) and extracted with DCM (30 mL × 2). The combined organic layers were concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC. The compound 6- [8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinol in-2-yl]-3-[3-[3-[1-[[1-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]-4-piperidyl]m ethyl]-4-piperidyl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid (23.9 mg, 21.6 µmol, 9.5% yield, 90.5% purity) was obtained as an off-white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.87 (s, 1H), 8.01 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.4 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.48 - 7.40 (m, 3H), 7.39 - 7.31 (m, 3H), 7.11 - 7.05 (m, 1H), 7.03 - 6.98 (m, 2H), 6.95 - 6.90 (m, 1H), 6.87 (d, J = 7.6 Hz, 1H), 6.64 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.32 (dd, J = 5.2, 9.6 Hz, 1H), 4.23 (s, 3H), 3.97 - 3.89 (m, 4H), 3.02 (t, J = 5.6 Hz, 2H), 2.90 - 2.83 (m, 3H), 2.65 - 2.62 (m, 2H), 2.30 - 2.12 (m, 5H), 1.92 - 1.82 (m, 7H), 1.78 - 1.62 (m, 6H), 1.38 - 1.23 (m, 6H), 1.20 - 1.10 (m, 2H) Example 80. Preparation of Compound 280 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[6-[2-[[3- (2, 6-dioxo-3-piperidyl) -1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]-3-pyridyl]propoxy ]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of methyl 2-(5-bromo-2-pyridyl) acetate To a solution of 2-(5-bromo-2-pyridyl) acetic acid (2.00 g, 9.26 mmol, 1 equiv.) in MeOH (20 mL) was added SOCl2 (2.20 g, 18.52 mmol, 1.34 mL, 2 equiv.) at 0 °C. The mixture was stirred at 25 °C for 3 hours. The reaction mixture was concentrated under reduced pressure to remove MeOH. The residue was diluted with water (20 mL) and extracted with DCM (20 mL × 3). The combined organic layers were washed with NaHCO3 (30 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give methyl 2-(5-bromo-2-pyridyl) acetate (2.1 g, crude) as a brown oil. 1 H NMR (400 MHz, CCDCl3-d) δ = 8.62 (d, J = 2.4 Hz, 1H), 7.80 (dd, J = 2.4, 8.4 Hz, 1H), 7.22 (d, J = 8.4 Hz, 1H), 3.83 (s, 2H), 3.73 (s, 3H) Step B. Procedure for preparation of methyl 2-[5-[3-[tert-butyl (dimethyl) silyl]oxypropyl]-2-pyridyl]acetate To an 15 mL vial equipped with a stir bar was added methyl 2-(5-bromo-2-pyridyl) acetate (950 mg, 4.13 mmol, 1 equiv.), 3-bromopropoxy-tert-butyl-dimethyl-silane (1.36 g, 5.37 mmol, 1.3 equiv.), bis[3, 5-difluoro-2-[5-(trifluoromethyl) -2-pyridyl]phenyl]iridium (1+) ;4-tert- butyl-2-(4-tert-butyl-2-pyridyl) pyridine;hexafluorophosphate (46.33 mg, 41.29 μmol, 0.01 equiv.), NiCl2. Dtbbpy (24.65 mg, 61.94 μmol, 0.015 equiv.), TTMSS (1.03 g, 4.13 mmol, 1.27 mL, 1 equiv.), and Na2CO3 (875.34 mg, 8.26 mmol, 2 equiv.) in DME (10 mL). The vial was sealed and placed under nitrogen was added. The reaction was stirred and irradiated with a 10 W blue LED lamp (3 cm away), with cooling water to keep the reaction temperature at 25 °C for 14 hours. The reaction mixture was concentrated and the residue was purified by flash silica gel chromatography (Eluent of 0 ~ 10% ethyl acetate/petroleum ether). The compound methyl 2-[5-[3-[tert-butyl (dimethyl) silyl]oxypropyl]-2-pyridyl]acetate (730 mg, 2.1 mmol, 51.3% yield, 94% purity) was obtained as a yellow oil. MS (ESI) m/z: 324.2 [M+H] + . 1 H NMR (400 MHz, CDCl 3 -d) δ = 8.40 (d, J = 2.0 Hz, 1H), 7.50 (dd, J = 2.4, 8.0 Hz, 1H), 7.22 (d, J = 8.0 Hz, 1H), 3.83 (s, 2H), 3.73 (s, 3H), 3.64 (t, J = 6.0 Hz, 2H), 2.73 - 2.65 (m, 2H), 1.84 - 1.79 (m, 2H), 0.91 (s, 8H), 0.08 - 0.02 (m, 6H) Step C. Procedure for preparation of methyl 2-[5-(3-hydroxypropyl) -2-pyridyl]acetate To a solution of methyl 2-[5-[3-[tert-butyl (dimethyl) silyl]oxypropyl]-2-pyridyl]acetate (370 mg, 1.14 mmol, 1 equiv.) in THF (1.5 mL) was added pyridine;hydrofluoride (1.65 g, 11.65 mmol, 1.5 mL, 70% purity, 10.19 equiv.) at 25 °C. The reaction mixture was stirred at 25 °C for 1 hour. The reaction mixture was diluted with water (5 mL) and extracted with EtOAc (10 mL × 5). The combined organic layers were dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The compound methyl 2-[5-(3-hydroxypropyl) -2- pyridyl]acetate (200 mg, crude) was obtained as a yellow oil. Step D. Procedure for preparation of methyl 2-[5-[3-(3-bromo-2-methyl-phenoxy) propyl]-2-pyridyl]acetate To a solution of 3-bromo-2-methyl-phenol (196.65 mg, 1.05 mmol, 1.1 equiv.) in toluene (4 mL) was added methyl 2-[5-(3-hydroxypropyl) -2-pyridyl]acetate (200 mg, 955.84 μmol, 1 equiv.) and 2-(tributyl-λ 5 -phosphanylidene) acetonitrile (299.90 mg, 1.24 mmol, 1.3 equiv.) at 25 °C. The reaction mixture was degassed and purged with N 2 three times, and then the mixture was stirred at 120 °C for 16 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 50% ethyl acetate/petroleum ether). The compound methyl 2-[5-[3-(3-bromo-2-methyl-phenoxy) propyl]-2-pyridyl]acetate (190 mg, 497.2 μmol, 52.0% yield, 99% purity) was obtained as a yellow oil. MS (ESI) m/z: 377.9 [M+H] + . 1 H NMR (400 MHz, CDCl3-d) δ = 8.43 (d, J = 1.6 Hz, 1H), 7.52 (dd, J = 2.0, 8.0 Hz, 1H), 7.24 (d, J = 8.0 Hz, 1H), 7.16 (d, J = 8.0 Hz, 1H), 6.99 (t, J = 8.0 Hz, 1H), 6.73 (d, J = 8.4 Hz, 1H), 3.97 (t, J = 6.0 Hz, 2H), 3.84 (s, 2H), 3.73 (s, 3H), 2.84 (t, J = 7.6 Hz, 2H), 2.34 (s, 3H), 2.18 - 2.09 (m, 2H) Step E. Procedure for preparation of tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[6-(2-methoxy-2-oxo-e thyl) -3-pyridyl]propoxy]- 2-methyl-phenyl]pyridine-2-carboxylate Tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-3-(4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridine-2-carboxylate (267.38 mg, 436.50 μmol, 1.1 equiv.), methyl 2-[5-[3-(3-bromo-2-methyl-phenoxy) propyl]-2-pyridyl]acetate (190 mg, 396.82 μmol, 79% purity, 1 equiv.), KF (69.17 mg, 1.19 mmol, 27.89 μL, 3 equiv.), and [2-(2- aminophenyl) phenyl]palladium (1+) ;bis (1-adamantyl) -butyl-phosphane;methanesulfonate (57.80 mg, 79.36 μmol, 0.2 equiv.) were taken up into a microwave tube in dioxane (2 mL) and H2O (0.2 mL). The sealed tube was heated at 100 °C for 1 hour under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 30% ethyl acetate/petroleum ether). The compound tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H- isoquinolin-2-yl]-3-[3-[3-[6-(2-methoxy-2-oxo-ethyl) -3-pyridyl]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (130 mg, 97.8 μmol, 24.6% yield, 59% purity) was obtained as a yellow solid. MS (ESI) m/z: 784.5 [M+H] + . Step F. Procedure for preparation of 2-[5-[3-[3-[6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) - 3, 4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyrid yl]-2-methyl- phenoxy]propyl]-2-pyridyl]acetic acid To a solution of tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H- isoquinolin-2-yl]-3-[3-[3-[6-(2-methoxy-2-oxo-ethyl) -3-pyridyl]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (130 mg, 97.84 μmol, 59% purity, 1 equiv.) in THF (1 mL) and H 2 O (0.1 mL) was added LiOH•H 2 O (12.32 mg, 293.52 μmol, 3 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was treated with water (4 mL). The pH was adjusted to around 3 by progressively adding diluted HCl. The mixture was filtered and concentrated to give a residue. The compound 2-[5-[3-[3-[6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]propyl]-2- pyridyl]acetic acid (168 mg, crude) was obtained as a yellow solid. MS (ESI) m/z: 770.5 [M+H] + . Step G. Procedure for preparation of tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) - 3, 4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[6-[2-[[3-(2, 6-dioxo-3-piperidyl) -1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-3-pyridyl]propoxy]-2-methyl -phenyl]pyridine-2- carboxylate To a solution of 2-[5-[3-[3-[6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]propyl]-2- pyridyl]acetic acid (160 mg, 207.82 μmol, 1 equiv.) in pyridine (0.5 mL) was added EDCI (47.81 mg, 249.38 μmol, 1.2 equiv.) and 3-(6-amino-1-methyl-indazol-3-yl) piperidine-2, 6- dione (64.41 mg, 249.38 μmol, 1.2 equiv.) at 25 °C. The reaction mixture was stirred at 25 °C for 16 hours. The mixture was triturated with water (5 mL) at 25 °C for 10 min. The mixture was filtered to give the compound tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4- dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[6-[2-[[3-(2, 6-dioxo-3-piperidyl) -1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-3-pyridyl]propoxy]-2-methyl-phenyl]py ridine-2-carboxylate (80 mg, 38.9 μmol, 18.7% yield, 49.2% purity) as a yellow solid. MS (ESI) m/z: 1010.3 [M+H] + . Step H. Procedure for preparation of 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4- dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[6-[2-[[3-(2, 6-dioxo-3-piperidyl) -1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-3-pyridyl]propoxy]-2-methyl-phenyl] pyridine-2-carboxylic acid To a solution of tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H- isoquinolin-2-yl]-3-[3-[3-[6-[2-[[3-(2, 6-dioxo-3-piperidyl) -1-methyl-indazol-6-yl]amino]-2- oxo-ethyl]-3-pyridyl]propoxy]-2-methyl-phenyl]pyridine-2-car boxylate (80 mg, 50.92 μmol, 64.3% purity, 1 equiv.) in DCM (0.5 mL) and TFA (0.5 mL) was stirred at 25 °C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC. The compound 6-[8-(1, 3-benzothiazol-2- ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[6-[2-[[3-(2, 6-dioxo-3-piperidyl) -1- methyl-indazol-6-yl]amino]-2-oxo-ethyl]-3-pyridyl]propoxy]-2 -methyl-phenyl]pyridine-2- carboxylic acid (21.3 mg, 20.9 μmol, 41.2% yield, 93.7% purity) was obtained as a yellow solid. MS (ESI) m/z: 954.3 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.86 (s, 1H), 10.43 (s, 1H), 8.39 (d, J = 2.0 Hz, 1H), 8.07 (s, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 7.6 Hz, 1H), 7.67 - 7.60 (m, 3H), 7.49 - 7.42 (m, 3H), 7.39 - 7.32 (m, 3H), 7.14 - 7.05 (m, 2H), 6.95 (d, J = 8.8 Hz, 1H), 6.86 (d, J = 8.0 Hz, 1H), 6.64 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.31 (dd, J = 5.2, 9.6 Hz, 1H), 4.00 - 3.96 (m, 2H), 3.94 - 3.90 (m, 2H), 3.89 (s, 3H), 3.85 (s, 2H), 3.02 (t, J = 5.6 Hz, 2H), 2.78 (t, J = 7.2 Hz, 2H), 2.67 - 2.62 (m, 2H), 2.37 - 2.29 (m, 1H), 2.21 - 2.13 (m, 1H), 2.10 - 2.02 (m, 2H), 1.92 (s, 3H) Example 81. Preparation of Compound 289b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-((1R)-6-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino )-2-oxoethyl)-6- azaspiro[2.5]octan-1-yl)ethoxy)-2-methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(3-(2-((1R)-6- (2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)am ino)-2-oxoethyl)-6- azaspiro[2.5]octan-1-yl)ethoxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of 6-(tert-butyl) 1-methyl 6-azaspiro[2.5]octane-1,6- dicarboxylate A mixture of 6-(tert-butoxycarbonyl)-6-azaspiro[2.5]octane-1-carboxylic acid (5 g, 19.58 mmol, 1 equiv.), Cs 2 CO 3 (7.66 g, 23.50 mmol, 1.2 equiv.) in DMF (50 mL) ,then added MeI (3.34 g, 23.50 mmol, 1.46 mL, 1.2 equiv.) to the mixture, the mixture was stirred at 25 °C for 2 hours, The mixture was diluted with water (10 mL) and extracted with EtOAc 15 mL (5 mL × 3). The combined organic layers were washed with brine (5 mL × 2), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give the compound 6-(tert-butyl) 1-methyl 6-azaspiro[2.5]octane-1,6-dicarboxylate (5 g, crude) as a colorless oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 3.60 (s, 3H), 3.41 - 3.26 (m, 4H), 3.17 - 3.09 (m, 1H), 1.72 - 1.43 (m, 4H), 1.39 (s, 9H), 0.99 (d, J = 6.8 Hz, 2H) Step B. Procedure for preparation of tert-butyl 1-(hydroxymethyl)-6-azaspiro[2.5]octane- 6-carboxylate To a solution of 6-(tert-butyl) 1-methyl 6-azaspiro[2.5]octane-1,6-dicarboxylate (5 g, crude) in THF (100 mL) was added LAH (1.69 g, 44.55 mmol, 1.2 equiv.) at 0 °C. Then the mixture was stirred at 20 °C for 2 hours. The reaction mixture was quenched by addition Na 2 SO 4 •10H 2 O (1 g) under 0 °C and N2 atmosphere, and then filtered and concentrated under reduced pressure to give a residue. The residue was used for next step without other purification. The compound tert-butyl 1-(hydroxymethyl)-6-azaspiro[2.5]octane-6-carboxylate (8.4 g, 34.8 mmol, 93.8% yield) was obtained as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 5.75 (s, 1H), 4.40 (t, J = 5.2 Hz, 1H), 3.56 - 3.43 (m, 2H), 3.38 - 3.33 (m, 1H), 3.30 - 3.21 (m, 3H), 1.39 (s, 9H), 1.36 - 1.25 (m, 2H), 1.22 - 1.15 (m, 1H), 0.87 - 0.77 (m, 1H), 0.43 (dd, J = 4.4, 8.8 Hz, 1H), 0.14 (t, J = 4.8 Hz, 1H) Step C. Procedure for preparation of tert-butyl 1-formyl-6-azaspiro[2.5]octane-6- carboxylate A mixture of DMSO (8.29 g, 106.08 mmol, 8.29 mL, 4 equiv.) in DCM (65 mL) was added dropwise to a solution of (COCl)2 (6.73 g, 53.04 mmol, 4.64 mL, 2 equiv.) in DCM (10 mL) at -70 °C under N2 atmosphere, The mixture was stirred at -70 °C for 1 hour, tert-butyl 1- (hydroxymethyl)-6-azaspiro[2.5]octane-6-carboxylate (6.4 g, 26.52 mmol, 1 equiv.) in DCM (10 mL) and was added dropwise at -70 °C. The solution was stirred for 1 hour at -70 °C. Then TEA (16.10 g, 159.12 mmol, 22.15 mL, 6 equiv.) was added into the solution. The solution was stirred at -70 °C for 0.5 hour under N 2 atmosphere. The mixture was diluted with H 2 O (10 mL) and extracted with DCM 15 mL (5 mL × 3). The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 30% ethyl acetate / petroleum ether) to give tert-butyl 1-formyl-6-azaspiro[2.5]octane-6-carboxylate (4.7 g, 19.6 mmol, 74.1% yield) as a yellow oil. Step D. Procedure for preparation of tert-butyl (E)-1-(2-methoxyvinyl)-6- azaspiro[2.5]octane-6-carboxylate A mixture of methoxymethyl(triphenyl)phosphonium;bromide (9.71 g, 25.07 mmol, 2 equiv.) in THF (25 mL) was added LiHMDS (1 M, 25.07 mL, 2 equiv.) at 0°C for 1 hour under N2 atmosphere, then tert-butyl 1-formyl-6-azaspiro[2.5]octane-6-carboxylate (3 g, 12.54 mmol, 1 equiv.) in THF (5 mL) was added dropwise at 0°C, the resulting mixture was stirred at 20°C for 1 hour under N2 atmosphere .The reaction mixture was quenched by addition sat. NH4Cl (5 mL) at 0°C, and then diluted with EtOAc (1 mL) and extracted with 3mL (1 mL × 3). The combined organic layers were washed with H 2 O (3 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 8% ethyl acetate / petroleum ether) to give tert-butyl (E)-1- (2-methoxyvinyl)-6-azaspiro[2.5]octane-6-carboxylate (4 g, crude) as a yellow oil. 1 H NMR (400 MHz, CDCl3) δ = 5.97 (dd, J = 0.8, 6.4 Hz, 1H), 4.07 (dd, J = 6.4, 9.2 Hz, 1H), 3.62 (s, 3H), 1.46 - 1.45 (m, 12H), 1.40 - 1.32 (m, 4H), 1.28 - 1.15 (m, 1H), 0.75 (dd, J = 4.4, 8.4 Hz, 1H), 0.35 - 0.29 (m, 2H) Step E. Procedure for preparation of tert-butyl 1-(2-oxoethyl)-6-azaspiro[2.5]octane-6- carboxylate A mixture of tert-butyl (E)-1-(2-methoxyvinyl)-6-azaspiro[2.5]octane-6-carboxylate (3 g, 11.22 mmol, 1 equiv.), TFA (3.84 g, 33.66 mmol, 2.49 mL, 3 equiv.), in H2O (30 mL) and CH3CN (120 mL) was stirred at 25 °C for 16 hours. The reaction mixture was quenched by addition NaHCO 3 (5 mL), and then diluted with H 2 O (20 mL) and extracted with DCM 90 mL (30 mL × 3). Then the combined organic layers were filtered and concentrated. The residue was used for next step without other purification. The compound tert-butyl 1-(2-oxoethyl)-6- azaspiro[2.5]octane-6-carboxylate (3 g, crude) was obtained as a yellow oil. Step F. Procedure for preparation of tert-butyl 1-(2-hydroxyethyl)-6-azaspiro[2.5]octane- 6-carboxylate To a solution of tert-butyl 1-(2-oxoethyl)-6-azaspiro[2.5]octane-6-carboxylate (3 g, 11.84 mmol, 1 equiv.) in MeOH (30 mL) was added NaBH4 (537.58 mg, 14.21 mmol, 1.2 equiv.) at 0 °C, The mixture was stirred at 20 °C for 1 hour. The reaction mixture was quenched by addition sat. NH 4 Cl (5 mL) at 0 °C, and then diluted with ethyl acetate (1 mL) and extracted with 3mL (1 mL × 3). The combined organic layers were washed with H2O 3 mL, dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 30% ethyl acetate / petroleum ether) to give tert-butyl 1-(2-hydroxyethyl)-6-azaspiro[2.5]octane-6-carboxylate (1.8 g, 7.0 mmol, 59.5% yield) as a colorless oil. 1 H NMR (400 MHz, CDCl 3 ) δ = 3.75 - 3.69 (m, 2H), 3.67 - 3.56 (m, 2H), 3.26 - 3.20 (m, 2H), 1.82 - 1.74 (m, 1H), 1.64 - 1.58 (m, 2H), 1.46 (s, 9H), 1.44 - 1.37 (m, 1H), 1.36 - 1.27 (m, 1H), 1.17 - 1.09 (m, 1H), 0.68 - 0.61 (m, 1H), 0.53 - 0.50 (m, 1H), 0.07 (t, J = 4.8 Hz, 1H) Step G. Procedure for preparation of tert-butyl 1-(2-(3-bromo-2-methylphenoxy)ethyl)- 6-azaspiro[2.5]octane-6-carboxylate A mixture of tert-butyl 1-(2-hydroxyethyl)-6-azaspiro[2.5]octane-6-carboxylate (1 g, 3.92 mmol, 1.2 equiv.), 3-bromo-2-methyl-phenol (610.38 mg, 3.26 mmol, 1 equiv.), 2-(tributyl-λ 5 - phosphanylidene)acetonitrile (1.18 g, 4.90 mmol, 1.5 equiv.), in toluene (10 mL) was stirred at 120 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 20% ethyl acetate / petroleum ether) to give tert-butyl 1-(2-(3-bromo-2-methylphenoxy)ethyl)-6- azaspiro[2.5]octane-6-carboxylate (1.3 g, 3.1 mmol, 93.9% yield) as a yellow oil. MS (ESI) m/z: 368.1[M+H] + . Step H. Procedure for preparation of 1-(2-(3-bromo-2-methylphenoxy)ethyl)-6- azaspiro[2.5]octane A mixture of tert-butyl 1-(2-(3-bromo-2-methylphenoxy)ethyl)-6-azaspiro[2.5]octane-6 - carboxylate (1.3 g, 3.06 mmol, 1 equiv.), in TFA (3 mL) and DCM (9 mL) was stirred at 25 °C for 2 hours. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was used for next step without other purification. The compound 1-(2-(3-bromo-2-methylphenoxy)ethyl)-6-azaspiro[2.5]octane (1.3 g, crude, TFA) was obtained as a white solid. MS (ESI) m/z: 323.9[M+H] + . Step I. Procedure for preparation of ethyl 2-(1-(2-(3-bromo-2-methylphenoxy)ethyl)-6- azaspiro[2.5]octan-6-yl)acetate A mixture of 1-(2-(3-bromo-2-methylphenoxy)ethyl)-6-azaspiro[2.5]octane (1.3 g, 2.97 mmol, 1 equiv., TFA), ethyl 2-bromoacetate (544.89 mg, 3.26 mmol, 360.85 µL, 1.1 equiv.), TEA (1.50 g, 14.83 mmol, 2.06 mL, 5 equiv.), in CH3CN (10 mL) was stirred at 25 °C for 1 hour. The combined organic layers were filtered and concentrated under reduced pressure to give a residue The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 50% ethyl acetate / petroleum ether) to give ethyl 2-(1-(2-(3-bromo-2-methylphenoxy)ethyl)-6- azaspiro[2.5]octan-6-yl)acetate (1 g, 2.4 mmol, 80.6% yield, 98.0% purity) as a colorless oil. MS (ESI) m/z: 410.0 [M+H] + . Step J. Procedure for preparation of ethyl (R)-2-(1-(2-(3-bromo-2-methylphenoxy)ethyl)- 6-azaspiro[2.5]octan-6-yl)acetate The ethyl 2-(1-(2-(3-bromo-2-methylphenoxy)ethyl)-6-azaspiro[2.5]octan -6-yl)acetate residue was purified by prep - HPLC to give ethyl (S)-2-(1-(2-(3-bromo-2- methylphenoxy)ethyl)-6-azaspiro[2.5]octan-6-yl)acetate (450 mg, 1.10 mmol, 45.00% yield) as a colorless oil and ethyl (R)-2-(1-(2-(3-bromo-2-methylphenoxy)ethyl)-6- azaspiro[2.5]octan-6-yl)acetate (390 mg, 950.4 µmol, 39.00% yield) as a colorless oil . Step K. Procedure for preparation of tert-butyl (R)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(6-(2- ethoxy-2-oxoethyl)-6- azaspiro[2.5]octan-1-yl)ethoxy)-2-methylphenyl)picolinate A mixture of ethyl (R)-2-(1-(2-(3-bromo-2-methylphenoxy)ethyl)-6-azaspiro[2.5]o ctan-6- yl)acetate (100.00 mg, 243.70 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tet ramethyl-1,3,2-dioxaborolan-2- yl)pyridine-2-carboxylate (179.13 mg, 292.44 µmol, 1.2 equiv.), [2-(2- aminophenyl)phenyl]palladium(1 + );bis(1-adamantyl)-butyl-phosphane;methanesulfonate (35.50 mg, 48.74 µmol, 0.2 equiv.), K2CO3 (1.5 M, 487.40 µL, 3 equiv.) in dioxane (1 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 100 °C for 1 hour under N2 atmosphere under MW. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 60% ethyl acetate / petroleum ether) to give tert-butyl (R)-6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2( 1H)-yl)-3-(3-(2-(6-(2-ethoxy- 2-oxoethyl)-6-azaspiro[2.5]octan-1-yl)ethoxy)-2-methylphenyl )picolinate (130 mg, 159.3 µmol, 65.4% yield) as a yellow oil. MS (ESI) m/z: 816.3 [M+H] + . Step L. Procedure for preparation of (R)-2-(1-(2-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)ethyl)-6-azaspiro[2.5]octan-6-yl)acetic acid A mixture of tert-butyl (R)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquino lin- 2(1H)-yl)-3-(3-(2-(6-(2-ethoxy-2-oxoethyl)-6-azaspiro[2.5]oc tan-1-yl)ethoxy)-2- methylphenyl)picolinate (130 mg, 159.31 µmol, 1 equiv.), LiOH•H 2 O (20.06 mg, 477.93 µmol, 3 equiv.), in THF (0.8 mL) and H2O (0.4 mL) was stirred at 25 °C for 2 hours. The reaction mixture was diluted with H2O (5 mL) and concentrated as a turbid liquid. Then the pH of the turbid liquid was adjusted to 3 with 1 N HCl (5 mL), the residue was extracted with DCM / MeOH(20:1), The combined organic layers were filtered and concentrated. The residue was used for next step without other purification. The compound (R)-2-(1-(2-(3-(6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)ethyl)-6-azaspi ro[2.5]octan-6-yl)acetic acid (100 mg, 126.9 µmol, 79.7% yield) was obtained as a yellow solid. MS (ESI) m/z: 788.4 [M+H] + . Step M. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((1R)-6-(2-((3-(2,6 -dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)-6-azaspiro[2.5]oct an-1-yl)ethoxy)-2- methylphenyl)picolinate A mixture of (R)-2-(1-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)ethyl)-6- azaspiro[2.5]octan-6-yl)acetic acid (90 mg, 114.22 µmol, 1 equiv.), 3-(6-amino-1-methyl- indazol-3-yl)piperidine-2,6-dione (35.40 mg, 137.06 µmol, 1.2 equiv.), EDCI (32.84 mg, 171.33 µmol, 1.5 equiv.), in pyridine (0.5 mL) was stirred at 25 °C for 2 hour The mixture was diluted with H2O mL(10 mL × 3) and extracted with ethyl acetate 24 mL (8 mL × 3). The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was used for next step without other purification. The compound tert- butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-((1R)- 6-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl) amino)-2-oxoethyl)-6- azaspiro[2.5]octan-1-yl)ethoxy)-2-methylphenyl)picolinate (90 mg, crude) was obtained as a yellow solid. MS (ESI) m/z: 1028.5 [M+H] + . Step N. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((1R)-6-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-6-azaspiro[2.5]octan-1-yl )ethoxy)-2- methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((1R)-6-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-6-azaspiro[2.5]octan-1-yl )ethoxy)-2- methylphenyl)picolinic acid] A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(2-((1R)-6-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-6-yl)amino)-2- oxoethyl)-6-azaspiro[2.5]octan-1-yl)ethoxy)-2-methylphenyl)p icolinate (90 mg, 87.53 µmol, 1 equiv.) in TFA (0.8 mL) and DCM (0.8 mL) was stirred at 25 °C for 2 hours, The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep–HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((1R)-6-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)-6-azaspiro[2.5]octan-1-yl)et hoxy)-2-methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(3-(2- ((1R)-6-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)amino)-2-oxoethyl)-6- azaspiro[2.5]octan-1-yl)ethoxy)-2-methylphenyl)picolinic acid] (98.7% purity) as a white solid. MS (ESI) m/z: 972.3 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.98 - 12.77 (m, 1H), 10.88 (s, 1H), 10.44 - 9.44 (m, 1H), 8.07 - 8.01 (m, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.69 - 7.59 (m, 2H), 7.49 - 7.32 (m, 5H), 7.23 - 7.15 (m, 1H), 7.10 (t, J = 7.6 Hz, 1H), 6.99 - 6.88 (m, 2H), 6.63 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.33 (dd, J = 5.2, 9.6 Hz, 1H), 4.13 - 4.02 (m, 2H), 3.94 - 3.89 (m, 5H), 3.47 - 3.35 (m, 6H), 3.02 ( t, J = 5.2 Hz, 2H), 2.69 - 2.53 (m, 4H), 2.37 - 2.31 (m, 1H), 2.21 - 2.13 (m, 1H), 1.92 (s, 3H), 1.76 - 1.57 (m, 2H), 1.47 - 1.01 (m, 2H), 0.92 - 0.68 (m, 1H), 0.61 - 0.35 (m, 1H), 0.23 - 0.00 (m, 1H) Example 82. Preparation or Compound 290 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[[1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-4-piperidyl]oxy]propyl]- 2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 4-prop-2-ynoxypiperidine-1-carboxylate A solution of tert-butyl 4-hydroxypiperidine-1-carboxylate (10 g, 49.69 mmol, 1.0 equiv.) in anhydrous THF (50 mL) was cooled to 0 °C. Subsequently, NaH (2.38 g, 59.62 mmol, 60% purity, 1.2 equiv.) was added under N2 atmosphere. The reaction mixture was stirred at 0 °C for 0.5 hours. Then, 3-bromoprop-1-yne (7.39 g, 49.69 mmol, 5.35 mL, 1.0 equiv.) was added. The resulting reaction mixture was stirred at 25 °C for 15.5 hours. Once completed, the reaction mixture was quenched with water (1 mL), then diluted with ethyl acetate (100 mL). The organic layers were washed with brine (20 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated in vacuo. The residue was purified by flash silica gel chromatography (Eluent of 0~10% ethyl acetate/petroleum ether) to give tert-butyl 4-prop-2-ynoxypiperidine- 1-carboxylate (8.1 g, 29.9 mmol, 60.1% yield, 88% purity) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 4.17 (d, J = 2.4 Hz, 2H), 3.69 - 3.56 (m, 3H), 3.41 - 3.36 (m, 1H), 3.03 (t, J = 9.6 Hz, 2H), 1.83 - 1.75 (m, 2H), 1.45 - 1.37 (m, 9H), 1.36 - 1.30 (m, 2H) Step B. Procedure for preparation of tert-butyl 4-[3-(3-bromo-2-methyl-phenyl) prop-2- ynoxy]piperidine-1-carboxylate A mixture of tert-butyl 4-prop-2-ynoxypiperidine-1-carboxylate (2.1 g, 8.78 mmol, 1.0 equiv.) , 1-bromo-3-iodo-2-methyl-benzene (2.61 g, 8.78 mmol, 1.0 equiv.), TEA (2.66 g, 26.33 mmol, 3.66 mL, 3.0 equiv.), CuI (334.25 mg, 1.76 mmol, 0.2 equiv.), and Pd(PPh 3 ) 2 Cl 2 (615.93 mg, 877.52 μmol, 0.1 equiv.) in DMF (30 mL) was degassed and purged with N 2 for three times, and then the mixture was stirred at 60 °C for 2 hours under N2 atmosphere. The reaction mixture was partitioned between ethyl acetate (150 mL) and H2O (150 mL). The organic phase was separated, washed with NaCl (aq.) (100 mL × 3), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~8% ethyl acetate/petroleum ether) to give tert-butyl 4-[3-(3- bromo-2-methyl-phenyl) prop-2-ynoxy]piperidine-1-carboxylate (2.6 g, 6.3 mmol, 71.5% yield, 99% purity) as a yellow solid. 1 H NMR (400 MHz, DMSO-d6) δ = 7.63 (dd, J = 0.8, 8.0 Hz, 1H), 7.49 - 7.43 (m, 1H), 7.15 (t, J = 8.0 Hz, 1H), 4.49 (s, 2H), 3.78 - 3.69 (m, 1H), 3.68 - 3.61 (m, 2H), 3.05 (s, 2H), 2.48 (s, 3H), 1.94 - 1.78 (m, 2H), 1.40 (s, 11H). Step C. Procedure for preparation of tert-butyl 4-[3-(3-bromo-2-methyl- phenyl)propoxy]piperidine-1-carboxylate To a solution of tert-butyl 4-[3-(3-bromo-2-methyl-phenyl) prop-2-ynoxy]piperidine-1- carboxylate (1.5 g, 3.67 mmol, 1.0 equiv.) in EtOAc (10 mL) was added Rh/Al2O3 (75.61 mg, 734.70 μmol, 0.2 equiv.).The mixture was stirred at 25 °C for 6 hours. Filtration of the reaction mixture then gives the crude product tert-butyl 4-[3-(3-bromo-2-methyl- phenyl)propoxy]piperidine-1-carboxylate (1.4 g, 3.5 mmol, 95.7% yield) as yellow oil. MS (ESI) m/z: 356.1 [M-56+H] + Step D. Procedure for preparation of 4-[3-(3-bromo-2-methyl-phenyl)propoxy] piperidine To a solution of tert-butyl 4-[3-(3-bromo-2-methyl-phenyl)propoxy]piperidine-1-carboxyla te (1.4 g, 3.40 mmol, 1.0 equiv.) in HCl/EtOAc (50 mL) and EtOAc (10 mL) .The mixture was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to remove HCl/EtOAc (50 mL) and EtOAc (10 mL) to give the crude product 4-[3-(3-bromo-2- methyl-phenyl)propoxy] piperidine (1.01 g, 3.2 mmol, 95.3% yield) as yellow oil. MS (ESI) m/z: 312.2 [M+H] + Step E. Procedure for preparation of ethyl 2-[4-[3-(3-bromo-2-methyl-phenyl)propoxy]- 1-piperidyl]acetate To a solution of ethyl 2-bromoacetate (420.10 mg, 2.52 mmol, 278.40 μL, 1.1 equiv.) and 4- [3-(3-bromo-2-methyl-phenyl)propoxy]piperidine (800 mg, 2.29 mmol, 1.0 equiv., HCl) and KI (380.84 mg, 2.29 mmol, 1.0 equiv.) in CH 3 CN (5 mL) was added TEA (696.44 mg, 6.88 mmol, 957.97 μL, 3.0 equiv.).The mixture was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure. The residue was diluted with H2O (50 mL) and extracted with ethyl acetate (50 mL × 3). The combined organic layers were washed with NaCl (aq.) (50 mL × 3), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 30~50% ethyl acetate/petroleum ether) to give ethyl 2-[4-[3-(3-bromo-2-methyl-phenyl)propoxy]-1- piperidyl]acetate (785 mg, 1.54 mmol, 67.00% yield, 78% purity) as a yellow oil. MS (ESI) m/z: 399.2 [M+H] + Step F. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[[1-(2-ethoxy-2-oxo -ethyl)-4- piperidyl]oxy]propyl]-2-methyl-phenyl]pyridine-2-carboxylate A mixture of ethyl 2-[4-[3-(3-bromo-2-methyl-phenyl)propoxy]-1-piperidyl]acetat e (400 mg, 1.00 mmol, 1.0 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (615.11 mg, 1.00 mmol, 1.0 equiv.), [2-(2-aminophenyl)phenyl]palladium(1+); bis(1- adamantyl)-butyl-phosphane; methanesulfonate (73.13 mg, 100.42 μmol, 0.1 equiv.), K 2 CO 3 (416.35 mg, 3.01 mmol, 3.0 equiv.) and in dioxane (8 mL) and H 2 O (1 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 100 °C for 1 hour under N2 atmosphere under microwave. The reaction mixture was concentrated under reduced pressure to remove dioxane (8 mL). The residue was diluted with H 2 O (50 mL) and extracted with ethyl acetate (50 mL × 3). The combined organic layers were dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 50~80% ethyl acetate/petroleum ether) to give tert-butyl 6-[8- (1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin- 2-yl]-3-[3-[3-[[1-(2-ethoxy-2- oxo-ethyl)-4-piperidyl]oxy]propyl]-2-methyl-phenyl]pyridine- 2-carboxylate (353 mg, 424.8 μmol, 42.3% yield, 96.8% purity) as yellow solid. MS (ESI) m/z: 804.6 [M+H] + Step G. Procedure for preparation of 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyr idyl]-2-methyl- phenyl]propoxy]-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[3-[[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]oxy]pro pyl]-2-methyl-phenyl]pyridine- 2-carboxylate (333 mg, 414.18 μmol, 1.0 equiv.) in THF (10 mL) was added LiOH•H 2 O (69.52 mg, 1.66 mmol, 4.0 equiv.) and H2O (10 mL).The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to remove THF. Then H 2 O (30 mL) was added to the mixture, and its pH was adjusted to 3~4 with HCl (1 N). The mixture was filtered to give 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenyl]propoxy]-1- piperidyl]acetic acid (253 mg, 326.0 μmol, 78.7% yield) as a yellow solid. MS (ESI) m/z: 776.5 [M+H] + Step H. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[[1-[2-[[3-(2,6-dio xo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-4-piperidyl]oxy]propyl]-2-m ethyl-phenyl]pyridine-2- carboxylate To a solution of 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenyl]propoxy]-1- piperidyl]acetic acid (200 mg, 257.75 μmol, 1.0 equiv.) in pyridine (8 mL) was added EDCI (74.12 mg, 386.62 μmol, 1.5 equiv.) and 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6- dione (66.57 mg, 257.75 μmol, 1.0 equiv.) The mixture was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. Tert-butyl 6-[8- (1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin- 2-yl]-3-[3-[3-[[1-[2-[[3-(2,6- dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl] -4-piperidyl]oxy]propyl]-2- methyl-phenyl]pyridine-2-carboxylate (215 mg, crude) was obtained as a yellow solid. MS (ESI) m/z: 1016.5 [M+H] + Step I. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[[1-[2-[[3-(2,6-dioxo-3-piperid yl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]oxy]propyl]-2-methyl-phen yl]pyridine-2-carboxylic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-inda zol-6-yl]amino]-2-oxo-ethyl]-4- piperidyl]oxy]propyl]-2-methyl-phenyl]pyridine-2-carboxylate (150 mg, 147.61 μmol, 1 equiv.) in TFA (10 mL) and DCM (10 mL) was stirred at 30 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to get a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]- 3-[3-[3-[[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-4- piperidyl]oxy]propyl]-2-methyl-phenyl]pyridine-2-carboxylic acid (90.0 mg, 93.3 μmol, 63.2% yield, 99.5% purity) as a yellow solid. MS (ESI) m/z: 960.5 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 13.07 - 12.64 (m, 1H), 10.89 (s, 1H), 9.88 ( s, 1H), 8.14 (s, 1H), 8.09 - 7.96 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 8.4 Hz, 2H), 7.52 - 7.43 (m, 3H), 7.37 (d, J = 7.6, 12.0 Hz, 2H), 7.24 (d, J = 8.4 Hz, 1H), 7.14 - 7.04 (m, 2H), 6.98 (d, J = 8.8 Hz, 1H), 6.87 (d, J = 6.8 Hz, 1H), 4.99 (s, 2H), 4.33 (dd, J = 5.2, 9.6 Hz, 1H), 3.92 (s, 5H), 3.44 (t, J = 6.0 Hz, 2H), 3.16 ( s, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.83 - 2.75 (m, 2H), 2.70 - 2.61 (m, 4H), 2.38 - 2.29 (m, 3H), 2.22 - 2.14 (m, 1H), 2.01 (s, 3H), 1.88 (d, J = 9.6 Hz, 2H), 1.80 - 1.70 (m, 2H), 1.64 - 1.53 (m, 2H) Example 83. Preparation of Compound 291 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4-[3-[[1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-4-piperidyl]oxy]propyl]- 2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 4-prop-2-ynoxypiperidine-1-carboxylate To a solution of tert-butyl 4-hydroxypiperidine-1-carboxylate (2 g, 9.94 mmol, 1 equiv.) in THF (20 mL) was cooled to 0 °C. NaH (476.94 mg, 11.92 mmol, 60% purity, 1.2 equiv.) was added into the mixture at 0°C. The reaction mixture was stirred at 0 °C for 0.5 hour. Then 3- bromoprop-1-yne (1.18 g, 9.94 mmol, 856.62 μL, 1 equiv.) was added into the mixture. The reaction mixture was stirred at 25 °C for 12 hours. Once completed, the reaction mixture was quenched with water (20 mL), then extracted with ethyl acetate (20 mL × 3). The organic layers were washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo. The residue was purified by flash silica gel chromatography (Eluent of 0~8% ethyl acetate/petroleum ether) to give tert-butyl 4-prop-2-ynoxypiperidine-1- carboxylate (2.4 g, 8.8 mmol, 88.8% yield, 88% purity) as a yellow solid. MS (ESI) m/z: 184.2 [M-56+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 4.17 (d, J = 2.4 Hz, 2H), 3.67 - 3.56 (m, 3H), 3.38 (t, J = 2.4 Hz, 1H), 3.02 (t, J = 9.6 Hz, 2H), 1.82 - 1.74 (m, 2H), 1.41 - 1.37 (m, 9H), 1.35 - 1.27 (m, 2H) Step B. Procedure for preparation of tert-butyl 4-[3-(4-bromo-3-methyl-phenyl)prop-2- ynoxy]piperidine-1-carboxylate A mixture of tert-butyl 4-prop-2-ynoxypiperidine-1-carboxylate (2 g, 8.36 mmol, 1 equiv.), 1- bromo-4-iodo-2-methyl-benzene (2.48 g, 8.36 mmol, 1 equiv.), TEA (2.54 g, 25.07 mmol, 3.49 mL, 3 equiv.), Pd(PPh3)2Cl2 (586.60 mg, 835.74 μmol, 0.1 equiv.), and CuI (318.33 mg, 1.67 mmol, 0.2 equiv.) in DMF (20 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 60 °C for 3 hours under N2 atmosphere. The reaction mixture was quenched by addition H2O (50 mL) and extracted with EtOAc (50 mL × 3). The combined organic layers were washed with saturated salt solution (50 mL × 1), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~5% Ethylacetate/petroleum ether) to give tert- butyl 4-[3-(4-bromo-3-methyl-phenyl)prop-2-ynoxy]piperidine-1-carb oxylate (2.4 g, 4.8 mmol, 57.0% yield, 81% purity) as a yellow oil. MS (ESI) m/z: 308.1 [M-100+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 7.58 (d, J = 8.0 Hz, 1H), 7.45 (d, J = 1.6 Hz, 1H), 7.20 (dd, J = 2.0, 8.4 Hz, 1H), 4.41 (s, 2H), 3.76 - 3.56 (m, 3H), 3.05 (t, J = 10.0 Hz, 2H), 2.33 (s, 3H), 1.90 - 1.77 (m, 2H), 1.39 (s, 9H), 1.38 - 1.32 (m, 2H) Step C. Procedure for preparation of tert-butyl 4-[3-(4-bromo-3-methyl-phenyl)propoxy] piperidine-1-carboxylate To a solution of tert-butyl 4-[3-(4-bromo-3-methyl-phenyl)prop-2-ynoxy]piperidine-1- carboxylate (2.2 g, 5.39 mmol, 1 equiv.) in EtOAc (20 mL) was added Rh/Al 2 O 3 (1.11 g, 538.78 μmol, 5% purity, 0.1 equiv.) under N 2 atmosphere. The suspension was degassed and purged with H2 three times. The mixture was stirred under H2 (15 psi) at 25 °C for 2 hours. The reaction solution was filtered to give tert-butyl 4-[3-(4-bromo-3-methyl-phenyl) propoxy]piperidine-1- carboxylate (2.2 g, 4.8 mmol, 90.8% yield, 91.7% purity) as a brown oil. MS (ESI) m/z: 356.2 [M-56+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 7.42 (d, J = 8.0 Hz, 1H), 7.06 (s, 1H), 6.91 - 6.85 (m, 1H), 3.81 - 3.72 (m, 2H), 3.46 - 3.38 (m, 3H), 3.13 - 3.03 (m, 2H), 2.63 (t, J = 7.6 Hz, 2H), 2.37 (s, 3H), 1.90 - 1.78 (m, 4H), 1.55 - 1.48 (m, 2H), 1.47 (s, 9H) Step D. Procedure for preparation of 4-[3-(4-bromo-3-methyl-phenyl)propoxy]piperidine A mixture of tert-butyl 4-[3-(4-bromo-3-methyl-phenyl)propoxy]piperidine-1-carboxyla te (2 g, 4.85 mmol, 1 equiv.) in HCl/EtOAc (20 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 25 °C for 2 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give 4-[3-(4-bromo-3-methyl- phenyl)propoxy]piperidine (2 g, crude) as a brown solid. 1 H NMR (400 MHz, DMSO-d6) δ = 7.43 (d, J = 8.0 Hz, 1H), 7.05 (d, J = 1.6 Hz, 1H), 6.86 (dd, J = 2.0, 8.0 Hz, 1H), 3.61 (s, 1H), 3.40 (t, J = 6.2 Hz, 2H), 3.35 - 3.14 (m, 4H), 2.66 - 2.57 (m, 2H), 2.38 (s, 3H), 2.13 (s, 1H), 2.10 - 2.06 (m, 2H), 1.99 - 1.91 (m, 2H), 1.90 - 1.81 (m, 2H) Step E. Procedure for preparation of ethyl 2-[4-[3-(4-bromo-3-methyl-phenyl)propoxy]- 1-piperidyl]acetate A mixture of 4-[3-(4-bromo-3-methyl-phenyl)propoxy]piperidine (2 g, 5.74 mmol, 1 equiv., HCl), ethyl 2-bromoacetate (862.05 mg, 5.16 mmol, 571.27 μL, 0.9 equiv.) and K2CO3 (2.38 g, 17.21 mmol, 3 equiv.) in CH 3 CN (20 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 40 °C for 4 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~28%Ethylacetate/petroleum ether) to give ethyl 2-[4-[3-(4-bromo-3-methyl-phenyl)propoxy]-1-piperidyl]acetat e (1.8 g, 4.3 mmol, 74.8% yield, 94.9% purity) as a yellow oil. MS (ESI) m/z: 398.2 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.42 (d, J = 8.0 Hz, 1H), 7.07 (d, J = 1.6 Hz, 1H), 6.88 (dd, J = 2.0, 8.0 Hz, 1H), 4.19 (q, J = 7.2 Hz, 2H), 3.41 (t, J = 6.4 Hz, 2H), 3.33 - 3.26 (m, 1H), 3.22 (s, 2H), 2.87 - 2.77 (m, 2H), 2.62 (t, J = 7.6 Hz, 2H), 2.41 - 2.32 (m, 4H), 1.98 - 1.81 (m, 4H), 1.71 - 1.65 (m, 2H), 1.28 (t, J = 7.2 Hz, 3H) Step F. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3, 2-dioxaborolan-2- yl)pyridine-2-carboxylate A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-bromo-pyridine-2-carboxylate (1.5 g, 2.65 mmol, 1 equiv.), 4,4,5,5-tetramethyl-1,3,2- dioxaborolane (1.70 g, 13.26 mmol, 1.92 mL, 5 equiv.), N,N-diethylethanamine (805.25 mg, 7.96 mmol, 1.11 mL, 3 equiv.), and cyclopentyl(diphenyl)phosphane;dichloropalladium;iron (194.09 mg, 265.26 umol, 0.1 equiv.) in THF (3 mL) and CH3CN (10 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 100 °C for 1 hour under N2 atmosphere in microwave. The reaction was filtered to give tert-butyl 6-[8-(1,3-benzothiazol- 2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-t etramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (2 g, 3.2 mmol, 41.0% yield) as a white solid. MS (ESI) m/z: 613.3 [M+H] + Step G. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[3-[[1-(2-ethoxy-2-oxo -ethyl)-4- piperidyl]oxy]propyl]-2-methyl-phenyl]pyridine-2-carboxylate A mixture of ethyl 2-[4-[3-(4-bromo-3-methyl-phenyl)propoxy]-1-piperidyl]acetat e (400 mg, 1.00 mmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (738.13 mg, 1.21 mmol, 1.2 equiv.), Ad2nBuP Pd G3 (73.13 mg, 100.42 μmol, 0.1 equiv.), and K2CO3 (416.35 mg, 3.01 mmol, 3 equiv.) in dioxane (5 mL) and H2O (2 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 100 °C for 1 hour under N 2 atmosphere in microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~51% ethyl acetate/petroleum ether) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[4-[3-[[1-(2-ethoxy-2-oxo-eth yl)-4-piperidyl]oxy]propyl]-2- methyl-phenyl]pyridine-2-carboxylate (500 mg, 590.8 μmol, 58.8% yield, 95% purity) as a yellow oil. MS (ESI) m/z: 403.0 [M/2+H] + Step H. Procedure for preparation of 2-[4-[3-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyr idyl]-3-methyl- phenyl]propoxy]-1-piperidyl]acetic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[4-[3-[[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]oxy]propy l]-2-methyl-phenyl]pyridine-2- carboxylate (200 mg, 248.75 μmol, 1 equiv.), LiOH•H 2 O (41.75 mg, 995.02 μmol, 4 equiv.) in THF (8 mL) and H 2 O (2 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 25 °C for 12 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to remove THF. The residue was diluted with H2O (5 mL) and adjusted to pH = 4~5 with HCl. Then the mixture was extracted with EtOAc (50 mL × 3). The combined organic layers were washed with saturated salt solution (50 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give 2-[4-[3-[4-[6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -2-tert-butoxycarbonyl-3- pyridyl]-3-methyl-phenyl]propoxy]-1-piperidyl]acetic acid (260 mg, crude) as a yellow solid. MS (ESI) m/z: 776.6 [M+H] + Step I. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[4-[3-[[1-[2-[[3-(2,6-dioxo-3 -piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-4-piperidyl]oxy]propyl]-2-methyl-ph enyl]pyridine-2- carboxylate A mixture of 2-[4-[3-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H-isoquinolin- 2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-3-methyl-phenyl]propo xy]-1-piperidyl]acetic acid (200.0 mg, 257.75 μmol, 1 equiv.), 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (79.88 mg, 309.30 μmol, 1.2 equiv.), EDCI (74.12 mg, 386.62 μmol, 1.5 equiv.) in pyridine (2 mL) was degassed and purged with N2 for three times, and then the mixture was stirred at 25 °C for 2 hours under N 2 atmosphere. The reaction mixture was quenched by addition H 2 O (20 mL) and extracted with EtOAc (20 mL × 5). The combined organic layers were washed with saturated salt solution (20 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[4-[3-[[1-[2-[[3-(2,6-dioxo-3-piperidyl) -1-methyl-indazol-6-yl]amino]-2- oxo-ethyl]-4-piperidyl]oxy]propyl]-2-methyl-phenyl]pyridine- 2-carboxylate (500 mg, crude) as a brown oil. MS (ESI) m/z: 1016.6 [M+H] + Step J. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[4-[3-[[1-[2-[[3-(2,6-dioxo-3-piperid yl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]oxy]propyl]-2-methyl-phen yl]pyridine-2-carboxylic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[4-[3-[[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-inda zol-6-yl]amino]-2-oxo-ethyl]-4- piperidyl]oxy]propyl]-2-methyl-phenyl]pyridine-2-carboxylate (150 mg, 147.61 μmol, 1 equiv.) in TFA (2 mL) and DCM (2 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 40 °C for 2 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep- HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4- [3-[[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl] amino]-2-oxo-ethyl]-4- piperidyl]oxy]propyl]-2-methyl-phenyl]pyridine-2-carboxylic acid (46.3 mg, 47.3 μmol, 32.0% yield, 98.9% purity) as a yellow solid. MS (ESI) m/z: 960.7 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.88 (s, 1H), 9.87 (s, 1H), 8.07 - 8.00 (m, 2H), 7.79 (d, J = 8.4 Hz, 1H), 7.63 (d, J = 8.8 Hz, 2H), 7.51 - 7.42 (m, 3H), 7.40 - 7.31 (m, 2H), 7.23 (d, J = 8.8 Hz, 1H), 7.05 (s, 1H), 7.01 - 6.89 (m, 3H), 4.97 (s, 2H), 4.32 (dd, J = 4.8, 9.2 Hz, 1H), 3.92 (s, 3H), 3.43 (t, J = 6.4 Hz, 4H), 3.15 (s, 2H), 3.02 ( t, J = 5.6 Hz, 2H), 2.78 (dd, J = 4.4, 6.4 Hz, 2H), 2.67 (s, 1H), 2.62 - 2.57 (m, 5H), 2.32 (d, J = 5.2 Hz, 2H), 2.21 - 2.13 (m, 1H), 2.04 (s, 3H), 1.87 (d, J = 9.6 Hz, 2H), 1.76 (s, 2H), 1.63 - 1.51 (m, 2H) Example 84. Preparation of Compound 122 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[4-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-oxo-e thyl]-1-piperidyl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[4-[2-[[3-(2,6-diox o-3-piperidyl)-1-methyl- indazol-7-yl]amino]-2-oxo-ethyl]-1-piperidyl]propoxy]-2-meth yl-phenyl]pyridine-2- carboxylate A mixture of 2-[1-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H-isoquinolin- 2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]prop yl]-4-piperidyl]acetic acid (107.79 mg, 138.92 µmol, 1 equiv.), 3-(7-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (71.76 mg, 277.84 µmol, 2.0 equiv.), HATU (105.64 mg, 277.84 µmol, 2.0 equiv.), and DIEA (17.95 mg, 138.92 µmol, 24.20 µL, 1 equiv.) in DMF (2 mL) was stirred at 25 o C for 12 hours. The reaction mixture was diluted with water (10 mL), filtered, and concentrated under reduced pressure to give the crude product tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[4-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol-7- yl]amino]-2-oxo-ethyl]-1-piperidyl]propoxy]-2-methyl-phenyl] pyridine-2-carboxylate (90 mg, 88.5 µmol, 63.7% yield) as a yellow solid. MS (ESI) m/z: 508.8 [M/2+H] + Step B. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[4-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-7- yl]amino]-2-oxo-ethyl]-1-piperidyl]propoxy]-2-methyl-phenyl] pyridine-2-carboxylic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[4-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indaz ol-7-yl]amino]-2-oxo-ethyl]-1- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate (90 mg, 88.56 µmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL) was stirred at 40 o C for 4 hours. The reaction mixture was concentrated under reduced pressure to give the crude product. The crude product was purified by prep-HPLC to give compound 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[3-[4-[2-[[3-(2,6-dioxo-3-piperidyl)- 1-methyl-indazol-7-yl]amino]-2- oxo-ethyl]-1-piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-c arboxylic acid (25 mg, 24.4 µmol, 27.6% yield, 93.9% purity) as a white solid. MS (ESI) m/z: 960.3 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 10.91 (s, 1H), 9.86 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.61 (dd, J = 7.6, 13.2 Hz, 2H), 7.45 - 7.35 (m, 6H), 7.11 - 7.06 (m, 3H), 6.94 - 6.87 (m, 2H), 6.65 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.36 (dd, J =5.2, 10.4 Hz, 1H), 4.05 (s, 3H), 3.94 - 3.90 (m, 5H), 3.05 - 2.94 (m, 6H), 2.73 - 2.58 (m, 3H), 2.36 - 2.32 (m, 3H), 2.22 - 2.00 (m, 4H), 1.91 (s, 3H), 1.74 ( d, J = 12.4 Hz, 2H), 1.33-1.29 (m, 2H) Example 85. Preparation of Compound 145 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(1-(2-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin -1-yl)-2-oxoethyl)piperidin-4- yl)ethoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(1-(2-(4-(3-(2,6-dioxop iperidin-3-yl)-1-methyl-1H- indazol-7-yl)piperazin-1-yl)-2-oxoethyl)piperidin-4-yl)ethox y)-2- methylphenyl)picolinate To a solution of 2-(4-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)ethyl)piperidin-1-yl)acetic acid (100 mg, 131.25 µmol, 1.00 equiv.) in DMF (1 mL) was added DIEA (50.89 mg, 393.74 µmol, 68.58 µL, 3.00 equiv.), HATU (59.88 mg, 157.50 µmol, 1.20 equiv.), and 3-(1-methyl- 7-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dione (52.53 mg, 144.37 µmol, 1.10 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction was diluted with water (20 mL) and extracted with DCM (30 mL × 3). The combined organic layers were washed with brine (20 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give tert- butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(1-(2- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pip erazin-1-yl)-2- oxoethyl)piperidin-4-yl)ethoxy)-2-methylphenyl)picolinate (130 mg, 119.8 µmol, 91.3% yield) as a yellow solid. MS (ESI) m/z: 1071.8 [M+H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(1-(2-(4-(3-(2,6-dioxop iperidin-3-yl)-1-methyl-1H- indazol-7-yl)piperazin-1-yl)-2-oxoethyl)piperidin-4-yl)ethox y)-2-methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(1-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-7-yl)piperazin- 1-yl)-2-oxoethyl)piperidin-4-yl)ethoxy)-2-methylphenyl)picol inate (130 mg, 121.35 µmol, 1.00 equiv.) in DCM (1 mL) was added TFA (1.67 g, 14.63 mmol, 1.0 mL, 120.57 equiv.). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(2-(1-(2-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin-1-yl )-2-oxoethyl)piperidin-4- yl)ethoxy)-2-methylphenyl)picolinic acid (20.6 mg, 18.4 µmol, 15.1% yield, 95.6% purity) as a white solid. MS (ESI) m/z: 1015.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.89 (s, 1H), 8.06 - 7.99 (m, 1H), 7.78 (d, J = 8.8 Hz, 1H), 7.63 (d, J = 7.6 Hz, 1H), 7.52 - 7.28 (m, 7H), 7.10 - 7.00 (m, 3H), 6.94 - 6.85 (m, 2H), 6.65 (d, J = 7.2 Hz, 1H), 5.02 - 4.96 (m, 2H), 4.37 - 4.33 (m, 1H), 4.28 (s, 3H), 4.00 (t, J = 6.0 Hz, 2H), 3.94 - 3.90 (m, 2H), 3.06 - 3.00 (m, 4H), 2.87 - 2.80 (m, 4H), 2.64 - 2.59 (m, 6H), 2.21 - 2.13 (m, 2H), 1.98 (d, J = 10.4 Hz, 2H), 1.89 (s, 3H), 1.74 - 1.67 (m, 4H), 1.52 - 1.47 (m, 1H), 1.28 - 1.20 (m, 4H) Example 86. Preparation of Compound 146b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4s)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pip erazin-1-yl)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) - 3, 4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-(3-ethoxy-3-oxo-propy l) cyclohexoxy]-2- methyl-phenyl]pyridine-2-carboxylate tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-3-(4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridine-2-carboxylate (737.46 mg, 1.20 mmol, 1.3 equiv.), ethyl 3-[4-(3-bromo-2-methyl-phenoxy) cyclohexyl]propanoate (400 mg, 926.10 μmol, 85.5% purity, 1 equiv.), KF (161.42 mg, 2.78 mmol, 65.09 μL, 3 equiv.), and [2-(2- aminophenyl) phenyl]palladium (1+) ;bis (1-adamantyl) -butyl-phosphane;methanesulfonate (134.89 mg, 185.22 μmol, 0.2 equiv.) were taken up into a microwave tube in dioxane (4 mL) and H 2 O (0.4 mL). The sealed tube was heated at 100 °C for 60 min under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 50% ethyl acetate/petroleum ether). The compound tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-(3-ethoxy-3-oxo-propy l) cyclohexoxy]-2-methyl- phenyl]pyridine-2-carboxylate (630 mg, 734.1 μmol, 79.3% yield, 90.3% purity) was obtained as a yellow oil. MS (ESI) m/z: 775.6 [M+H] + . Step B. Procedure for preparation of 3-[4-[3-[6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyrid yl]-2-methyl- phenoxy]cyclohexyl]propanoic acid To a solution of tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H- isoquinolin-2-yl]-3-[3-[4-(3-ethoxy-3-oxo-propyl) cyclohexoxy]-2-methyl-phenyl]pyridine-2- carboxylate (600 mg, 774.23 μmol, 1 equiv.) in THF (6 mL) and H 2 O (0.6 mL) was added LiOH•H2O (97.47 mg, 2.32 mmol, 3 equiv.). The mixture was stirred at 25 °C for 40 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was treated with water (4 mL). The pH of the mixture was adjusted to around 3 by progressively adding diluted HCl. The mixture was filtered to give 3-[4-[3-[6-[8-(1, 3-benzothiazol-2- ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyrid yl]-2-methyl- phenoxy]cyclohexyl]propanoic acid (557 mg, 618.96 μmol, 79.95% yield, 83% purity) as a yellow solid. MS (ESI) m/z: 747.6 [M+H] + . Step C. Procedure for preparation of tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) - 3, 4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[4-[3-(2, 6-dioxo-3-piperidyl) -1-methyl- indazol-7-yl]piperazin-1-yl]-3-oxo-propyl] cyclohexoxy]-2-methyl-phenyl]pyridine-2- carboxylate To a solution of 3-[4-[3-[6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]cyclohexyl]propanoic acid (200 mg, 267.77 μmol, 1 equiv.) in DMF (1 mL) was added HATU (152.72 mg, 401.65 μmol, 1.5 equiv.), DIPEA (103.82 mg, 803.31 μmol, 139.92 μL, 3 equiv.), and 3-(1-methyl-7- piperazin-1-yl-indazol-3-yl) piperidine-2, 6-dione (131.49 mg, 401.65 μmol, 1.5 equiv.) at 25 °C. The reaction mixture was stirred at 25 °C for 16 hours. The mixture was triturated with water (5 mL) and filtered to give a residue. The compound tert-butyl 6-[8-(1, 3-benzothiazol- 2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[4-[3-(2, 6-dioxo-3-piperidyl) - 1-methyl-indazol-7-yl]piperazin-1-yl]-3-oxo-propyl]cyclohexo xy]-2-methyl-phenyl]pyridine- 2-carboxylate (270 mg, 180.9 μmol, 67.5% yield, 70.8% purity) was obtained as a yellow solid. MS (ESI) m/z: 1056.4 [M+H] + . Step D. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4s)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)piperazin-1-yl)-3-oxopropyl)cyclohexyl)oxy)- 2-methylphenyl)picolinic acid A solution of tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin- 2-yl]-3-[3-[4-[3-[4-[3-(2, 6-dioxo-3-piperidyl) -1-methyl-indazol-7-yl]piperazin-1-yl]-3-oxo- propyl] cyclohexoxy]-2-methyl-phenyl]pyridine-2-carboxylate (240 mg, 227.21 μmol, 1 eq) in DCM (1 mL) and TFA (1 mL) was stirred at 25 °C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC. The compound 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4s)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-7- yl)piperazin-1-yl)-3-oxopropyl)cyclohexyl)oxy)-2-methylpheny l)picolinic acid (67.2 mg, 66.5 μmol, 29.2% yield, 98.9% purity) was obtained as a white solid. MS (ESI) m/z: 1000.4 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.99 - 12.23 (m, 2H), 10.89 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.52 - 7.31 (m, 6H), 7.06 (d, J = 8.0 Hz, 1H), 7.04 - 6.99 (m, 2H), 6.96 (d, J = 8.8 Hz, 1H), 6.89 (d, J = 8.4 Hz, 1H), 6.60 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.61 (s, 1H), 4.53 - 4.40 (m, 1H), 4.34 (dd, J = 5.2, 9.6 Hz, 1H), 4.26 (s, 3H), 3.91 (t, J = 5.6 Hz, 3H), 3.25 - 3.10 (m, 2H), 3.02 (t, J = 5.6 Hz, 2H), 2.72 - 2.52 (m, 4H), 2.47 - 2.26 (m, 4H), 2.18 (d, J = 5.4 Hz, 1H), 2.01 - 1.84 (m, 5H), 1.67 - 1.18 (m, 10H) Example 87. Preparation of Compound 150 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[3-[4-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]piperazin-1-yl ]propyl]phenoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo -3-piperidyl)-1-methyl- indazol-7-yl]piperazin-1-yl]propyl]phenoxy]-2-methyl-phenyl] pyridine-2-carboxylate A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(2-methyl-3-(4-(3-oxopropyl)phenoxy)phenyl)picolinate (100 mg, 137.96 µmol, 1.0 equiv.), 3-(1-methyl-7-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (54.20 mg, 165.55 µmol, 1.2 equiv.), HCOOH (6.63 mg, 137.96 µmol, 1.0 equiv.), and NaBH(OAc)3 (43.86 mg, 206.94 µmol, 1.5 equiv.) in DCM (2 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 15 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The compound tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[4-[3-[4-[3-(2,6-dioxo- 3-piperidyl)-1-methyl-indazol-7-yl]piperazin-1-yl]propyl]phe noxy]-2-methyl- phenyl]pyridine-2-carboxylate (150 mg, crude) was obtained as a yellow oil. MS (ESI) m/z: 1036.7 [M+H] + . Step B. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo-3-piperidyl )-1-methyl-indazol-7- yl]piperazin-1-yl]propyl]phenoxy]-2-methyl-phenyl]pyridine-2 -carboxylic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazo l-7-yl]piperazin-1- yl]propyl]phenoxy]-2-methyl-phenyl]pyridine-2-carboxylate (150 mg, 144.75 µmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL) was stirred at 40 °C for 21 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo-3-piperidyl)-1 -methyl-indazol-7-yl]piperazin-1- yl]propyl]phenoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid (30.1 mg, 29.9 µmol, 20.6% yield, 97.4% purity) as a white solid. MS (ESI) m/z: 981.0 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.89 (s, 1H), 8.15 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 7.6 Hz, 1H), 7.53 (d, J = 8.8 Hz, 1H), 7.50 - 7.31 (m, 6H), 7.21 - 7.15 (m, 3H), 7.05 - 6.98 (m, 3H), 6.89 (d, J = 7.6 Hz, 1H), 6.86 - 6.80 (m, 3H), 5.00 (s, 2H), 4.34 (dd, J = 5.2, 9.6 Hz, 1H), 4.23 (s, 3H), 3.93 (t, J = 6.0 Hz, 2H), 3.04 (d, J = 5.6 Hz, 2H), 2.98 - 2.84 (m, 4H), 2.71 - 2.55 (m, 7H), 2.40 (t, J = 6.8 Hz, 2H), 2.36 - 2.28 (m, 2H), 2.21 - 2.13 (m, 1H), 1.91 (s, 3H), 1.81 - 1.74 (m, 2H) Example 88. Preparation of Compound 162b 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[(1R)-3-[1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-o xo-ethyl]-4-piperidyl]-1- methyl-propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 4-[(3R)-3-(3-bromo-2-methyl- phenoxy)butyl]piperidine-1-carboxylate A mixture of tert-butyl 4-[(3S)-3-hydroxybutyl]piperidine-1-carboxylate (1.08 g, 4.20 mmol, 1 equiv.), 3-bromo-2-methyl-phenol (784.85 mg, 4.20 mmol, 1 equiv.), and 2-(tributyl-λ 5 - phosphanylidene)acetonitrile (1.22 g, 5.04 mmol, 1.2 equiv.) in toluene (2 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 120 °C for 1 hour under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 10/1 to 3/1) to give tert-butyl 4-[(3R)-3-(3-bromo-2-methyl-phenoxy)butyl]piperidine-1- carboxylate (1.3 g, 3.0 mmol, 72.6% yield) as a colorless oil. 1 H NMR (400 MHz, CDCl 3 ,) δ = 7.13 (t, J = 8.4 Hz, 1H), 6.98 (t, J = 8.0 Hz, 1H), 6.76 (d, J = 8.4 Hz, 1H), 4.36 - 4.25 (m, 1H), 4.16 - 4.01 (m, 2H), 2.68 (t, J = 12.4 Hz, 2H), 2.30 (s, 3H), 1.80 - 1.58 (m, 5H), 1.47 (s, 9H), 1.44 - 1.35 (m, 2H), 1.29 (d, J = 6.0 Hz, 3H), 1.16 - 1.05 (m, 2H). Step B. Procedure for preparation of 4-[(3R)-3-(3-bromo-2-methyl- phenoxy)butyl]piperidine A solution of tert-butyl 4-[(3R)-3-(3-bromo-2-methyl-phenoxy)butyl]piperidine-1-carbo xylate (1.3 g, 3.05 mmol, 1 equiv.) in HCl/EtOAc (1 mL) was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=10/1 to 3/1) to give 4-[(3R)-3-(3-bromo-2-methyl-phenoxy)butyl]piperidine (650 mg, 1.9 mmol, 65.3% yield) as a colorless oil. 1 H NMR (400 MHz, CDCl 3 ,) δ = 9.59 (s, 1H), 7.13 (d, J = 8.0 Hz, 1H), 6.98 (t, J = 8.0 Hz, 1H), 6.74 (d, J = 8.4 Hz, 1H), 4.39 - 4.20 (m, 1H), 3.48 (d, J = 12.4 Hz, 2H), 2.84 (m, J = 12.0 Hz, 2H), 2.28 (s, 3H), 1.90 (d, J = 13.6 Hz, 2H), 1.71 - 1.37 (m, 7H), 1.28 (d, J = 6.0 Hz, 3H). Step C. Procedure for preparation of ethyl 2-[4-[(3R)-3-(3-bromo-2-methyl-phenoxy) butyl]-1-piperidyl] acetate To a solution of 4-[(3R)-3-(3-bromo-2-methyl-phenoxy)butyl]piperidine (650 mg, 1.99 mmol, 1 equiv.), and ethyl 2-bromoacetate (332.70 mg, 1.99 mmol, 220.33 µL, 1 equiv.) in CH 3 CN (3 mL) was added K 2 CO 3 (275.34 mg, 1.99 mmol, 1 equiv.). The mixture was stirred at 60 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=10/1 to 2/1) to give ethyl 2-[4-[(3R)-3-(3-bromo-2-methyl-phenoxy) butyl]-1- piperidyl] acetate (600 mg, 1.46 mmol, 73.04% yield) as a yellow oil. MS (ESI) m/z: 414.2 [M+H] + 1 H NMR (400 MHz, CDCl 3 ,) δ = 7.13 (d, J = 8.0 Hz, 1H), 6.98 (t, J = 8.0 Hz, 1H), 6.76 (d, J = 8.4 Hz, 1H), 4.35 - 4.26 (m, 1H), 4.19 (q, J = 7.2 Hz, 2H), 3.19 (s, 2H), 2.93 (d, J = 11.2 Hz, 2H), 2.30 (s, 3H), 2.13 (t, J = 11.2 Hz, 2H), 1.80 - 1.56 (m, 6H), 1.36 - 1.26 (m, 9H). Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[(1R)-3-[1-(2-ethoxy-2 -oxo-ethyl)-4-piperidyl]-1- methyl-propoxy]-2-methyl-phenyl]pyridine-2-carboxylate A mixture of ethyl 2-[4-[(3R)-3-(3-bromo-2-methyl-phenoxy)butyl]-1-piperidyl]ac etate (700 mg, 1.70 mmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (1.04 g, 1.70 mmol, 1 equiv.), [2-(2-aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-buty l- phosphane;methanesulfonate (123.63 mg, 169.75 µmol, 0.1 equiv.), KF (1.5 M, 3.40 mL, 3 equiv.) in dioxane (5 mL) and water (1 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 100 °C for 1 hour under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 10/1 to 2/1) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[(1R)-3-[1-(2- ethoxy-2-oxo-ethyl)-4-piperidyl]-1-methyl-propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (850 mg, 1.0 mmol, 61.21% yield) as a yellow oil. MS (ESI) m/z: 818.7 [M+H] + 1 H NMR (400 MHz, CDCl 3 ,) δ = 7.86 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 8.0 Hz, 1H), 7.57 (d, J = 7.2 Hz, 1H), 7.43 - 7.28 (m, 5H), 7.08 (t, J = 8.0 Hz, 1H), 6.90 (d, J = 8.8 Hz, 1H), 6.80 (d, J = 8.0 Hz, 1H), 6.67 (d, J = 7.6 Hz, 1H), 5.23 - 4.85 (m, 2H), 4.42 - 4.29 (m, 1H), 4.14 - 4.10 (m, 4H), 3.22 (s, 2H), 3.07 (t, J = 6.4 Hz, 2H), 3.01 - 2.88 (m, 2H), 2.25 - 2.09 (m, 2H), 2.05 (s, 5H), 1.68 (s, 4H), 1.50 - 1.41 (m, 2H), 1.32 (d, J = 5.6 Hz, 3H), 1.26 (d, J = 3.2 Hz, 4H), 1.14 (s, 9H). Step E. Procedure for preparation of 2-[4-[(3R)-3-[3-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-2- methyl-phenoxy]butyl]-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[(1R)-3-[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]-1- methyl-propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (200 mg, 244.49 µmol, 1 equiv.) in THF (0.5 mL), H2O (0.5 mL) was added LiOH•H2O (51.30 mg, 1.22 mmol, 5 equiv.). The mixture was stirred at 25 °C for 2 hours. The pH was adjusted to 6 with 1M HCl. The residue was extracted with DCM (2mL × 3). The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=10/1 to 3/1) to give 2-[4-[(3R)-3-[3-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-2-methyl- phenoxy]butyl]-1-piperidyl]acetic acid (160 mg, 202.54 µmol, 82.84% yield) as a yellow solid. MS (ESI) m/z: 790.7 [M+H] + Step F. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[(1R)-3-[1-[2-[[3-(2,6 -dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-4-piperidyl]-1-methyl-propo xy]-2-methyl- phenyl]pyridine-2-carboxylate To a solution of 2-[4-[(3R)-3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]butyl]-1- piperidyl]acetic acid (80 mg, 101.27 µmol, 1 equiv.),3-(6-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (28.77 mg, 111.40 µmol, 1.1 equiv.) in DMF (1 mL) was added HATU (57.76 mg, 151.90 µmol, 1.5 equiv.) and DIEA (39.26 mg, 303.81 µmol, 52.92 µL, 3 equiv.). The mixture was stirred at 25 °C for 2 hours. The mixture was quenched by addition of water (2 mL) at 25 °C when precipitation occurred. The mixture was then filtered and concentrated under reduced pressure to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[(1R)-3-[1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]-1-methyl-propoxy]-2-meth yl-phenyl]pyridine-2- carboxylate (100 mg, 97.0 µmol, 95.8% yield) as a white solid. MS (ESI) m/z: 1030.8 [M+H] Step G. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[(1R)-3-[1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]-1-methyl-propoxy]-2-meth yl-phenyl]pyridine-2- carboxylic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[(1R)-3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo- ethyl]-4-piperidyl]-1-methyl-propoxy]-2-methyl-phenyl]pyridi ne-2-carboxylate (100 mg, 97.06 µmol, 1 equiv.) in DCM (1 mL) and TFA (1 mL) was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[(1R)-3-[1-[2-[[3-(2,6-dioxo-3-piperi dyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]-1-methyl-propoxy]-2-meth yl-phenyl]pyridine-2- carboxylic acid (31.6 mg, 31.2 µmol, 32.1% yield, 95.9% purity) as a white solid. MS (ESI) m/z: 974.7[M+H] 1 H NMR (400 MHz, DMSO-d6,) δ = 13.07 - 12.65 (s, 1H), 10.87 (s, 1H), 9.99 - 9.72 (s, 1H), 8.13 (s, 1H), 8.08 - 7.97 (m, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.66 - 7.59 (m, 2H), 7.50 - 7.41 (m, 3H), 7.40 - 7.31 (m, 2H), 7.20 (d, J = 8.4 Hz, 1H), 7.12 - 7.04 (m, 1H), 6.96 (d, J = 8.8 Hz, 1H), 6.89 (d, J = 8.8 Hz, 1H), 6.61 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.45 - 4.37 (m, 1H), 4.32 (dd, J = 4.8, 9.6 Hz, 1H), 3.92 (s, 5H), 3.02 (t, J = 5.6 Hz, 2H), 2.94 - 2.85 (m, 2H), 2.70 - 2.59 (m, 4H), 2.38 - 2.28 (m, 2H), 2.21 - 2.13 (m, 2H), 1.88 (s, 3H), 1.71 - 1.63 (m, 3H), 1.45 - 1.21 (m, 9H). Example 89. Preparation of Compound 164a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-((2S)-1- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino )-1-oxopropan-2-yl)piperidin- 4-yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of (S)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(1-ethoxy-1-oxop ropan-2-yl)piperidin-4- yl)propoxy)-2-methylphenyl)picolinate A mixture of (S)-ethyl 2-(4-(3-(3-bromo-2-methylphenoxy)propyl)piperidin-1-yl)propa noate (450 mg, 1.09 mmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)picolinate (735.30 mg, 1.20 mmol, 1.1 equiv.) in dioxane (10 mL) was added KF (1.5 M, 2.18 mL, 3 equiv.) and Ad2nBuP Pd G3(cataCXium® A Pd G3 ) (79.47 mg, 109.13 µmol, 0.1 equiv.). After addition, the mixture was degassed and purged with N2 three times, and then the mixture was stirred at 100 °C for 2 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~45% ethyl acetate/petroleum ether) to give (S)-tert-butyl 6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2( 1H)-yl)-3-(3-(3-(1-(1-ethoxy- 1-oxopropan-2-yl)piperidin-4-yl)propoxy)-2-methylphenyl)pico linate (600 mg, 690.7 µmol, 63.2% yield, 94.1% purity) as a yellow solid. MS (ESI) m/z: 818.5 [M+H] + Step B. Procedure for preparation of (S)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)propyl)piperidin-1-yl)propanoic acid To a solution of (S)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(1-ethoxy-1-oxopropan-2-yl)piperidin-4- yl)propoxy)-2- methylphenyl)picolinate (600 mg, 733.47 µmol, 1 equiv.) in THF (6 mL) was added LiOH•H 2 O (92.34 mg, 2.20 mmol, 3 equiv.) and H 2 O (1.5 mL). The mixture was stirred at 40 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to remove THF. The aqueous phase was adjusted to pH=4~5 with 1M HCl. The reaction mixture was filtered and diluted in ethyl acetate. The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give (S)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)propyl)piperidin-1-yl)propanoic acid (600 mg, crude) as a yellow solid. MS (ESI) m/z: 790.8 [M+H] + Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-((2S)-1-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-1-oxopropan-2-yl)piperidin-4-yl)propo xy)-2- methylphenyl)picolinate To a solution of (S)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)piperidin-1- yl)propanoic acid (100 mg, 126.59 µmol, 1 equiv.), 3-(6-amino-1-methyl-1H-indazol-3- yl)piperidine-2,6-dione (39.23 mg, 151.90 µmol, 1.2 equiv.) in pyridine (1 mL) was added EDCI (36.40 mg, 189.88 µmol, 1.5 equiv.). The mixture was stirred at 25 °C for 3 hours. The reaction mixture was quenched by addition H2O 2mL. The reaction mixture was filtered and washed with 5 mL of water. After addition, the filter cake was diluted in DCM 10 mL. The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-((2S)-1-((3-(2,6-dioxopiperidin-3-yl)-1 -methyl-1H-indazol-6-yl)amino)- 1-oxopropan-2-yl)piperidin-4-yl)propoxy)-2-methylphenyl)pico linate (100 mg, crude) as a red solid. MS (ESI) m/z: 1030.8 [M+H] + Step D. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-((2S)-1-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-1-oxopropan-2-yl)piperidin-4-yl)propo xy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-((2S)-1-((3-(2,6-dioxopiperidin-3-yl)-1 -methyl-1H-indazol-6-yl)amino)- 1-oxopropan-2-yl)piperidin-4-yl)propoxy)-2-methylphenyl)pico linate (100 mg, 89.71 µmol, 92.42% purity, 1 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 150.56 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1- ((2S)-1-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)amino)-1-oxopropan-2- yl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinic acid (63.4 mg, 64.2 µmol, 71.6% yield, 98.6% purity) as a white solid. MS (ESI) m/z: 974.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 13.22 - 12.35 (m, 1H), 10.88 (s, 1H), 9.92 (s, 1H), 8.14 (s, 1H), 8.08 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 8.6 Hz, 2H), 7.51 - 7.41 (m, 3H), 7.41 - 7.30 (m, 2H), 7.19 (dd, J = 1.6, 7.2 Hz, 1H), 7.14 - 7.03 (m, 1H), 6.96 (d, J = 8.8 Hz, 1H), 6.87 (d, J = 8.4 Hz, 1H), 6.62 (d, J = 8.4 Hz, 1H), 4.98 (s, 2H), 4.33 - 4.29 (m, 1H), 3.97 - 3.88 (m, 7H), 3.02 (t, J = 5.6 Hz, 2H), 2.87 - 2.79 (m, 2H), 2.67 - 2.59 (m, 2H), 2.38 - 2.27 (m, 2H), 2.21 - 2.11 (m, 2H), 1.89 (s, 3H), 1.81 - 1.65 (m, 4H), 1.43 - 1.34 (m, 2H), 1.33 - 1.12 (m, 7H). Example 90. Preparation of Compound 168a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pip erazin-1-yl)-2,2- difluoropropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of 3-[7-[4-[3-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]-2,2-difluoro-propyl]piperazin-1-yl]-1-me thyl-indazol-3- yl]piperidine-2,6-dione A mixture of 3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2,2-difluo ropropyl trifluoromethanesulfonate (270 mg, 545.13 µmol, 1 equiv.), 3-(1-methyl-7-piperazin-1-yl- indazol-3-yl)piperidine-2,6-dione (151.70 mg, 463.36 µmol, 0.85 equiv.), and K2CO3 (188.35 mg, 1.36 mmol, 2.5 equiv.) in CH 3 CN (1.5 mL) was stirred at 50 °C for 10 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 20% ethyl acetate / petroleum ether) to give 3-[7-[4-[3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-2,2-difl uoro- propyl]piperazin-1-yl]-1-methyl-indazol-3-yl]piperidine-2,6- dione (150 mg, 223.0 µmol, 40.9% yield) as a colorless oil. MS (ESI) m/z: 673.6 [M+H] + . Step B. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo -3-piperidyl)-1-methyl- indazol-7-yl]piperazin-1-yl]-2,2-difluoro-propyl]cyclohexoxy ]-2-methyl- phenyl]pyridine-2-carboxylate A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine- 2-carboxylate (191.25 mg, 312.22 µmol, 1.4 equiv.), 3-[7-[4-[3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-2,2-difl uoro- propyl]piperazin-1-yl]-1-methyl-indazol-3-yl]piperidine-2,6- dione (150 mg, 223.01 µmol, 1 equiv.), [2-(2-aminophenyl)phenyl]palladium(1 + );bis(1-adamantyl)-butyl- phosphane;methanesulfonate (32.48 mg, 44.60 µmol, 0.2 equiv.), and KF (1.5 M, 223.01 uL, 1.5 eq) in dioxane (0.4 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 100 °C for 1 hour under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 50% ethyl acetate / petroleum ether) to give tert-butyl 6-[8- (1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin- 2-yl]-3-[3-[4-[3-[4-[3-(2,6- dioxo-3-piperidyl)-1-methyl-indazol-7-yl]piperazin-1-yl]-2,2 -difluoro-propyl]cyclohexoxy]- 2-methyl-phenyl]pyridine-2-carboxylate (200 mg, 185.5 umol, 83.2% yield) as a pale yellow solid. MS (ESI) m/z: 1079.7 [M+H] + . Step C. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)-2,2-difluoropropyl)cy clohexyl)oxy)-2- methylphenyl)picolinic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazo l-7-yl]piperazin-1-yl]-2,2- difluoro-propyl]cyclohexoxy]-2-methyl-phenyl]pyridine-2-carb oxylate (200 mg, 185.48 µmol, 1 equiv.) in TFA (0.5 mL) and DCM (0.5 mL) was stirred at 25 °C for 10 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-7- yl)piperazin-1-yl)-2,2-difluoropropyl)cyclohexyl)oxy)-2-meth ylphenyl)picolinic acid (97.3 mg, 91.9 µmol, 49.6% yield, 96.6% purity) as a off-white solid. MS (ESI) m/z: 1023.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.97 - 12.44 (m, 2H), 10.88 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 6.8 Hz, 1H), 7.49 - 7.33 (m, 6H), 7.13 - 6.91 (m, 5H), 6.62 (d, J = 7.6 Hz, 1H), 5.05 - 4.94 (m, 2H), 4.36 - 4.30 (m, 1H), 4.24 (s, 3H), 3.92 (t, J = 5.2 Hz, 2H), 3.02 (t, J = 5.6 Hz, 2H), 2.95 - 2.77 (m, 4H), 2.95 - 2.58 (m, 5H), 2.39 - 2.26 (m, 2H), 2.21 - 1.84 (m, 12H), 1.75 - 1.67 (m, 1H), 1.46 - 1.35 (m, 2H), 1.27 - 1.18 (m, 2H). F NMR (400 MHz, DMSO-d6) δ = -73.67, -94.95. Example 91. Preparation of Compound 169a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[(1S)-3-[1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-o xo-ethyl]-4-piperidyl]-1- methyl-propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[(1S)-3-[1-[2-[[3-(2,6 -dioxo-3-piperidyl)-1-methyl- indazol-7-yl]amino]-2-oxo-ethyl]-4-piperidyl]-1-methyl-propo xy]-2-methyl- phenyl]pyridine-2-carboxylate To a solution of 2-[4-[(3S)-3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]butyl]-1- piperidyl]acetic acid (100 mg, 126.59 µmol, 1 equiv.) and 3-(7-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (39.23 mg, 151.90 µmol, 1.2 equiv.) in DMF (0.5 mL) was added HATU (72.20 mg, 189.9 µmol, 1.5 equiv.) and DIEA (81.80 mg, 632.93 µmol, 110.24 µL, 5 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was quenched by addition water (10 mL), filtered, and concentrated under reduced pressure to give tert-butyl 6- [8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinol in-2-yl]-3-[3-[(1S)-3-[1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-o xo-ethyl]-4-piperidyl]-1- methyl-propoxy]-2-methyl-phenyl]pyridine-2-carboxylate (120 mg, 116.5 µmol, 92.0% yield) as a yellow solid. MS (ESI) m/z: 1030.9 [M+H] + Step B. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[(1S)-3-[1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-7- yl]amino]-2-oxo-ethyl]-4-piperidyl]-1-methyl-propoxy]-2-meth yl-phenyl]pyridine-2- carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[(1S)-3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methy l-indazol-7-yl]amino]-2-oxo- ethyl]-4-piperidyl]-1-methyl-propoxy]-2-methyl-phenyl]pyridi ne-2-carboxylate (120 mg, 116.48 µmol, 1 equiv.) in DCM (1 mL) was added TFA (13.28 mg, 116.48 µmol, 8.62 µL, 1 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8- (1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin- 2-yl]-3-[3-[(1S)-3-[1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-oxo-e thyl]-4-piperidyl]-1-methyl- propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid (20.7 mg, 19.5 µmol, 16.8% yield, 91.9% purity) as a white solid. MS (ESI) m/z: 974.5 [M+H] + 1 H NMR (400 MHz, DMSO-d6,) δ = 10.91 (s, 1H), 9.86 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.56 (d, J = 8.4 Hz, 1H), 7.50 - 7.42 (m, 3H), 7.40 - 7.32 (m, 2H), 7.30 (d, J = 7.2 Hz, 1H), 7.11 - 7.03 (m, 2H), 6.95 (d, J = 8.8 Hz, 1H), 6.88 (d, J = 8.4 Hz, 1H), 6.61 (d, J = 7.2 Hz, 1H), 4.97 (s, 2H), 4.49 - 4.33 (m, 2H), 4.10 (s, 3H), 3.91 (t, J = 5.6 Hz, 2H), 3.16 (br s, 2H), 3.02 (t, J = 5.6 Hz, 2H), 2.93 (d, J = 10.4 Hz, 2H), 2.69 - 2.59 (m, 2H), 2.39 - 2.31 (m, 1H), 2.23 - 2.11 (m, 3H), 1.88 (s, 3H), 1.74 - 1.56 (m, 4H), 1.38 - 1.18 (m, 8H). Example 92. Preparation of Compound 169b 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[(1R)-3-[1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-o xo-ethyl]-4-piperidyl]-1- methyl-propoxy]-2-methyl-phenyl] pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((2R)-4-(1-(2-((3-(2,6-di oxopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-2-oxoethyl)piperidin-4-yl)butan-2-yl) oxy)-2- methylphenyl)picolinate To a solution of 2-[4-[(3R)-3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]butyl]-1- piperidyl]acetic acid (80 mg, 101.27 µmol, 1 equiv.) and 3-(7-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (28.77 mg, 111.40 µmol, 1.1 equiv.) in DMF (1 mL) was added HATU (57.76 mg, 151.91 µmol, 1.5 equiv.) and DIEA (39.26 mg, 303.81 µmol, 52.92 µL, 3 equiv.). The mixture was stirred at 25 °C for 2 hours. The mixture was quenched by water (2 mL) at 25 °C, filtered, and concentrated under reduced pressure to give compound tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((2R)-4-(1-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino)-2- oxoethyl)piperidin-4-yl)butan- 2-yl)oxy)-2-methylphenyl)picolinate (100 mg, 97.1 µmol, 95.9% yield) as a white solid. MS (ESI) m/z: 1030.8[M+H] + Step B. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[(1R)-3-[1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-7- yl]amino]-2-oxo-ethyl]-4-piperidyl]-1-methyl-propoxy]-2-meth yl-phenyl] pyridine-2- carboxylic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((2R)-4-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-meth yl-1H-indazol-7-yl)amino)-2- oxoethyl)piperidin-4-yl)butan-2-yl)oxy)-2-methylphenyl)picol inate (100 mg, 97.06 µmol, 1 equiv.) in DCM (1 mL) and TFA (1 mL) was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep- HPLC to give compound 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[(1R)-3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methy l-indazol-7-yl]amino]-2-oxo- ethyl]-4-piperidyl]-1-methyl-propoxy]-2-methyl-phenyl] pyridine-2-carboxylic acid (31.9 mg, 32.3 µmol, 33.3% yield, 98.8% purity) as an off-white solid. MS (ESI) m/z: 974.7 [M+H] + 1 H NMR (400 MHz, DMSO-d6,) δ = 12.95 - 12.78 (m, 1H), 12.73 - 12.38 (m, 1H), 10.91 (s, 1H), 10.04 - 9.46 (m, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 6.8 Hz, 2H), 7.50 - 7.42 (m, 3H), 7.40 - 7.32 (m, 2H), 7.29 - 7.18 (m, 1H), 7.08 (t, J = 8.0 Hz, 2H), 6.96 (d, J = 8.4 Hz, 1H), 6.89 (d, J = 8.0 Hz, 1H), 6.61 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.46 - 4.34 (m, 2H), 4.08 (s, 3H), 3.91 (t, J = 5.6 Hz, 2H), 3.65 - 3.38 (m, 2H), 3.02 (t, J = 5.6 Hz, 4H), 2.71 - 2.61 (m, 2H), 2.42 - 2.30 (m, 2H), 2.23 - 2.09 (m, 2H), 1.88 (s, 3H), 1.73 - 1.56 (m, 3H), 1.53 - 1.19 (m, 9H). Example 93. Preparation of Compound 170 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-oxo-e thyl]-4-piperidyl]-2,2-difluoro- propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-diox o-3-piperidyl)-1-methyl- indazol-7-yl]amino]-2-oxo-ethyl]-4-piperidyl]-2,2-difluoro-p ropoxy]-2-methyl- phenyl]pyridine-2-carboxylate To a solution of 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]-2,2-difluoro-propyl]- 1-piperidyl]acetic acid (100 mg, 123.16 µmol, 1 equiv.) and 3-(7-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (38.17 mg, 147.80 µmol, 1.2 equiv.) in DMF (1 mL) was added HATU (56.20 mg, 147.80 µmol, 1.2 equiv.) and DIEA (47.75 mg, 369.49 µmol, 64.36 µL, 3 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was added into water (5 mL) and filtered to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)- 1-methyl-indazol-7-yl]amino]-2- oxo-ethyl]-4-piperidyl]-2,2-difluoro-propoxy]-2-methyl-pheny l]pyridine-2-carboxylate (110 mg, crude) as a yellow solid. MS (ESI) m/z: 1052.5 [M+H] + Step B. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-7- yl]amino]-2-oxo-ethyl]-4-piperidyl]-2,2-difluoro-propoxy]-2- methyl-phenyl]pyridine-2- carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-ind azol-7-yl]amino]-2-oxo-ethyl]-4- piperidyl]-2,2-difluoro-propoxy]-2-methyl-phenyl]pyridine-2- carboxylate (110 mg, 104.54 µmol, 1 equiv.) in DCM (1.1 mL) was added TFA (1.69 g, 14.86 mmol, 1.1 mL, 142.11 equiv.). The mixture was stirred at 25 °C for 12 hours. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[3-[1-[2-[[3-(2,6-dioxo- 3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-oxo-ethyl]-4-pip eridyl]-2,2-difluoro-propoxy]- 2-methyl-phenyl]pyridine-2-carboxylic acid (35.5 mg, 35.4 µmol, 33.9% yield, 99.1% purity, FA) as an off-white solid. MS (ESI) m/z: 996.6 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.27 - 11.99 (m, 1H), 10.90 (s, 1H), 9.85 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.56 (d, J = 8.0 Hz, 1H), 7.50 - 7.42 (m, 3H), 7.40 - 7.32 (m, 2H), 7.29 (d, J = 7.2 Hz, 1H), 7.16 - 7.10 (m, 1H), 7.07 (t, J = 7.6 Hz, 1H), 7.00 - 6.92 (m, 2H), 6.71 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.41 - 4.34 (m, 1H), 4.29 (t, J = 12 Hz, 2H), 4.10 (s, 3H), 3.92 (t, J = 6.0 Hz, 2H), 3.15 (s, 2H), 3.02 ( t, J = 5.6 Hz, 2H), 2.95 - 2.88 (m, 2H), 2.71 - 2.57 (m, 2H), 2.39 - 2.32 (m, 1H), 2.26 - 2.12 (m, 3H), 2.09 - 1.97 (m, 2H), 1.92 (s, 3H), 1.80 - 1.65 (m, 3H), 1.50 - 1.37 (m, 2H) Example 94. Preparation of Compound 173b 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[(3R)-3-[1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-o xo-ethyl]-4-piperidyl]butoxy]- 2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[(3R)-3-[1-[2-[[3-(2,6 -dioxo-3-piperidyl)-1-methyl- indazol-7-yl]amino]-2-oxo-ethyl]-4-piperidyl]butoxy]-2-methy l-phenyl]pyridine-2- carboxylate To a solution of 2-[4-[(1R)-3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]-1-methyl-propyl]-1- piperidyl]acetic acid (100 mg, 126.59 µmol, 1 equiv.) and HATU (72.20 mg, 189.89 µmol, 1.5 equiv.) in DMF (1 mL) was added DIEA (49.08 mg, 379.77 µmol, 66.15 µL, 3 equiv.) and 3- (7-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (39.23 mg, 151.91 µmol, 1.2 equiv.). The mixture was stirred at 25 °C for 1 hour. The mixture was added into H 2 O (5 mL) and filtered to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[(3R)-3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methy l-indazol-7-yl]amino]-2-oxo- ethyl]-4-piperidyl]butoxy]-2-methyl-phenyl]pyridine-2-carbox ylate (105 mg, crude) as a yellow solid. MS (ESI) m/z: 1030.8 [M+H] + Step B. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[(3R)-3-[1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-7- yl]amino]-2-oxo-ethyl]-4-piperidyl]butoxy]-2-methyl-phenyl]p yridine-2-carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[(3R)-3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methy l-indazol-7-yl]amino]-2-oxo- ethyl]-4-piperidyl]butoxy]-2-methyl-phenyl]pyridine-2-carbox ylate (105 mg, 101.92 µmol, 1 equiv.) in DCM (0.5 mL) was added TFA (770.00 mg, 6.75 mmol, 0.5 mL, 66.26 equiv.). The mixture was stirred at 25 °C for 12 hours. The mixture was concentrated under reduced pressure to remove DCM. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[(3R)-3-[ 1-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol-7-yl]amino]-2-oxo-ethyl]-4-piper idyl]butoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid (22.3 mg, 22.2 µmol, 21.8% yield, 96.9% purity) as a white solid. MS (ESI) m/z: 974.6 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.37 - 11.91 (m, 1H), 10.90 (s, 1H), 9.87 (s, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 8.0 Hz, 1H), 7.56 (d, J = 8.0 Hz, 1H), 7.49 - 7.40 (m, 3H), 7.40 - 7.29 (m, 3H), 7.08 (q, J = 7.6 Hz, 2H), 6.96 - 6.87 (m, 2H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.40 - 4.34 (m, 1H), 4.11 (s, 3H), 4.06 - 3.95 (m, 2H), 3.91 (t, J = 5.2 Hz, 2H), 3.15 (s, 2H), 3.04 - 2.97 (m, 4H), 2.70 - 2.60 (m, 2H), 2.41 - 2.29 (m, 2H), 2.18 - 2.13 (m, 2H), 1.89 (s, 3H), 1.66 - 1.52 (m, 4H), 1.49 - 1.38 (m, 2H), 1.32 - 1.15 (m, 2H), 0.92 (d, J = 6.8 Hz, 3H) Example 95. Preparation of Compound 175 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(4-(4-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin -1-yl)butyl)phenoxy)-2- methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(4-(4-(3-(2,6-dioxo piperidin-3-yl)-1-methyl-1H- indazol-7-yl)piperazin-1-yl)butyl)phenoxy)-2-methylphenyl)pi colinate A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(2-methyl-3-(4-(4-oxobutyl)phenoxy)phenyl)picolinate (75.00 mg, 101.50 µmol, 1 equiv.), 3-(1-methyl-7-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (39.88 mg, 121.80 µmol, 1.2 equiv.), 4A MS (10 mg, 101.50 µmol, 1.00 equiv.) and HCOOH (97.52 ug, 2.03 µmol, 0.02 equiv.) in EtOH (2.4 mL) and THF (2.4 mL) was stirred at 25 °C for 2 h. NaBH3CN (19.14 mg, 304.51 µmol, 3 equiv.) was added to the mixture, and the mixture was stirred at 25 °C for 14 h. The mixture was diluted with H 2 O (2 mL × 2) and extracted with ethyl acetate (2 mL × 3). The combined organic layers were filtered and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- (3-(4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -7-yl)piperazin-1- yl)butyl)phenoxy)-2-methylphenyl)picolinate (100 mg, crude) as a yellow oil. It was used for next step without other purification. MS (ESI) m/z:1050.6 [M+H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(4-(4-(3-(2,6-dioxopipe ridin-3-yl)-1-methyl-1H- indazol-7-yl)piperazin-1-yl)butyl)phenoxy)-2-methylphenyl)pi colinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-7-yl)piperazin-1- yl)butyl)phenoxy)-2-methylphenyl)picolinate (80 mg, 75.73 µmol, 1 equiv.), in TFA (0.8 mL) and DCM (0.8 mL) was stirred at 40 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin-1-yl )butyl)phenoxy)-2- methylphenyl)picolinic acid (28.2 mg, 27.9 µmol, 36.9% yield, 98.3% purity) as a white solid. MS (ESI) m/z: 994.8 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.87 (s, 1H), 8.18 (s, 1H), 8.02 (d, J = 7.2 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.61 (d, J = 7.2 Hz,1H), 7.50 - 7.43 (m, 3H), 7.39 - 7.32 (m, 3H), 7.19 - 7.13 (m, 3H), 7.02 - 6.94 (m, 3H), 6.89 (d, J = 7.2 Hz, 1H), 6.81 (d, J = 8.0 Hz, 3H), 4.98 (s, 2H), 4.33 (dd, J = 5.2, 9.2 Hz, 1H), 4.22 (s, 3H), 3.91 (t, J = 5.6 Hz, 2H), 3.05 - 3.00 (m, 4H), 2.91 - 2.87 (m, 2H), 2.69 - 2.58 (m, 6H), 2.39 (t, J = 7.2 Hz, 2H), 2.35 - 2.28 (m, 2H), 2.23 - 2.10 (m, 2H), 1.91 (s, 3H), 1.64 - 1.56 (m, 2H), 1.50 - 1.48 (m, 2H) Example 96. Preparation of Compound 191 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(4-(4-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin -1-yl)-4-oxobutyl)phenoxy)-2- methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(4-(4-(3-(2,6-dioxopipe ridin-3-yl)-1-methyl-1H- indazol-7-yl)piperazin-1-yl)-4-oxobutyl)phenoxy)-2-methylphe nyl)picolinate A mixture of 4-(4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroiso quinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)phe nyl)butanoic acid (100 mg, 132.47 µmol, 1 equiv.), HATU (75.55 mg, 198.70 µmol, 1.5 equiv.), and DIEA (51.36 mg, 397.41 µmol, 69.22 µL, 3 equiv.) in DMAC (2 mL) was stirred at 25 °C for 0.25 h. 3-(1- Methyl-7-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dione (52.04 mg, 158.96 µmol, 1.2 equiv.) was added to the mixture and stirred at 25 °C for 16 h. The mixture was diluted with H2O (2 mL × 2) and extracted with ethyl acetate (2 mL × 3). The combined organic layers were filtered and concentrated under reduced pressure to give the crude compound tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin-1-yl )-4-oxobutyl)phenoxy)-2- methylphenyl)picolinate (100 mg, crude) as a yellow solid, it was used for next step without other purification. MS (ESI) m/z: 1064.8 [M+H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(4-(4-(3-(2,6-dioxopipe ridin-3-yl)-1-methyl-1H- indazol-7-yl)piperazin-1-yl)-4-oxobutyl)phenoxy)-2-methylphe nyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-7-yl)piperazin-1-yl)-4- oxobutyl)phenoxy)-2-methylphenyl)picolinate (100 mg, 93.96 µmol, 1 equiv.) in DCM (0.8 mL) and TFA (0.8 mL) was stirred at 40 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep–HPLC to give 6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2( 1H)-yl)-3-(3-(4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin-1-yl )-4-oxobutyl)phenoxy)-2- methylphenyl)picolinic acid (33.1 mg, 30.5 µmol, 32.5% yield, 93.1% purity) as a white solid. MS (ESI) m/z: 1008.8 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.20 - 12.33 (m, 1H), 10.88 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.54 - 7.36 (m, 6H), 7.20 - 7.14 (m, 3H), 7.05 - 6.97 (m, 3H), 6.90 - 6.81 (m, 4H), 4.99 (s, 2H), 4.56 - 4.42 (m, 1H), 4.34 (dd, J = 4.8, 9.6 Hz, 1H), 4.26 (s, 3H), 4.00 - 3.81 (m, 4H), 3.05 - 3.01 (m, 2H), 2.69 - 2.55 (m, 6H), 2.43 - 2.24 (m, 6H), 2.21 - 2.12 (m, 1H), 1.90 (s, 3H), 1.85 - 1.79 (m, 2H) Example 97. Preparation of Compound 196 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino)-2- oxoethyl)piperidin-4-yl)-3,3- difluoropropoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-7-yl)amino)-2-oxoethyl)piperidin-4-yl)-3,3-difluorop ropoxy)-2- methylphenyl)picolinate To a solution of 2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)-1,1- difluoropropyl)piperidin-1-yl)acetic acid (50.0 mg, 61.6 µmol, 1.0 equiv.) in DMF (0.50 mL) were added 3-(7-amino-1-methyl-1H-indazol-3-yl)piperidine-2,6-dione (17.5 mg, 67.7 µmol, 1.1 equiv.), HATU (28.1 mg, 73.9 µmol, 1.2 equiv.), and DIEA (23.9 mg, 185 µmol, 32.2 µL, 3.0 equiv.). The mixture was stirred at 20 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino)-2- oxoethyl)piperidin-4-yl)-3,3- difluoropropoxy)-2-methylphenyl)picolinate (30.0 mg, 28.5 µmol, 46.3% yield) as a yellow solid. MS (ESI) m/z: 1052.5 [M+H] + Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-7-yl)amino)-2-oxoethyl)piperidin-4-yl)-3,3-difluorop ropoxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-meth yl-1H-indazol-7-yl)amino)-2- oxoethyl)piperidin-4-yl)-3,3-difluoropropoxy)-2-methylphenyl )picolinate (30.0 mg, 28.5 µmol, 1.0 equiv.) in CH2Cl2 (1.00 mL) was added TFA (924 mg, 8.10 mmol, 600 µL, 284 equiv.). The mixture was stirred at 20 °C for 6 hours. After completion, the reaction mixture was concentrated under reduced pressure and the residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino)-2- oxoethyl)piperidin-4-yl)-3,3- difluoropropoxy)-2-methylphenyl)picolinic acid (10.64 mg, 10.6 µmol, 37.1% yield, 99.0% purity) as a yellow solid. MS (ESI) m/z: 996.7 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.01 - 12.74 (m, 1H), 12.71 - 12.41 (m, 1H), 10.90 (s, 1H), 9.86 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 7.6 Hz, 1H), 7.68 - 7.52 (m, 2H), 7.51 - 7.41 (m, 3H), 7.41 - 7.31 (m, 2H), 7.27 (d, J = 7.2 Hz, 1H), 7.16 - 7.03 (m, 2H), 7.01 - 6.89 (m, 2H), 6.66 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.43 - 4.32 (m, 1H), 4.16 (t, J = 5.6 Hz, 2H), 4.09 (s, 3H), 3.95 - 3.89 (m, 2H), 3.17 (s, 2H), 3.03 (t, J = 5.6 Hz, 4H), 2.66 - 2.54 (m, 2H), 2.46 - 2.36 (m, 3H), 2.23 - 2.12 (m, 3H), 2.04 - 1.94 (m, 1H), 1.90 (s, 3H), 1.86 - 1.75 (m, 2H), 1.70 - 1.56 (m, 2H). Example 98. Preparation of Compound 199 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4-[3-[1-[2-[4-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]piperazin-1-yl ]ethyl]-4-piperidyl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[4-[3-[1-[2-[4-[3-(2,6-dioxo-3-piperi dyl)-1-methyl-indazol-6- yl]piperazin-1-yl]ethyl]-4-piperidyl]propoxy]-2-methyl-pheny l]pyridine-2-carboxylic acid To a solution of 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (83.72 mg, 255.74 µmol, 0.9 equiv.) and NaBH(OAc) 3 (180.67 mg, 852.46 µmol, 3.0 equiv.) in IPA (2 mL) and DCM (2 mL) was added 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[2-methyl-4-[3-[1-(2-oxoethyl)-4-piperid yl]propoxy]phenyl]pyridine-2- carboxylic acid (200 mg, 284.15 µmol, 1.0 equiv.) at 0 °C slowly. The mixture was stirred at 0 °C for 1 hour, and then the mixture was warmed to 25 °C and stirred at 25 °C for 12 hours. The mixture was concentrated under reduced pressure to remove DCM and IPA. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[4-[3-[1-[2-[4-[3-(2,6-dioxo-3-piperidyl )-1-methyl-indazol-6- yl]piperazin-1-yl]ethyl]-4-piperidyl]propoxy]-2-methyl-pheny l]pyridine-2-carboxylic acid (21.17 mg, 20.8 µmol, 7.3% yield, 99.8% purity) as a yellow solid. MS (ESI) m/z: 1016.0 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 10.84 (s, 1H), 8.02 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.52 - 7.28 (m, 6H), 6.98 - 6.80 (m, 4H), 6.79 - 6.75 (m, 1H), 6.72 - 6.65 (m, 1H), 4.95 (s, 2H), 4.27 - 4.23 (m, 1H), 3.94 - 3.89 (m, 4H), 3.88 (s, 3H), 3.23 - 3.17 (m, 6H), 3.03 - 3.97 (m, 4H), 2.62 - 2.55 (m, 7H), 2.35 - 2.23 (m, 2H), 2.19 - 2.08 (m, 3H), 2.04 (s, 3H), 1.74 - 1.66 (m, 2H), 1.66 - 1.59 (m, 2H), 1.33 - 1.23 (m, 3H), 1.18 - 1.06 (m, 2H) Example 99. Preparation of Compound 202b 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[(3R)-1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-o xo-ethyl]-3-piperidyl]butoxy]- 2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[(3R)-1-[2-[[3-(2,6 -dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-3-piperidyl]butoxy]-2-methy l-phenyl]pyridine-2- carboxylate A mixture of 2-[(3R)-3-[4-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]butyl]-1- piperidyl]acetic acid (91 mg, 115.19 µmol, 1 equiv.), 3-(6-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (35.70 mg, 138.23 µmol, 1.2 equiv.), HATU (52.56 mg, 138.23 µmol, 1.2 equiv.), and DIEA (44.66 mg, 345.58 µmol, 60.19 µL, 3 equiv.) in DMF (3 mL) was stirred at 25 °C for 12 hours. The reaction mixture was quenched by addition of water (3 mL), and then extracted with ethyl acetate (3 mL × 3). The organic layers were combined and concentrated under reduced pressure to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[(3R)-1-[2-[[3-(2,6 -dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-3-piperidyl]butoxy]-2-methy l-phenyl]pyridine-2- carboxylate (100 mg, crude) as a purple oil. MS (ESI) m/z: 1030.6 [M+H] + . Step B. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[4-[(3R)-1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-3-piperidyl]butoxy]-2-methyl-phenyl]p yridine-2-carboxylic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[4-[(3R)-1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo- ethyl]-3-piperidyl]butoxy]-2-methyl-phenyl]pyridine-2-carbox ylate (100 mg, 97.06 µmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL) was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep- HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3- [4-[(3R)-1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6 -yl]amino]-2-oxo-ethyl]-3- piperidyl]butoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid (24.2 mg, 24.6 µmol, 25.3% yield, 98.8% purity) as a yellow solid. MS (ESI) m/z: 974.9 [M+H] + . 1 H NMR (400 MHz, DMSO-d6,) δ = 10.87 (s, 1H), 9.84 (s, 1H), 8.15 (s, 1H), 8.05 - 7.99 (m, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.64 - 7.59 (m, 2H), 7.48 - 7.32 (m, 5H), 7.10 - 7.30 (m, 1H), 7.10 - 7.03 (m, 1H), 6.94 (d, J = 8.8 Hz, 1H), 6.83 (d, J = 8.0 Hz, 1H), 6.61 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.31 (dd, J = 5.2, 9.6 Hz, 1H), 3.94 - 3.88 (m, 7H), 3.12 (d, J = 11.2 Hz, 2H), 3.02 (t, J = 5.6 Hz, 2H), 2.85 - 2.85 (m, 1H), 2.85 - 2.77 (m, 2H), 2.67 - 2.58 (m, 2H), 2.37 - 2.27 (m, 1H), 2.19 - 2.10 (m, 2H), 1.87 (s, 4H), 1.77 - 1.40 (m, 9H), 1.30 - 1.23 (m, 2H). Example 100. Preparation of Compound 210a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((3S)-1-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino )-2-oxoethyl)piperidin-3- yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3S)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-2-oxoethyl)piperidin-3-yl)propoxy)-2- methylphenyl)picolinate To a solution of (S)-2-(3-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)piperidin-1- yl)acetic acid (150 mg, 193.31 μmol, 1 equiv.) in DMF (1.5 mL) was added 3-(7-amino-1- methyl-1H-indazol-3-yl)piperidine-2,6-dione (49.93 mg, 193.31 μmol, 1 equiv.), HATU (80.85 mg, 212.64 μmol, 1.1 equiv.) and DIPEA (74.95 mg, 579.93 μmol, 101.01 μL, 3 equiv.) at 25 °C. The reaction mixture was stirred at 25 °C for 2 hours. The mixture was triturated with water (5 mL) and filtered to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3S)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl-1H- indazol-7-yl)amino)-2-oxoethyl)piperidin-3-yl)propoxy)-2-met hylphenyl)picolinate (132 mg, 105.2 μmol, 54.4% yield, 81% purity) as a yellow solid. MS (ESI) m/z: 1016.7 [M+H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3S)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-2-oxoethyl)piperidin-3-yl)propoxy)-2- methylphenyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-((3S)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-7-yl)amino)-2- oxoethyl)piperidin-3-yl)propoxy)-2-methylphenyl)picolinate (132 mg, 105.86 μmol, 81.5% purity, 1 equiv.) in DCM (1 mL) and TFA (1 mL) was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-((3S)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-7-yl)amino)-2- oxoethyl)piperidin-3-yl)propoxy)-2-methylphenyl)picolinic acid (54.61 mg, 55.0 μmol, 51.9% yield, 96.7% purity) as a yellow solid. MS (ESI) m/z: 960.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.01 - 12.36 (m, 2H), 10.92 (s, 1H), 10.55 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.73 - 7.57 (m, 2H), 7.52 - 7.41 (m, 3H), 7.40 - 7.32 (m, 2H), 7.21 (d, J = 7.2 Hz, 1H), 7.17 - 7.07 (m, 2H), 6.97 (d, J = 8.8 Hz, 1H), 6.88 (d, J = 8.4 Hz, 1H), 6.64 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.44 - 4.36 (m, 1H), 4.27 (s, 2H), 4.07 (s, 3H), 3.99 - 3.90 (m, 4H), 3.76 (s, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.89 - 2.77 (m, 1H), 2.75 - 2.55 (m, 4H), 2.43 - 2.31 (m, 1H), 2.22 - 2.11 (m, 1H), 2.01 - 1.94 (m, 1H), 1.90 (s, 3H), 1.89 - 1.83 (m, 2H), 1.82 - 1.72 (m, 3H), 1.48 - 1.36 (m, 2H) Example 101. Preparation of Compound 212a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1-yl)-2,2- difluoropropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-2,2- difluoro-propan-1-ol A mixture of ethyl 3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2,2- difluoropropanoate (200 mg, 493.49 µmol, 1 equiv.), LiAlH 4 (15.92 mg, 419.47 µmol, 0.85 equiv.)in THF (2 mL) was stirred at 0 °C for 2 hours. The reaction mixture was quenched by addition Na 2 SO 4 •10H 2 O (100 mg) under 0 °C and N 2 atmosphere, and then filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 10% ethyl acetate / petroleum ether) to give 3-((1r,4r)-4-(3- bromo-2-methylphenoxy)cyclohexyl)-2,2-difluoropropan-1-ol (130 mg, 357.9 µmol, 72.5% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.17 - 7.11 (m, 1H), 7.09 - 7.00 (m, 2H), 5.46 (s, 1H), 4.28 - 4.17 (m, 1H), 3.55 (t, J = 13.6 Hz, 2H), 2.21 (s, 3H), 2.06 - 1.97 (m, 2H), 1.85 - 1.57 (m, 5H), 1.44 - 1.34 (m, 2H), 1.21 - 1.11 (m, 2H). F NMR (400 MHz, DMSO-d6) δ = -103.576 Step B. Procedure for preparation of 3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2,2-difluoropropyl trifluoromethanesulfonate To a solution of 3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2,2-difluo ropropan-1-ol (200 mg, 550.61 µmol, 1 equiv.) and pyridine (65.33 mg, 825.91 µmol, 66.66 µL, 1.5 equiv.) in CH3CN (1.5 mL) was added trifluoromethylsulfonyl trifluoromethanesulfonate (170.88 mg, 605.67 µmol, 99.93 µL, 1.1 equiv.). The mixture was stirred at 0 °C for 1 hour. The reaction mixture was diluted with H 2 O (2 mL) and extracted with DCM (10 mL). The combined organic layers were washed with 1N citric acid (2 mL) and 1N NaHCO 3 (2 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was used for next step without other purification. The compound 3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2,2-difluoropropyl trifluoromethanesulfonate (270 mg, 545.13 µmol, 99.00% yield) was obtained as a colorless oil. Step C. Procedure for preparation of 3-(6-(4-(3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2,2-difluoropropyl)piperazin-1-yl) -1-methyl-1H-indazol-3- yl)piperidine-2,6-dione A mixture of 3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2,2-difluo ropropyl trifluoromethanesulfonate (270 mg, 545.13 µmol, 1 equiv.), 3-(1-methyl-6-(piperazin-1-yl)- 1H-indazol-3-yl)piperidine-2,6-dione (151.70 mg, 463.36 µmol, 0.85 equiv.), K2CO3 (188.35 mg, 1.36 mmol, 2.5 equiv.) in CH3CN (1.5 mL) was stirred at 50 °C for 10 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~20% ethyl acetate / petroleum ether) to give 3-(6-(4-(3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2 ,2- difluoropropyl)piperazin-1-yl)-1-methyl-1H-indazol-3-yl)pipe ridine-2,6-dione (120 mg, 178.4 µmol, 32.7% yield) as a white solid. MS (ESI) m/z: 673.5 [M+H] + . Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-2,2-difluoropropyl)cy clohexyl)oxy)-2- methylphenyl)picolinate A mixture of 3-(6-(4-(3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2 ,2- difluoropropyl)piperazin-1-yl)-1-methyl-1H-indazol-3-yl)pipe ridine-2,6-dione (153.00 mg, 249.78 µmol, 1.4 equiv.), 3-[6-[4-[3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-2,2- difluoro-propyl]piperazin-1-yl]-1-methyl-indazol-3-yl]piperi dine-2,6-dione (120 mg, 178.41 µmol, 1 equiv.), [2-(2-aminophenyl)phenyl]palladium(1 + );bis(1-adamantyl)-butyl- phosphane;methanesulfonate (25.99 mg, 35.68 µmol, 0.2 equiv.), and KF (1.5 M, 178.41 µL, 1.5 equiv.) in dioxane (0.4 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 100 °C for 1 hour under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 50% ethyl acetate / petroleum ether) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1-yl)-2,2- difluoropropyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (100 mg, 92.7 µmol, 51.9% yield) as a pale yellow solid. MS (ESI) m/z: 1080.4 [M+H] + . Step E. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-2,2-difluoropropyl)cy clohexyl)oxy)-2- methylphenyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1r,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)piperazin-1- yl)-2,2-difluoropropyl)cyclohexyl)oxy)-2-methylphenyl)picoli nate (100 mg, 92.74 µmol, 1 equiv.) in TFA (0.5 mL) and DCM (0.5 mL) was stirred at 25 °C for 10 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1r,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)piperazin-1- yl)-2,2-difluoropropyl)cyclohexyl)oxy)-2-methylphenyl)picoli nic acid (37.0 mg, 35.5 µmol, 38.3% yield, 98.1% purity) as a white solid. MS (ESI) m/z: 1022.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.54 - 11.96 (m, 2H), 10.85 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.65 - 7.59 (m, 1H), 7.53 - 7.43 (m, 4H), 7.41 - 7.32 (m, 2H), 7.10 - 7.05 (m, 1H), 6.98 - 6.86 (m, 4H), 6.61 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.28 - 4.19 (m, 2H), 3.93 - 3.88 (m, 5H), 3.22 (s, 2H), 3.05 - 2.99 (m, 2H), 2.80 - 2.59 (m, 8H), 2.34 - 2.05 (m, 6H), 1.98 - 1.86 (m, 7H), 1.73 - 1.66 (m, 1H), 1.44 - 1.37 (m, 2H), 1.24 - 1.18 (m, 2H). F NMR (400 MHz, DMSO-d6) δ = -73.56, -94.83. Example 102. Preparation of Compound 216a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-((2S)-1- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino )-1-oxopropan-2-yl)piperidin- 4-yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-((2S)-1-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-1-oxopropan-2-yl)piperidin-4-yl)propo xy)-2- methylphenyl)picolinate To a solution of (S)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)piperidin-1- yl)propanoic acid (100 mg, 126.59 µmol, 1 equiv.), and 3-(7-amino-1-methyl-1H-indazol-3- yl)piperidine-2,6-dione (39.23 mg, 151.91 µmol, 1.2 equiv.) in pyridine (1 mL) was added EDCI (36.40 mg, 189.89 µmol, 1.5 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was quenched by addition of H2O (2mL). The reaction mixture was filtered, and the solid was diluted in DCM (10 mL). The mixture was dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-((2S)-1-((3-(2,6 -dioxopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-1-oxopropan-2-yl)piperidin-4-yl)propo xy)-2- methylphenyl)picolinate (100 mg, crude) as a yellow solid. MS (ESI) m/z: 1030.8 [M+H] + Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-((2S)-1-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-1-oxopropan-2-yl)piperidin-4-yl)propo xy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-((2S)-1-((3-(2,6-dioxopiperidin-3-yl)-1 -methyl-1H-indazol-7-yl)amino)- 1-oxopropan-2-yl)piperidin-4-yl)propoxy)-2-methylphenyl)pico linate (100 mg, 87.33 µmol, 89.97% purity, 1 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 154.66 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1- ((2S)-1-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7- yl)amino)-1-oxopropan-2- yl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinic acid (51.4 mg, 52.5 µmol, 60.1% yield, 99.3% purity) as a white solid. MS (ESI) m/z: 975.0 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 13.16 - 12.54 (m, 1H), 10.90 (s, 1H), 10.05 - 9.73 (m, 1H), 8.14 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.66 - 7.54 (m, 2H), 7.50 - 7.41 (m, 3H), 7.41 - 7.32 (m, 2H), 7.25 (d, J = 7.2 Hz, 1H), 7.08 (q, J = 8.0 Hz, 2H), 6.96 (d, J = 9.2 Hz, 1H), 6.91 - 6.84 (m, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.39 - 4.35 (m, 1H), 4.10 (s, 3H), 3.97 - 3.89 (m, 4H), 3.02 (t, J = 5.6 Hz, 2H), 2.98 - 2.85 (m, 2H), 2.70 - 2.59 (m, 2H), 2.44 - 2.30 (m, 2H), 2.28 - 2.11 (m, 2H), 1.89 (s, 3H), 1.83 - 1.65 (m, 4H), 1.50 - 1.09 (m, 9H). Example 103. Preparation of Compound 216b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-((2R)-1- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino )-1-oxopropan-2-yl)piperidin- 4-yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-((2R)-1-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-1-oxopropan-2-yl)piperidin-4-yl)propo xy)-2- methylphenyl)picolinate To a solution of (R)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)piperidin-1- yl)propanoic acid (100 mg, 112.93 µmol, 89.21% purity, 1 equiv.) and 3-(7-amino-1-methyl- 1H-indazol-3-yl)piperidine-2,6-dione (35.00 mg, 135.51 µmol, 1.2 equiv.) in pyridine (1 mL) was added EDCI (32.47 mg, 169.39 µmol, 1.5 equiv.). The mixture was stirred at 25 °C for 3 hours. The reaction mixture was quenched by addition of H 2 O (2mL). The reaction mixture was filtered and the filter cake was washed with water (5 mL). The filter cake was then diluted in DCM (10 mL). The organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-((2R)-1-((3-(2,6-dio xopiperidin-3-yl)-1-methyl-1H- indazol-7-yl)amino)-1-oxopropan-2-yl)piperidin-4-yl)propoxy) -2-methylphenyl)picolinate (100 mg, crude) as a brown solid. MS (ESI) m/z: 1030.8 [M+H] + Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-((2R)-1-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-1-oxopropan-2-yl)piperidin-4-yl)propo xy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-((2R)-1-((3-(2,6-dioxopiperidin-3-yl)-1 -methyl-1H-indazol-7-yl)amino)- 1-oxopropan-2-yl)piperidin-4-yl)propoxy)-2-methylphenyl)pico linate (100 mg, 97.06 µmol, 1 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 139.14 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-(1-((2R)-1-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino)-1- oxopropan-2-yl)piperidin-4- yl)propoxy)-2-methylphenyl)picolinic acid (55.0 mg, 55.0 µmol, 56.7% yield, 97.5% purity) as a white solid. MS (ESI) m/z: 975.0 [M+H] + . 1 H NMR (400 MHz, DMSO-d6,) δ = 10.89 (s, 1H), 9.86 (s, 1H), 8.15 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.56 (d, J = 8.0 Hz, 1H), 7.51 - 7.41 (m, 3H), 7.40 - 7.31 (m, 2H), 7.25 (d, J = 7.2 Hz, 1H), 7.08 (q, J = 8.0 Hz, 2H), 6.96 (d, J = 8.8 Hz, 1H), 6.87 (d, J = 8.4 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.38 - 4.34 (m, 1H), 4.10 (s, 3H), 3.97 - 3.89 (m, 4H), 3.02 (t, J = 5.6 Hz, 2H), 2.91 - 2.84 (m, 2H), 2.68 - 2.60 (m, 2H), 2.39 - 2.30 (m, 2H), 2.24 - 2.12 (m, 2H), 1.89 (s, 3H), 1.81 - 1.68 (m, 4H), 1.41 - 1.40 (m, 2H), 1.35 - 1.17 (m, 7H). Example 104. Preparation of Compound 217 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[8-[4-[3-(2,6- dioxo-3-piperidyl)-1-methyl-indazol-7-yl]piperazin-1-yl]octo xy]-2-methyl-phenyl]pyridine- 2-carboxylic acid Step A. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[8-[4-[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol- 7-yl]piperazin-1-yl]octoxy]-2-methyl-phenyl]pyridine-2-carbo xylate A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[2-methyl-3-(8-oxooctoxy)phenyl]pyridine-2-carboxylate (100 mg, 139.10 µmol, 1 equiv.), 3-(1-methyl-7-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (54.65 mg, 166.92 µmol, 1.2 equiv.), NaBH(OAc)3 (88.44 mg, 417.30 µmol, 3 equiv.), and AcOH (8.35 mg, 139.10 µmol, 7.96 µL, 1 equiv.) in DCM (2 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 3 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was used in the next step without purification. tert-Butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[8-[4-[3-(2,6-dioxo-3-piperidyl)-1-me thyl-indazol-7-yl]piperazin-1- yl]octoxy]-2-methyl-phenyl]pyridine-2-carboxylate (100 mg, crude) was obtained as a white solid. MS (ESI) m/z: 1030.8 [M+H] + Step B. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[8-[4-[3-(2,6-dioxo-3-piperidyl)-1 -methyl-indazol-7- yl]piperazin-1-yl]octoxy]-2-methyl-phenyl]pyridine-2-carboxy lic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[8-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7 -yl]piperazin-1-yl]octoxy]-2- methyl-phenyl]pyridine-2-carboxylate (90.00 mg, 87.35 µmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[8-[4- [3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]piperazin-1 -yl]octoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid (21.5 mg, 20.4 µmol, 23.4% yield, 96.9% purity, FA) as a white solid. MS (ESI) m/z: 975.1 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.90 - 12.82 (m, 1H), 10.89 (s, 1H), 8.14 (s, 1H), 8.01 (d, J = 7.6 Hz, 1H), 7.77 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.42 - 7.39 (m, 7H), 7.11 - 7.01 (m, 3H), 6.96 (d, J = 8.8 Hz, 1H), 6.88 (d, J = 8.4 Hz, 1H), 6.63 (d, J = 7.2 Hz, 1H), 5.00 - 4.95 (m, 2H), 4.36 - 4.33 (m, 1H), 4.25 - 4.22 (m, 3H), 3.99 - 3.89 (m, 5H), 3.08 - 2.96 (m, 6H), 2.92 - 2.85 (m, 3H), 2.70 - 2.56 (m, 3H), 2.34 - 2.30 (m, 1H), 2.18 - 2.14 (m, 1H), 1.90 (s, 3H), 1.78 - 1.72 (m, 2H), 1.64 - 1.55 (m, 2H), 1.49 - 1.44 (m, 2H), 1.41 - 1.25 (m, 7H). Example 105. Preparation of Compound 224a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(((1R,3s,5S)- 8-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl) amino)-2-oxoethyl)-8- azabicyclo[3.2.1]octan-3-yl)oxy)ethoxy)-2-methylphenyl)picol inic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(((1R,3s,5S)-8-(2-((3-( 2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)amino)-2-oxoethyl)-8-azabicyclo[3.2.1 ]octan-3-yl)oxy)ethoxy)-2- methylphenyl)picolinate To a solution of 2-((1R,3s,5S)-3-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)ethoxy)-8-azabicyclo[3.2.1]octan-8-yl)acetic acid (110 mg, 136.82 µmol, 1 equiv.) in DMF (1 mL) was added HATU (62.43 mg, 164.19 µmol, 1.2 equiv.) and DIEA (53.05 mg, 410.47 µmol, 71.49 µL, 3 equiv.). Then 3-(7-amino-1-methyl-1H-indazol-3- yl)piperidine-2,6-dione (38.87 mg, 150.50 µmol, 1.1 equiv.) was added into the mixture and stirred at 25 °C for 12 hours. DCM 20 mL and water 20 mL were added and layers were separated. The aqueous was washed with DCM 20 mL (10 mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated under vacuum to give compound tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(2-(((1R,3s,5S)-8-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino )-2-oxoethyl)-8- azabicyclo[3.2.1]octan-3-yl)oxy)ethoxy)-2-methylphenyl)picol inate (100 mg, 95.7 µmol, 69.9% yield) as a yellow solid. MS (ESI) m/z: 1044.9 [M+H] + Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(((1R,3s,5S)-8-(2-((3-( 2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)amino)-2-oxoethyl)-8-azabicyclo[3.2.1 ]octan-3-yl)oxy)ethoxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(((1R,3s,5S)-8-(2-((3-(2,6-dioxopiperidin- 3-yl)-1-methyl-1H-indazol-7- yl)amino)-2-oxoethyl)-8-azabicyclo[3.2.1]octan-3-yl)oxy)etho xy)-2-methylphenyl)picolinate (100 mg, 95.76 µmol, 1 equiv.) in DCM (2 mL) was added TFA (3.08 g, 27.01 mmol, 2 mL, 282.07 equiv.) and. The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by prep- HPLC to give compound 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(((1R,3s,5S)-8-(2-((3-(2,6-dioxopiperidin- 3-yl)-1-methyl-1H-indazol-7- yl)amino)-2-oxoethyl)-8-azabicyclo[3.2.1]octan-3-yl)oxy)etho xy)-2-methylphenyl)picolinic acid (20.7 mg, 18.8 µmol, 19.7% yield, 94.0% purity, FA) as a yellow solid. MS (ESI) m/z: 988.8 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.89 (s, 1H), 9.99 (s, 1H), 8.15 (s, 1H), 8.02 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.56 (d, J = 8.0 Hz, 1H), 7.49 - 7.40 (m, 3H), 7.39 - 7.29 (m, 3H), 7.12 - 7.03 (m, 2H), 6.95 (d, J = 8.8 Hz, 1H), 6.89 (d, J = 8.0 Hz, 1H), 6.64 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.41 - 4.31 (m, 1H), 4.12 (s, 3H), 4.08 - 4.03 (m, 2H), 3.93 - 3.89 (m, 2H), 3.76 - 3.73 (m, 2H), 3.72 - 3.65 (m, 2H), 3.21 (s, 2H), 3.02 (t, J = 5.2 Hz, 2H), 2.66 (d, J = 4.0 Hz, 2H), 2.33 (d, J = 1.6 Hz, 1H), 2.23 - 2.10 (m, 2H), 1.94 - 1.84 (m, 7H), 1.66 (t, J = 10.8 Hz, 2H), 1.59 (d, J = 7.6 Hz, 2H) Example 106. Preparation of Compound 231a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[2-[(3S)-1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-o xo-ethyl]pyrrolidin-3- yl]ethoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[(3S)-1-[2-[[3-(2,6 -dioxo-3-piperidyl)-1-methyl- indazol-7-yl]amino]-2-oxo-ethyl]pyrrolidin-3-yl]ethoxy]-2-me thyl-phenyl]pyridine-2- carboxylate A mixture of 2-[(3S)-3-[2-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]ethyl]pyrrolidin-1- yl]acetic acid ((110 mg, 147.08 µmol, 1 equiv.), 3-(7-amino-1-methyl-indazol-3-yl)piperidine- 2,6-dione (37.99 mg, 147.08 µmol, 1 equiv.), and EDCI (42.29 mg, 220.62 µmol, 1.5 equiv.) in pyridine (1 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 4 hours under N 2 atmosphere. The reaction mixture was diluted with water 5 mL and extracted with ethyl acetate 10 mL (5 mL × 2). The combined organic layers were washed with brine (5 mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[(3S)-1-[2-[[3-(2,6 -dioxo-3-piperidyl)-1-methyl- indazol-7-yl]amino]-2-oxo-ethyl]pyrrolidin-3-yl]ethoxy]-2-me thyl-phenyl]pyridine-2- carboxylate (186 mg, crude) was obtained as a red oil. Step B. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[2-[(3S)-1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-7- yl]amino]-2-oxo-ethyl]pyrrolidin-3-yl]ethoxy]-2-methyl-pheny l]pyridine-2-carboxylic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[2-[(3S)-1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl- indazol-7-yl]amino]-2-oxo- ethyl]pyrrolidin-3-yl]ethoxy]-2-methyl-phenyl]pyridine-2-car boxylate (100 mg, 101.20 µmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 24 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep- HPLC. The compound 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[2-[(3S)-1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl- indazol-7-yl]amino]-2-oxo- ethyl]pyrrolidin-3-yl]ethoxy]-2-methyl-phenyl]pyridine-2-car boxylic acid (48.5 mg, 51.3 µmol, 50.7% yield, 98.6% purity) was obtained as a yellow solid. MS (ESI) m/z: 933.0 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.89 - 12.83 (m, 1H), 12.67 - 12.45 (m, 1H), 10.91 (s, 1H), 10.45 (d, J = 8.8 Hz, 1H), 10.29 - 10.07 (m, 1H), 8.02 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 7.6 Hz, 1H), 7.71 - 7.56 (m, 2H), 7.50 - 7.30 (m, 5H), 7.25 - 7.06 (m, 3H), 7.01 - 6.87 (m, 2H), 6.65 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.43 - 4.37 (m, 2H), 4.05 (s, 5H), 3.91 (t, J = 6.0 Hz, 2H), 3.71 - 3.62 (m, 1H), 3.03 (t, J = 5.6 Hz, 2H), 2.74 - 2.60 (m, 3H), 2.43 - 2.26 (m, 6H), 2.23 - 2.09 (m, 2H), 1.99 - 1.85 (m, 5H) Example 107. Preparation of Compound 231b 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[2-[(3R)-1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-o xo-ethyl]pyrrolidin-3- yl]ethoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[(3R)-1-[2-[[3-(2,6 -dioxo-3-piperidyl)-1-methyl- indazol-7-yl]amino]-2-oxo-ethyl]pyrrolidin-3-yl]ethoxy]-2-me thyl-phenyl]pyridine-2- carboxylate A mixture of 2-[(3R)-3-[2-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]ethyl]pyrrolidin-1- yl]acetic acid (65 mg, 86.91 µmol, 1 equiv.), 3-(7-amino-1-methyl-indazol-3-yl)piperidine-2,6- dione (26.94 mg, 104.29 µmol, 1.2 eq), HATU (33.05 mg, 86.91 µmol, 1 equiv.), and DIPEA (33.70 mg, 260.73 µmol, 45.41 µL, 3 equiv.) in DMF (3 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 4 hours under N2 atmosphere. The reaction mixture was added to water (3 mL), and then filtered. The mixture was concentrated to give a residue under reduced pressure. The residue was used in the next step without purification. The compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[2-[(3R)-1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-7- yl]amino]-2-oxo-ethyl]pyrrolidin-3-yl]ethoxy]-2-methyl-pheny l]pyridine-2-carboxylate (70 mg, crude) was obtained as a yellow solid. MS (ESI) m/z: 988.8 [M+H] + Step B. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[2-[(3R)-1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-7- yl]amino]-2-oxo-ethyl]pyrrolidin-3-yl]ethoxy]-2-methyl-pheny l]pyridine-2-carboxylic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[2-[(3R)-1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl- indazol-7-yl]amino]-2-oxo- ethyl]pyrrolidin-3-yl]ethoxy]-2-methyl-phenyl]pyridine-2-car boxylate (70 mg, 70.84 µmol, 1 equiv.) in TFA (2 mL) and DCM (2 mL) was stirred at 40 °C for 3 hours. The reaction mixture was concentrated under reduced pressure to remove DCM and TFA. The residue was purified by prep-HPLC. The compound 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[2-[(3R)-1-[2-[[3-(2,6-dioxo-3-piperi dyl)-1-methyl-indazol-7- yl]amino]-2-oxo-ethyl]pyrrolidin-3-yl]ethoxy]-2-methyl-pheny l]pyridine-2-carboxylic acid (24.2 mg, 25.8 µmol, 36.5% yield, 99.3% purity) was obtained as a yellow solid. MS (ESI) m/z: 932.0 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 13.55 - 12.12 (m, 1H), 10.90 (s, 1H), 9.90 (s, 1H), 8.14 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.57 (d, J = 8.0 Hz, 1H), 7.49 - 7.42 (m, 3H), 7.39 - 7.32 (m, 2H), 7.26 (d, J = 7.2 Hz, 1H), 7.11 - 7.04 (m, 2H), 6.96 (d, J = 8.8 Hz, 1H), 6.90 (d, J = 8.0 Hz, 1H), 6.63 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.39 - 4.35 (m, 1H), 4.07 (s, 3H), 4.01 - 3.98 (m, 2H), 3.93 - 3.90 (m, 2H), 3.05 - 3.00 (m, 4H), 2.89 - 2.80 (m, 2H), 2.71 - 2.61 (m, 4H), 2.40 - 2.36 (m, 1H), 2.18 - 2.13 (m, 1H), 2.10 - 2.05 (m, 2H), 1.92 - 1.85 (m, 5H), 1.59 - 1.52 (m, 1H). Example 108. Preparation of Compound 237

6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[9-[4-[3-(2,6- dioxo-3-piperidyl)-1-methyl-indazol-7-yl]piperazin-1-yl]nono xy]-2-methyl-phenyl]pyridine- 2-carboxylic acid Step A. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[9-[4-[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol- 7-yl]piperazin-1-yl]nonoxy]-2-methyl-phenyl]pyridine-2-carbo xylate To a solution of 3-(1-methyl-7-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (44.67 mg, 136.44 µmol, 1 equiv.) in DCM (5 mL) was added tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[2-methyl-3- (9- oxononoxy)phenyl]pyridine-2-carboxylate (100 mg, 136.44 µmol, 1 equiv.) and AcOH (8.19 mg, 136.44 µmol, 7.80 µL, 1 equiv.). The mixture was stirred at 25 °C for 1 hour, and then NaBH(OAc)3 (28.92 mg, 136.44 µmol, 1 equiv.) was added. The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to remove solvent to give the crude product tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[9-[4-[3-(2,6-dioxo-3-piperidyl)-1-me thyl-indazol-7-yl]piperazin-1- yl]nonoxy]-2-methyl-phenyl]pyridine-2-carboxylate (100 mg, 95.7 µmol, 70.1% yield) as a yellow solid. MS (ESI) m/z: 523.1 [M/2+H] + Step B. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[9-[4-[3-(2,6-dioxo-3-piperidyl)-1 -methyl-indazol-7- yl]piperazin-1-yl]nonoxy]-2-methyl-phenyl]pyridine-2-carboxy lic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[9-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol -7-yl]piperazin-1-yl]nonoxy]-2- methyl-phenyl]pyridine-2-carboxylate (100 mg, 95.76 µmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL). The mixture was stirred at 40 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by reverse- phase HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[9-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7 -yl]piperazin-1-yl]nonoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid (35.9 mg, 35.3 µmol, 36.9% yield, 97.1% purity) as a white solid. MS (ESI) m/z: 988.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.38 - 12.40 (m, 1H), 10.92 (s, 1H), 8.20 (s, 1H), 8.09 (d, J = 8.0 Hz, 1H), 7.85 (d, J = 8.0 Hz, 1H), 7.68 (d, J = 7.6 Hz, 1H), 7.58 - 7.47 (m, 4H), 7.38 (s, 2H), 7.17 - 7.12 (m, 1H), 7.03 - 6.90 (m, 4H), 6.69 (d, J = 7.6 Hz, 1H), 5.03 (s, 2H), 4.32 (dd, J = 5.2, 9.2 Hz, 1H), 4.02 ( t, J = 6.0 Hz, 2H), 3.98 - 3.93 (m, 5H), 3.44 - 3.42 (m, 5H), 3.05 ( t, J = 5.6 Hz, 2H), 2.75 - 2.64 (m, 6H), 2.41 - 2.29 (m, 2H), 2.26 - 2.18 (m, 1H), 1.96 (s, 3H), 1.82 - 1.75 (m, 2H), 1.57 - 1.48 (m, 4H), 1.36 (s, 8H). Example 109. Preparation of Compound 242 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4-[3-[4-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-oxo-e thyl]piperazin-1-yl]propoxy]- 2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[3-[4-[2-[[3-(2,6-diox o-3-piperidyl)-1-methyl- indazol-7-yl]amino]-2-oxo-ethyl]piperazin-1-yl]propoxy]-2-me thyl-phenyl]pyridine-2- carboxylate To a solution of 2-[4-[3-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-3-methyl- phenoxy]propyl]piperazin-1- yl]acetic acid (80 mg, 102.97 µmol, 1 equiv.) in DMF (1 mL) was added HATU (78.30 mg, 205.94 µmol, 2 equiv.), DIPEA (39.92 mg, 308.90 µmol, 53.81 µL, 3 equiv.), and 3-(7-amino- 1-methyl-indazol-3-yl)piperidine-2,6-dione (53.19 mg, 205.94 µmol, 2 equiv.). The mixture was stirred at 25 °C for 3 hours. The reaction mixture was quenched by addition of H 2 O (2 mL). The reaction mixture was filtered and washed with water (5 mL). The mixture was then diluted with DCM (10 mL). The organic layers were dried over Na2SO4, filtered, and concentrated under reduced pressure to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[3-[4-[2-[[3-(2,6-diox o-3-piperidyl)-1-methyl-indazol- 7-yl]amino]-2-oxo-ethyl]piperazin-1-yl]propoxy]-2-methyl-phe nyl]pyridine-2-carboxylate (100 mg, crude) as a yellow solid. MS (ESI) m/z: 1017.5 [M+H] + . Step B. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[4-[3-[4-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-7- yl]amino]-2-oxo-ethyl]piperazin-1-yl]propoxy]-2-methyl-pheny l]pyridine-2-carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[4-[3-[4-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-ind azol-7-yl]amino]-2-oxo- ethyl]piperazin-1-yl]propoxy]-2-methyl-phenyl]pyridine-2-car boxylate (100 mg, 98.31 µmol, 1 equiv.) in DCM (0.5 mL) was added TFA (770.00 mg, 6.75 mmol, 500.00 µL, 68.69 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product was purified by prep-HPLC to give 6- [8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinol in-2-yl]-3-[4-[3-[4-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-oxo-e thyl]piperazin-1-yl]propoxy]- 2-methyl-phenyl]pyridine-2-carboxylic acid (13.2 mg, 12.3 µmol, 12.5% yield, 89.4% purity) as a yellow solid. MS (ESI) m/z: 961.7 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.88 (s, 1H), 9.83 (s, 1H), 8.14 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.4 Hz, 1H), 7.62 (d, J = 6.4 Hz, 1H), 7.57 (d, J = 7.6 Hz, 1H), 7.51 - 7.41 (m, 3H), 7.40 - 7.29 (m, 3H), 7.08 (t, J = 7.6 Hz, 1H), 6.98 - 6.89 (m, 2H), 6.79 (d, J = 1.6 Hz, 1H), 6.73 - 6.69 (m, 1H), 4.97 (s, 2H), 4.32 – 4.33 (m, 1H), 4.12 (s, 2H), 4.07 - 3.95 (m, 3H), 3.91 (t, J = 5.2 Hz, 2H), 3.64 - 3.56 (m, 2H), 3.20 (s, 3H), 3.03 (t, J = 5.6 Hz, 4H), 2.96 (s, 1H), 2.64 - 2.58 (m, 4H), 2.25 - 2.13 (m, 3H), 2.07 - 1.99 (m, 4H), 1.91 - 1.83 (m, 2H). Example 110. Preparation of Compound 246a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((2R)-4-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino )-2-oxoethyl)-2- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((2R)-4-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-2-oxoethyl)-2-(trifluoromethyl)pipera zin-1-yl)propoxy)-2- methylphenyl)picolinate To a solution of (R)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)-3- (trifluoromethyl)piperazin-1-yl)acetic acid (90 mg, 106.52 µmol, 1.0 equiv.) and 3-(7-amino- 1-methyl-1H-indazol-3-yl)piperidine-2,6-dione (27.51 mg, 106.52 µmol, 1.0 equiv.) in pyridine (1.5 mL) was added EDCI (30.63 mg, 159.77 µmol, 1.5 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was diluted with water (2 mL) and extracted with ethyl acetate (2 mL × 5). The combined organic layers were washed with brine (5 mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give tert-butyl 6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2( 1H)-yl)-3-(3-(3-((2R)-4-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino)-2- oxoethyl)-2- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linate (100 mg, crude) as a white solid. MS (ESI) m/z: 1085.4 [M+H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((2R)-4-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-2-oxoethyl)-2-(trifluoromethyl)pipera zin-1-yl)propoxy)-2- methylphenyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-((2R)-4-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-7-yl)amino)-2- oxoethyl)-2-(trifluoromethyl)piperazin-1-yl)propoxy)-2-methy lphenyl)picolinate (100 mg, 92.15 µmol, 1.0 equiv.) in DCM (1 mL) was added TFA (770.00 mg, 6.75 mmol, 0.5 mL, 73.28 equiv.) was stirred at 25 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-((2R)-4-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino)-2- oxoethyl)-2- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linic acid (72.0 mg, 69.5 µmol, 75.4% yield, 99.3% purity) as a white solid. MS (ESI) m/z: 1029.4 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 13.13 - 12.14 (m, 2H), 10.90 (s, 1H), 9.90 - 9.62 (m, 1H), 8.13 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.61 (t, J = 6.4 Hz, 2H), 7.51 - 7.41 (m, 3H), 7.41 - 7.31 (m, 2H), 7.21 (d, J = 7.2 Hz, 1H), 7.13 - 7.05 (m, 2H), 6.97 (d, J = 8.8 Hz, 1H), 6.88 (d, J = 8.4 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.38 (dd, J = 4.8, 5.2 Hz, 1H), 4.07 (s, 3H), 4.00 (t, J = 5.6 Hz, 2H), 3.92 (t, J = 5.6 Hz, 2H), 3.65 - 3.46 (m, 2H), 3.03 (t, J = 5.6 Hz, 3H), 2.95 - 2.80 (m, 4H), 2.78 - 2.54 (m, 6H), 2.43 - 2.31 (m, 1H), 2.23 - 2.11 (m, 1H), 1.97 - 1.86 (m, 5H). Example 111. Preparation of Compound 255 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[1-[2-[4-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]-1-piperidyl]e thyl]-4-piperidyl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 4-[3-(2,6-dibenzyloxy-3-pyridyl)-1- methyl-indazol-6-yl]-3,6-dihydro-2H-pyridine-1-carboxylate A mixture of 6-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (3.2 g, 6.40 mmol, 1.00 equiv.), tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydro- 2H- pyridine-1-carboxylate (2 g, 6.47 mmol, 1.01 equiv.), K 3 PO 4 (4.07 g, 19.19 mmol, 3.00 equiv.), Ad2nBuP Pd G3 (465.73 mg, 639.51 µmol, 0.10 equiv.), and H2O (3.20 g, 177.63 mmol, 3.20 mL, 27.78 equiv.) in dioxane (25 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 60 °C for 16 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~20% ethyl acetate/petroleum ether) to give tert-butyl 4-[3- (2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-yl]-3,6-dihyd ro-2H-pyridine-1-carboxylate (3.5 g, 5.8 mmol, 90.8% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ = 7.92 (d, J = 8.0 Hz, 1H), 7.64 (d, J = 8.8 Hz, 1H), 7.59 (s, 1H), 7.51 - 7.45 (m, 2H), 7.42 - 7.28 (m, 8H), 7.17 (dd, J = 1.2, 8.4 Hz, 1H), 6.59 (d, J = 8.0 Hz, 1H), 6.26 (s, 1H), 5.46 (s, 2H), 5.43 (s, 2H), 4.08 - 4.06 (m, 3H), 4.05 - 4.03 (m, 2H), 3.59 (t, J = 5.6 Hz, 2H), 2.62 - 2.55 (m, 2H), 1.45 (s, 9H) Step B. Procedure for preparation of tert-butyl 4-[3-(2,6-dioxo-3-piperidyl)-1-methyl- indazol-6-yl]piperidine-1-carboxylate A mixture of tert-butyl 4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-yl]-3,6- dihydro- 2H-pyridine-1-carboxylate (3.5 g, 5.81 mmol, 1.00 equiv.), Pd/C (3 g, 5.81 mmol, 10% purity, 1.00 equiv.), and EtOH (15 mL) in THF (15 mL) was degassed and purged with H2 (15 Psi) three times, and then the mixture was stirred at 25 °C for 16 hours under H2 (15 Psi) atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give crude tert-butyl 4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]piperidin e-1-carboxylate (2.5 g, crude) as a black solid. MS (ESI) m/z 427.3 [M+H] + . Step C. Procedure for preparation of 3-[1-methyl-6-(4-piperidyl)indazol-3-yl]piperidine- 2,6-dione To a solution of tert-butyl 4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]piperidin e-1- carboxylate (2.5 g, 5.86 mmol, 1.00 equiv.) in HCl/dioxane (20 mL) stirred at 25 °C for 1 hour. The reaction mixture was filtered to give crude 3-[1-methyl-6-(4-piperidyl)indazol-3- yl]piperidine-2,6-dione (1.5 g, 4.6 mmol, 78.4% yield) as a white solid. Step D. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[4-[3-(2,6-dioxo-3-piperi dyl)-1-methyl-indazol-6-yl]-1- piperidyl]ethyl]-4-piperidyl]propoxy]-2-methyl-phenyl]pyridi ne-2-carboxylic acid A solution of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[2- methyl-3-[3-[1-(2-oxoethyl)-4-piperidyl]propoxy]phenyl]pyrid ine-2-carboxylic acid (80 mg, 113.66 µmol, 1.00 equiv.) and 3-[1-methyl-6-(4-piperidyl)indazol-3-yl]piperidine-2,6-dione (37.10 mg, 113.66 µmol, 1.00 equiv.) in DCM (1 mL) and IPA (0.1 mL) was stirred at 25 °C for 15 hours. Then NaBH(OAc)3 (72.27 mg, 340.98 µmol, 3 equiv.)was added into the solution. The mixture was stirred at 25 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8- (1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin- 2-yl]-3-[3-[3-[1-[2-[4-[3-(2,6- dioxo-3-piperidyl)-1-methyl-indazol-6-yl]-1-piperidyl]ethyl] -4-piperidyl]propoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid (14.0 mg, 11.9 µmol, 10.5% yield, 90.5% purity) as a yellow solid. MS (ESI) m/z 1014.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.87 (s, 1H), 8.23 (s, 2H), 8.01 (d, J = 7.6 Hz, 1H), 7.77 (d, J = 8.0 Hz, 1H), 7.64 - 7.56 (m, 2H), 7.49 - 7.43 (m, 1H), 7.42 - 7.38 (m, 2H), 7.37 - 7.30 (m, 3H), 7.09 - 6.99 (m, 2H), 6.89 - 6.81 (m, 2H), 6.66 (d, J = 7.6 Hz, 1H), 4.94 (s, 2H), 4.31 (dd, J = 5.2, 9.6 Hz, 1H), 4.00 - 3.95 (m, 2H), 3.95 (s, 3H), 3.87 (t, J = 5.2 Hz, 2H), 3.04 - 2.94 (m, 8H), 2.63 - 2.59 (m, 2H), 2.18 - 2.03 (m, 6H), 1.90 (s, 3H), 1.82 - 1.65 (m, 8H), 1.62 - 1.51 (m, 2H), 1.40 - 1.24 (m, 4H), 1.16 - 0.99 (m, 2H) Example 112. Preparation of Compound 261 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[2-[1-[[1-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]-4-piperidyl]m ethyl]-4-piperidyl]ethoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[2-[1-[[1-[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-7-yl]-4- piperidyl]methyl]-4-piperidyl]ethoxy]-2-methyl-phenyl]pyridi ne-2-carboxylic acid To a solution of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- [2-methyl-3-[2-(4-piperidyl)ethoxy]phenyl]pyridine-2-carboxy lic acid (130 mg, 189.99 µmol, 1.0 equiv., HCl) and 1-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]piperidin e-4- carbaldehyde (74.07 mg, 208.99 µmol, 1.1 equiv.) in DCM (1 mL) and IPA (0.1 mL) was stirred at 25 °C for 12 hours. Then NaBH(OAc)3 (120.80 mg, 569.97 µmol, 3.0 equiv.) was added at 0 °C. After addition, the mixture was stirred at 25 °C for 0.5 hour. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[2-[1- [[1-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]-4-pipe ridyl]methyl]-4- piperidyl]ethoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid (36.1 mg, 33.8 µmol, 17.8% yield, 96.5% purity) as a pink solid. MS (ESI) m/z: 986.5 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.87 (s, 1H), 8.02 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.49 - 7.30 (m, 6H), 7.08 (t, J = 8.0 Hz, 1H), 7.03 - 6.97 (m, 2H), 6.95 - 6.86 (m, 2H), 6.64 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.32 (dd, J = 5.2, 9.6 Hz, 1H), 4.23 (s, 3H), 4.02 - 3.96 (m, 2H), 3.91 (t, J = 5.6 Hz, 2H), 3.25 - 3.21 (m, 4H), 3.04 - 3.00 (m, 2H), 2.86 (d, J = 9.6 Hz, 2H), 2.70 - 2.60 (m, 4H), 2.25 - 2.12 (m, 3H), 1.95 - 1.81 (m, 7H), 1.72 - 1.65 (m, 4H), 1.54 - 1.45 (m, 1H), 1.41 - 1.29 (m, 2H), 1.28 - 1.19 (m, 2H) Example 113. Preparation of Compound 284b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1R,4r)-4- ((2R)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 7-yl)piperazin-1-yl)-2- methylpropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1R,4r)-4- ((2R*)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -7-yl)piperazin-1-yl)-2- methylpropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of 3-(7-(4-((R)-3-((1r,4R)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2-methylpropyl)piperazin-1-yl)-1-m ethyl-1H-indazol-3- yl)piperidine-2,6-dione To a solution of 3-(1-methyl-7-(piperazin-1-yl)-1H-indazol-3-yl)piperidine-2, 6-dione (128.69 mg, 353.70 µmol, 1.2 equiv., HCl) in DCM (2 mL) was added NaBH(OAc)3 (187.41 mg, 884.26 µmol, 3 equiv.). Then (R)-3-((1r,4R)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2- methylpropanal (100 mg, 294.75 µmol, 1 equiv.) was added into the mixture and stirred at 0 °C for 1 hour. Then the mixture was stirred at 25 °C for 15 hours. DCM (30 mL) and water (30 mL) were added, and layers were separated. The aqueous phase was extracted with DCM (10 mL × 2). The combined organic extracts were dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=4/1 to DCM: MeOH = 10:1) to give 3-(7-(4-((R)-3-((1r,4R)-4- (3-bromo-2-methylphenoxy)cyclohexyl)-2-methylpropyl)piperazi n-1-yl)-1-methyl-1H- indazol-3-yl)piperidine-2,6-dione (80 mg, 122.95 µmol, 41.71% yield) as a yellow solid. MS (ESI) m/z: 652.1 [M+H] + ( 80 Br) Step B. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-((2R)-3-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)-2-methylpropyl)cycloh exyl)oxy)-2- methylphenyl)picolinate A mixture of 3-(7-(4-((R)-3-((1r,4R)-4-(3-bromo-2-methylphenoxy)cyclohexy l)-2- methylpropyl)piperazin-1-yl)-1-methyl-1H-indazol-3-yl)piperi dine-2,6-dione (80.00 mg, 122.95 µmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)picolinate (90.38 mg, 147.55 µmol, 1.2 equiv.), KF (21.43 mg, 368.86 µmol, 8.64 µL, 3 equiv.), H2O (0.1 mL), and [2-(2-aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-buty l- phosphane;methanesulfonate (8.95 mg, 12.30 µmol, 0.1 equiv.) in dioxane (1 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 100 °C for 1 hour under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate 3/1, DCM: MeOH = 10:1) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-((2R)-3-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)-2-methylpropyl)cycloh exyl)oxy)-2- methylphenyl)picolinate (90 mg, 85.2 µmol, 69.3% yield) as a yellow solid. MS (ESI) m/z: 1056.4 [M+H] + Step C. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-((2R)-3-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)-2-methylpropyl)cycloh exyl)oxy)-2- methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-((2R*)-3-(4-(3 -(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)-2-methylpropyl)cycloh exyl)oxy)-2- methylphenyl)picolinic acid] To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1R,4r)-4-((2R)-3-(4-(3-(2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-7- yl)piperazin-1-yl)-2-methylpropyl)cyclohexyl)oxy)-2-methylph enyl)picolinate (90 mg, 85.20 µmol, 1 equiv.) in DCM (1 mL) was added TFA (2.31 g, 20.26 mmol, 1.50 mL, 237.78 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1R,4r)-4-((2R)-3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pip erazin-1-yl)-2- methylpropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r) -4-((2R*)-3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin-1-yl )-2- methylpropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] (40.4 mg, 39.5 µmol, 46.3% yield, 97.7% purity) as a white solid. MS (ESI) m/z: 1000.5 [M+H] + . 1 H NMR (400 MHz, DMSO) δ = 12.87 (s, 1H), 12.68 - 12.47 (m, 1H), 10.90 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 8.4 Hz, 1H), 7.50 - 7.42 (m, 4H), 7.40 - 7.32 (m, 2H), 7.12 - 7.03 (m, 3H), 6.99 - 6.91 (m, 2H), 6.62 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.39 - 4.33 (m, 1H), 4.26 (s, 3H), 4.23 - 4.16 (m, 1H), 3.91 ( t, J = 6.0 Hz, 2H), 3.70 - 3.50 (m, 2H), 3.22 - 3.11 (m, 6H), 3.02 (t, J = 5.6 Hz, 2H), 2.63 - 2.52 (m, 4H), 2.18 (d, J = 5.6 Hz, 1H), 2.16 - 2.02 (m, 4H), 1.87 (s, 3H), 1.83 - 1.72 (m, 2H), 1.52 - 1.28 (m, 4H), 1.27 - 1.21 (m, 1H), 1.18 - 1.09 (m, 2H), 1.00 (d, J = 6.0Hz, 3H) Example 114. Preparation of Compound 287 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[2-[1-[3-[1-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]-4-piperidyl]p ropyl]-4-piperidyl]ethoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 3-(2,6-dibenzyloxy-3-pyridyl)-6-[4-(3,3- dimethoxypropyl)-1-piperidyl]-1-methyl-indazole A mixture of 6-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (200.00 mg, 399.69 µmol, 1.0 equiv.), 4-(3,3-dimethoxypropyl)piperidine (97.31 mg, 519.60 µmol, 1.3 equiv.), Cs2CO3 (390.68 mg, 1.20 mmol, 3.0 equiv.), and 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5- dichloro-2H-imidazol-1-ium-2-ide;3-chloropyridine;dichloropa lladium (38.88 mg, 39.97 µmol, 0.1 equiv.) in 2-methyl-2-butanol (2 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 100 °C for 16 hours under N2 atmosphere. The mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~30% ethyl acetate/petroleum ether) to give 3-(2,6- dibenzyloxy-3-pyridyl)-6-[4-(3,3-dimethoxypropyl)-1-piperidy l]-1-methyl-indazole (150 mg, 236.6 µmol, 59.2% yield, 95.7% purity) as a green oil. MS (ESI) m/z: 607.5 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.88 (d, J = 8.0 Hz, 1H), 7.50 - 7.44 (m, 3H), 7.42 - 7.24 (m, 8H), 6.83 - 6.77 (m, 2H), 6.56 (d, J = 8.0 Hz, 1H), 5.44 (s, 2H), 5.41 (s, 2H), 4.33 (t, J = 5.6 Hz, 1H), 3.95 (s, 3H), 3.79 - 3.72 (m, 2H), 3.22 (s, 6H), 2.73 - 2.64 (m, 2H), 1.80 - 1.72 (m, 2H), 1.60 - 1.52 (m, 2H), 1.47 - 1.38 (m, 1H), 1.30 - 1.22 (m, 4H) Step B. Procedure for preparation of 3-[6-[4-(3,3-dimethoxypropyl)-1-piperidyl]-1- methyl-indazol-3-yl]piperidine-2,6-dione To a solution of 3-(2,6-dibenzyloxy-3-pyridyl)-6-[4-(3,3-dimethoxypropyl)-1-p iperidyl]-1- methyl-indazole (150 mg, 247.22 µmol, 1.0 equiv.) in THF (2 mL) and EtOH (2 mL) was added Pd(OH) 2 (100 mg, 712.05 µmol, 2.88 equiv.) and Pd/C (100 mg, 10% purity) under N 2 . The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H2 (15 psi) at 20°C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give 3-[6-[4-(3,3-dimethoxypropyl)-1-piperidyl]-1- methyl-indazol-3-yl]piperidine-2,6-dione (90 mg, crude) as a yellow oil. MS (ESI) m/z: 429.2 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 10.84 (s, 1H), 7.47 (d, J = 9.2 Hz, 1H), 6.91 - 6.87 (m, 1H), 6.83 - 6.80 (m, 1H), 4.37 - 4.30 (m, 2H), 4.27 - 4.22 (m, 1H), 3.88 (s, 3H), 3.79 - 3.73 (m, 2H), 3.46 - 3.42 (m, 1H), 3.22 (s, 6H), 2.72 - 2.58 (m, 4H), 1.80 - 1.72 (m, 2H), 1.59 - 1.53 (m, 2H), 1.38 (s, 1H), 1.28 - 1.21 (m, 4H) Step C. Procedure for preparation of 3-[1-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6- yl]-4-piperidyl]propanal A mixture of 3-[6-[4-(3,3-dimethoxypropyl)-1-piperidyl]-1-methyl-indazol- 3-yl]piperidine- 2,6-dione (90 mg, 210.02 µmol, 1.0 equiv.) and HCOOH (10.09 mg, 210.02 µmol, 1 mL, 1.0 equiv.) was stirred at 90 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give 3-[1-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]-4-pip eridyl]propanal (75 mg, crude) as a pink oil. MS (ESI) m/z: 383.3 [M+H] + Step D. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[2-[1-[3-[1-[3-(2,6-dioxo-3-piperi dyl)-1-methyl-indazol-6-yl]-4- piperidyl]propyl]-4-piperidyl]ethoxy]-2-methyl-phenyl]pyridi ne-2-carboxylic acid To a solution of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- [2-methyl-3-[2-(4-piperidyl)ethoxy]phenyl]pyridine-2-carboxy lic acid (100 mg, 146.15 µmol, 1.0 equiv., HCl) and 3-[1-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]-4- piperidyl]propanal (72.66 mg, 189.99 µmol, 1.3 equiv.) in DCM (3 mL) and t-BuOH (1 mL) was stirred at 20 °C for 3 hours. NaBH(OAc)3 (278.77 mg, 1.32 mmol, 9.0 equiv.) was added at 0 °C. Then the mixture was warmed to 20 °C and stirred at 20 °C for 0.5 hour. The mixture was concentrated under reduced pressure to remove DCM and t-BuOH. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[2-[1-[3-[1-[3-(2,6-dioxo-3-piperidyl )-1-methyl-indazol-6-yl]-4- piperidyl]propyl]-4-piperidyl]ethoxy]-2-methyl-phenyl]pyridi ne-2-carboxylic acid (58.69 mg, 57.38 μmol, 39.26% yield, 99.16% purity) as a white solid. MS (ESI) m/z: 1015.0 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.85 (s, 1H), 8.01 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 6.4 Hz, 1H), 7.50 - 7.39 (m, 3H), 7.39 - 7.27 (m, 3H), 7.10 - 7.04 (m, 1H), 6.92 - 6.82 (m, 3H), 6.81 (s, 1H), 6.66 (d, J = 7.6 Hz, 1H), 4.96 (s, 2H), 4.26 - 4.22 (m, 1H), 3.98 - 3.94 (m, 2H), 3.92 - 3.89 (m, 2H), 3.87 (s, 3H), 3.80 - 3.71 (m, 4H), 3.00 (t, J = 5.6 Hz, 2H), 2.98 - 2.92 (m, 2H), 2.70 - 2.64 (m, 2H), 2.64 - 2.58 (m, 2H), 2.48 - 2.41 (m, 2H), 2.18 - 2.12 (m, 2H), 1.90 (s, 3H), 1.74 (d, J = 11.6 Hz, 2H), 1.70 - 1.58 (m, 4H), 1.52 (s, 3H), 1.43 - 1.32 (m, 1H), 1.31 - 1.11 (m, 6H) Example 115. Preparation of Compound 288 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[2-[1-[3-[1-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]-4-piperidyl]p ropyl]-4-piperidyl]ethoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of benzyl 4-(3-oxopropyl)piperidine-1-carboxylate To a solution of (COCl) 2 (1.83 g, 14.42 mmol, 1.26 mL, 2.0 equiv.) in DCM (20 mL) was added DMSO (2.25 g, 28.84 mmol, 2.25 mL, 4.0 equiv.) dropwise slowly at -70 °C under N2 atmosphere, and then the mixture was stirred at -70 °C for 1 hour. To the mixture was added benzyl 4-(3-hydroxypropyl)piperidine-1-carboxylate (2 g, 7.21 mmol, 1.0 equiv.) dropwise slowly at -70 °C, and the mixture was stirred for 1 hour. Then TEA (4.38 g, 43.27 mmol, 6.02 mL, 6.0 equiv.) was added dropwise slowly at -70 °C, and then the mixture was stirred at 20 °C for 0.5 hour under N 2 atmosphere. The reaction mixture was diluted with H 2 O (50 mL) and extracted with DCM (50 mL × 3). The combined organic layers were washed with H 2 O (50 mL × 3), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue to benzyl 4-(3-oxopropyl)piperidine-1-carboxylate (1.97 g, crude) as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 9.69 – 9.63 (m, 1H), 7.48 – 7.20 (m, 5H), 5.05 (s, 2H), 4.04 – 3.93 (m, 2H), 2.83 – 2.66 (m, 2H), 2.48 – 2.42 (m, 2H), 1.68 – 1.57 (m, 2H), 1.50 – 1.42 (m, 2H), 1.42 – 1.33 (m, 1H), 1.04 – 0.91 (m, 2H) Step B. Procedure for preparation of benzyl 4-(3,3-dimethoxypropyl)piperidine-1- carboxylate To a solution of benzyl 4-(3-oxopropyl)piperidine-1-carboxylate (1.97 g, 7.15 mmol, 1 equiv.) in MeOH (20 mL) was added TsOH (1.48 g, 8.59 mmol, 1.2 equiv.) and 4A MS (3 g, 7.15 mmol, 1.0 equiv.). The mixture was stirred at 80 °C for 24 hours. The mixture was filtered to give a filtrate, which was treated with saturated NaHCO3 solution to pH= 8 and concentrated under reduced pressure to give a residue. The residue was diluted with H 2 O (50 mL) and extracted with DCM (50 mL × 3). The combined organic layers were dried over Na2SO4, filtered, and concentrated under reduced pressure to give benzyl 4-(3,3- dimethoxypropyl)piperidine-1-carboxylate (1.9 g, crude) as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.40 - 7.28 (m, 5H), 5.05 (s, 2H), 4.30 (t, J = 5.6 Hz, 1H), 4.01 -3.94 (m, 2H), 3.20 (s, 6H), 2.84 - 2.66 (m, 2H), 1.67 - 1.60 (m, 2H), 1.54 - 1.47 (m, 2H), 1.43 - 1.33 (m, 1H), 1.23 - 1.16 (m, 2H), 1.02 - 0.91 (m, 2H) Step C. Procedure for preparation of 4-(3,3-dimethoxypropyl)piperidine To a solution of benzyl 4-(3,3-dimethoxypropyl)piperidine-1-carboxylate (1.9 g, 5.91 mmol, 1.0 equiv.) in THF (50 mL) was added Pd/C (500 mg, 10% purity) under N 2 . The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H2 (50 psi) at 50°C for 16 hours. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~100% Methanol / Dichloromethane) to give 4-(3,3- dimethoxypropyl)piperidine (655 mg, 3.1 mmol, 53.2% yield, 90.0% purity) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 4.29 (t, J = 5.6 Hz, 1H), 3.19 (s, 6H), 2.97 - 2.88 (m, 2H), 2.8 – 2.40 (m, 2H), 1.61 - 1.53 (m, 2H), 1.53 - 1.46 (m, 2H), 1.30 - 1.20 (m, 1H), 1.20 - 1.13 (m, 2H), 1.04 - 0.92 (m, 2H) Step D. Procedure for preparation of 3-(2,6-dibenzyloxy-3-pyridyl)-7-[4-(3,3- dimethoxypropyl)-1-piperidyl]-1-methyl-indazole A mixture of 7-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (200 mg, 399.69 µmol, 1.0 equiv.), 4-(3,3-dimethoxypropyl)piperidine (97.31 mg, 519.60 µmol, 1.3 equiv.), Cs 2 CO 3 (390.68 mg, 1.20 mmol, 3.0 equiv.), and 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5- dichloro-2H-imidazol-1-ium-2-ide;3-chloropyridine;dichloropa lladium (38.88 mg, 39.97 µmol, 0.1 equiv.) in 2-methyl-2-butanol (2 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 100 °C for 16 hours under N2 atmosphere. The mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~15% ethyl acetate/petroleum ether) to give 3-(2,6- dibenzyloxy-3-pyridyl)-7-[4-(3,3-dimethoxypropyl)-1-piperidy l]-1-methyl-indazole (100 mg, 148.3 µmol, 37.1% yield, 90.0% purity) as a yellow oil. MS (ESI) m/z: 607.4 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 7.84 (d, J = 8.0 Hz, 1H), 7.50 - 7.44 (m, 2H), 7.42 - 7.33 (m, 5H), 7.32 - 7.26 (m, 4H), 7.01 - 6.96 (m, 1H), 6.95 - 6.89 (m, 1H), 6.58 (d, J = 8.0 Hz, 1H), 5.42 (d, J = 5.6 Hz, 4H), 4.35 (t, J = 5.6 Hz, 1H), 4.31 (s, 3H), 3.23 (s, 8H), 2.71 - 2.63 (m, 2H), 1.85 - 1.76 (m, 2H), 1.61 - 1.55 (m, 2H), 1.44 - 1.36 (m, 3H), 1.34 - 1.29 (m, 2H) Step E. Procedure for preparation of 3-[7-[4-(3,3-dimethoxypropyl)-1-piperidyl]-1- methyl-indazol-3-yl]piperidine-2,6-dione To a solution of 3-(2,6-dibenzyloxy-3-pyridyl)-7-[4-(3,3-dimethoxypropyl)-1-p iperidyl]-1- methyl-indazole (100 mg, 164.81 µmol, 1.0 equiv.) in THF (1 mL) and EtOH (1 mL) was added Pd(OH) 2 (23.15 mg, 164.81 µmol, 1.0 equiv.) and Pd/C (50 mg, 10% purity) under N 2 . The suspension was degassed under vacuum and purged with H2 several times. The mixture was stirred under H 2 (15 psi) at 20°C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give 3-[7-[4-(3,3-dimethoxypropyl)-1-piperidyl]-1- methyl-indazol-3-yl]piperidine-2,6-dione (65 mg, crude) as a yellow oil. MS (ESI) m/z: 429.1 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.39 - 7.32 (m, 1H), 7.03 - 6.95 (m, 2H), 4.37 - 4.30 (m, 2H), 4.23 (s, 3H), 3.23 (s, 6H), 3.21 - 3.18 (m, 2H), 2.74 - 2.58 (m, 4H), 2.38 - 2.25 (m, 2H), 1.84 - 1.76 (m, 2H), 1.61 - 1.54 (m, 2H), 1.42 - 1.37 (m, 2H), 1.35 - 1.26 (m, 3H) Step F. Procedure for preparation of 3-[1-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7- yl]-4-piperidyl]propanal A mixture of 3-[7-[4-(3,3-dimethoxypropyl)-1-piperidyl]-1-methyl-indazol- 3-yl]piperidine- 2,6-dione (60 mg, 140.02 µmol, 1.0 equiv.) and HCOOH (6.73 mg, 140.02 µmol, 1 mL, 1.0 equiv.) was stirred at 90 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give 3-[1-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]-4-pip eridyl]propanal (60 mg, crude) as a pink oil. MS (ESI) m/z: 383.2 [M+H] + Step G. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[2-[1-[3-[1-[3-(2,6-dioxo-3-piperi dyl)-1-methyl-indazol-7-yl]-4- piperidyl]propyl]-4-piperidyl]ethoxy]-2-methyl-phenyl]pyridi ne-2-carboxylic acid To a solution of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- [2-methyl-3-[2-(4-piperidyl)ethoxy]phenyl]pyridine-2-carboxy lic acid (80 mg, 123.50 µmol, 1.0 equiv.) and NaBH(OAc) 3 (235.57 mg, 1.11 mmol, 9.0 equiv.) in DCM (2 mL) and t-BuOH (0.5 mL), and then 3-[1-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]-4- piperidyl]propanal (56.68 mg, 148.20 µmol, 1.2 equiv.) was added dropwise at 0 °C. After addition, the mixture was warmed to 20 °C and stirred at 20 °C for 16 hours. The mixture was concentrated under reduced pressure to remove DCM and t-BuOH. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[2-[1-[3-[1-[3-(2,6-dioxo-3-piperidyl)-1-methyl-ind azol-7-yl]-4-piperidyl]propyl]-4- piperidyl]ethoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid (46.3 mg, 41.8 µmol, 33.8% yield, 95.6% purity, FA) as a white solid. MS (ESI) m/z: 1015.5 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.89 (s, 1H), 8.01 (d, J = 8.0 Hz, 1H), 7.77 (d, J = 8.0 Hz, 1H), 7.61 (d, J = 7.6 Hz, 1H), 7.48 - 7.42 (m, 1H), 7.42 - 7.29 (m, 5H), 7.10 - 7.04 (m, 1H), 7.03 - 6.96 (m, 2H), 6.89 - 6.82 (m, 2H), 6.66 (d, J = 7.6 Hz, 1H), 4.95 (s, 2H), 4.35 - 4.31 (m, 1H), 4.22 (s, 3H), 3.98 - 3.94 (m, 2H), 3.90 (t, J = 5.6 Hz, 2H), 3.24 - 3.15 (m, 4H), 3.0 - 2.95 (m, 4H), 2.69 - 2.59 (m, 4H), 2.35 - 2.28 (m, 1H), 2.25 - 2.10 (m, 3H), 1.90 (s, 3H), 1.82 - 1.74 (m, 2H), 1.71 - 1.60 (m, 4H), 1.54 (s, 3H), 1.36 (s, 3H), 1.29 - 1.14 (m, 4H) Example 116. Preparation of Compound 289a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-((1S)-6-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino )-2-oxoethyl)-6- azaspiro[2.5]octan-1-yl)ethoxy)-2-methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(3-(2-((1S)-6- (2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)am ino)-2-oxoethyl)-6- azaspiro[2.5]octan-1-yl)ethoxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of tert-butyl (S)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(6-(2-ethoxy-2-oxoe thyl)-6-azaspiro[2.5]octan- 1-yl)ethoxy)-2-methylphenyl)picolinate A mixture of ethyl (S)-2-(1-(2-(3-bromo-2-methylphenoxy)ethyl)-6-azaspiro[2.5]o ctan-6- yl)acetate (100 mg, 243.70 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3, 2-dioxaborolan-2-yl)pyridine-2- carboxylate (179.13 mg, 292.44 µmol, 1.2 equiv.), [2-(2- aminophenyl)phenyl]palladium(1 + );bis(1-adamantyl)-butyl-phosphane;methanesulfonate (35.50 mg, 48.74 µmol, 0.2 equiv.), and K2CO3 (1.5 M, 487.40 µL, 3 equiv.) in dioxane (1 mL) and H 2 O (1 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 100 °C for 1 hour under N 2 atmosphere under microwave. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 60% ethyl acetate / petroleum ether) to give tert- butyl (S)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquino lin-2(1H)-yl)-3-(3-(2-(6- (2-ethoxy-2-oxoethyl)-6-azaspiro[2.5]octan-1-yl)ethoxy)-2-me thylphenyl)picolinate (140 mg, 171.6 µmol, 70.4% yield) as a yellow oil. MS (ESI) m/z: 816.4 [M+H] + . Step B. Procedure for preparation of (S)-2-(1-(2-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)ethyl)-6-azaspiro[2.5]octan-6-yl)acetic acid A mixture of tert-butyl (S)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquino lin- 2(1H)-yl)-3-(3-(2-(6-(2-ethoxy-2-oxoethyl)-6-azaspiro[2.5]oc tan-1-yl)ethoxy)-2- methylphenyl)picolinate (140 mg, 171.57 µmol, 1 equiv.) and LiOH•H2O (21.60 mg, 514.70 µmol, 3 equiv.) in THF (0.8 mL) and H 2 O (0.4 mL) was stirred at 25 °C for 2 hours. The reaction mixture was diluted with H2O (5 mL) and concentrated as a turbid liquid. Then the pH of the turbid liquid was adjusted to 3 with 1 N HCl (5 mL), and the mixture was extracted with DCM / MeOH (20:1), The combined organic layers were filtered and concentrated to give (S)- 2-(1-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin-2(1H)-yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)ethyl)-6-azaspi ro[2.5]octan-6-yl)acetic acid (120 mg, 152.3 µmol, 88.8% yield) as a yellow solid. MS (ESI) m/z: 788.3 [M+H] + Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((1S)-6-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-6-azaspiro[2.5]octan-1-yl )ethoxy)-2- methylphenyl)picolinate A mixture of (S)-2-(1-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)ethyl)-6- azaspiro[2.5]octan-6-yl)acetic acid (110 mg, 139.60 µmol, 1 equiv.), 3-(6-amino-1-methyl- indazol-3-yl)piperidine-2,6-dione (43.27 mg, 167.52 µmol, 1.2 equiv.), and EDCI (40.14 mg, 209.40 µmol, 1.5 equiv.) in pyridine (1.2 mL) was stirred at 25 °C for 2 hours. The mixture was diluted with H2O (10 mL × 3) and extracted with ethyl acetate (8 mL × 3). The combined organic layers were filtered and concentrated under reduced pressure to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(2-((1S)-6-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2- oxoethyl)-6- azaspiro[2.5]octan-1-yl)ethoxy)-2-methylphenyl)picolinate (100 mg, crude) as a yellow solid. MS (ESI) m/z: 1028.6 [M+H] + Step D. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((1S)-6-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-6-azaspiro[2.5]octan-1-yl )ethoxy)-2- methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((1S)-6-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-6-azaspiro[2.5]octan-1-yl )ethoxy)-2- methylphenyl)picolinic acid] A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(2-((1S)-6-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-6-yl)amino)-2- oxoethyl)-6-azaspiro[2.5]octan-1-yl)ethoxy)-2-methylphenyl)p icolinate (100 mg, 97.26 µmol, 1 equiv.) in TFA (0.1 mL) and DCM (0.1 mL) was stirred at 25 °C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep–HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((1S)-6-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)-6-azaspiro[2.5]octan-1-yl)et hoxy)-2-methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(3-(2- ((1S)-6-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)amino)-2-oxoethyl)-6- azaspiro[2.5]octan-1-yl)ethoxy)-2-methylphenyl)picolinic acid] (98.4% purity) as a yellow solid. MS (ESI) m/z: 972.4 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 12.94 - 12.80 (m, 1H), 10.93 - 10.82 (m, 1H), 10.19 - 9.74 (m, 1H), 8.06 - 7.99 (m, 2H), 7.81 - 7.76 (m, 1H), 7.67 - 7.60 (m, 2H), 7.47 - 7.34 (m, 5H), 7.21 - 7.07 (m, 2H), 6.98 - 6.89 (m, 2H), 6.63 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.33 (dd, J = 5.2, 9.6 Hz, 1H), 4.11 - 4.03 (m, 2H), 3.94 - 3.90 (m, 5H), 3.50 - 3.39 (m, 4H), 3.02 ( t, J = 5.2 Hz, 2H), 2.68 - 2.54 (m, 6H), 2.36 - 2.29 (m, 1H), 2.21 - 2.14 (m, 1H), 1.92 (s, 3H), 1.73 - 1.54 (m, 2H), 1.47 - 0.98 (m, 2H), 0.94 - 0.72 (m, 1H), 0.66 - 0.41 (m, 1H), 0.36 - 0.17 (m, 1H) Example 117. Preparation of Compound 101 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[2-[1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-oxo-e thyl]-4-piperidyl]ethoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 2-(tert-butyl) 8-methyl 3,4-dihydroisoquinoline- 2,8(1H)-dicarboxylate [SMILEs: O=C(N1CC2=C(C(OC)=O)C=CC=C2CC1)OC(C)(C)C] To a solution of 2-tert-butoxycarbonyl-3,4-dihydro-1H-isoquinoline-8-carboxyl ic acid (62 g, 223.57 mmol, 1 equiv.) in DMF (600 mL) was added MeI (38.08 g, 268.29 mmol, 16.70 mL, 1.2 equiv.) and K2CO3 (61.80 g, 447.15 mmol, 2 equiv.). The mixture was stirred at 50°C for 2 hours. The mixture was poured into ice water (800 mL) (pH>8), extracted with EtOAc (300 mL × 3). The organic layers were combined and washed with brine (300 mL ×3 ), dried over Na2SO4, and concentrated in vacuo to give product. The crude product 2-(tert-butyl) 8-methyl 3,4-dihydroisoquinoline-2,8(1H)-dicarboxylate (66 g, crude) as a brown oil was used in the next step without further purification. Step B. Procedure for preparation of tert-butyl 8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinoline-2-carboxylate To a solution of 1, 3-benzothiazol-2-amine (35.73 g, 237.87 mmol, 1.05 equiv.) in 2-MeTHF (350 mL) was added LiHMDS (1 M, 453.08 mL, 2 equiv.) at 0°C, and the solution was stirred at 0°C for 30 min. Then a solution of 2-(tert-butyl) 8-methyl 3,4-dihydroisoquinoline-2,8(1H)- dicarboxylate (66.87 g, 226.54 mmol, 98.7% purity, 1 equiv.) in 2-MeTHF (350 mL) was added to the above solution at 0°C and stirred for 30 min again. The resulting mixture was warmed to 60°C and stirred for 17 hours. The reaction mixture was poured into sat.NH4Cl (1L), extracted with EtOAc (500 mL × 3). The combined organic layer was washed with brine, dried over Na2SO4, concentrated in vacuo to give a crude product. The crude product was purified by re-crystallization from EtOAc (500 mL) at 25 o C. The filtrate was concentrated in vacuo to give a residue. The residue was dissolved EtOAc (300 mL), the solution was washed with 10% citric acid (100 mL × 3) and brine (150 mL), dried over Na 2 SO 4 , and concentrated under reduced pressure to give a residue. The residue was purified by re-crystallization from ethyl acetate/petroleum ether=1/1 (600 ml) at 25°C. The second recrystallized mother liquor was concentrated in vacuo to give residue. The residue was purified by flash silica gel chromatography (Eluent of 0~40% DCM/petroleum ether) to give title product (5g). Combined three batch products to afford tert-butyl 8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinoline-2-carboxylate (66.7 g, 162.88 mmol, 72.85% yield of two steps) as a light yellow solid. MS (ESI) m/z: 410.0 [M+H] + . 1 H NMR (400 MHz, CDCl3) δ = 7.87 - 7.79 (m, 1H), 7.51 (d, J = 7.4 Hz, 1H), 7.33 - 7.26 (m, 3H), 7.22 (d, J = 7.4 Hz, 1H), 7.17 - 7.10 (m, 1H), 4.91 (s, 2H), 3.64 (s, 2H), 2.88 (s, 2H), 1.57 - 1.34 (m, 9H) Step C. Procedure for preparation of N-(1,3-benzothiazol-2-yl)-1,2,3,4- tetrahydroisoquinoline-8-carboxamide To a mixture of tert-butyl 8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinoli ne- 2-carboxylate (61.7 g, 150.67 mmol, 1 equiv.) in DCM (1000 mL) was added HCl/dioxane (600 mL). The mixture was stirred at 25°C for 6 hours. TLC (petroleum ether: ethyl acetate = 3:1, R f = 0) showed the reaction was completed. The reaction mixture was concentrated in vacuo to give N-(1,3-benzothiazol-2-yl)-1,2,3,4-tetrahydroisoquinoline-8-c arboxamide (59 g, crude, 2HCl) as a white solid. The solid was used in the next step without further purification. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 9.63 (br s, 2H), 8.02 (d, J = 7.8 Hz, 1H), 7.87 - 7.61 (m, 2H), 7.55 - 7.22 (m, 4H), 4.47-4.36 (m, 2H), 3.56 (s, 3H), 3.43 - 3.28 (m, 2H), 3.11 (t, J = 5.8 Hz, 2H) Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-bromo-pyridine-2-carboxyl ate To a mixture of N-(1,3-benzothiazol-2-yl)-1,2,3,4-tetrahydroisoquinoline-8-c arboxamide (30 g, 73.76 mmol, 94% purity, 1 equiv., 2HCl) in DMA (300 mL) was added Cs 2 CO 3 (120.17 g, 368.81 mmol, 5 equiv.) and tert-butyl 3-bromo-6-chloro-pyridine-2-carboxylate (21.80 g, 74.50 mmol, 1.01 equiv.). The mixture was stirred at 120°C for 12 hours. Then tert-butyl 3- bromo-6-chloro-pyridine-2-carboxylate (5.8 g, 19.83 mmol, 2.69 e -1 equiv.) was added to the reaction mixture, and the resulting mixture was stirred at 120°C for 7 hours. The reaction mixture was cooled to room temperature, and poured onto ice water (1 L). The mixture was stirred for 10 min and filtered. The cake was washed with EtOH and collected. The cake was triturated with EtOH (200 mL) at 25 o C for 30 min to give tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-bromo-pyridi ne-2-carboxylate (24.6 g, 43.50 mmol, 58.98% yield) was obtained as a white solid. MS (ESI) m/z: 565.0 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.70 (brs, 1H), 8.01 (d, J = 7.8 Hz, 1H), 7.77 (dd, J = 2.8, 8.4 Hz, 2H), 7.58 (d, J = 7.4 Hz, 1H), 7.50 - 7.39 (m, 2H), 7.39 - 7.30 (m, 2H), 6.86 (d, J = 9.2 Hz, 1H), 4.93 (s, 2H), 3.77 (t, J = 6.0 Hz, 2H), 3.00 (t, J = 5.8 Hz, 2H), 1.34 (s, 9H) Step E. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3, 2-dioxaborolan-2-yl) pyridine-2-carboxylate tert-Butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-bromo- pyridine-2-carboxylate (1.5 g, 2.65 mmol, 1 equiv.), 4,4,5,5-tetramethyl-1,3,2-dioxaborolane (746.84 mg, 5.84 mmol, 846.76 µL, 2.2 equiv.), TEA (836.05 mg, 8.26 mmol, 1.15 mL, 3.11 equiv.), and Pd(dppf)Cl 2 .CH 2 Cl 2 (108.31 mg, 132.63 µmol, 0.05 equiv.) were taken up into a microwave tube in CH3CN (15 mL) and THF (5 mL). The sealed tube was heated at 100°C for 30 min under microwave. The reaction mixture was poured into water (200 mL) and extracted with ethyl acetate (150 mL × 2). The combined organic phase was concentrated in vacuo to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=6/1 to 1/2) to afford title product. The compound tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl) pyridine-2-carboxylate (9.2 g, 14.27 mmol, 55.0% yield, 95% purity) was obtained as a white solid. MS (ESI) m/z: 613.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.84 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.2 Hz, 1H), 7.68 (d, J = 8.6 Hz, 1H), 7.56 (d, J = 7.2 Hz, 1H), 7.50 - 7.39 (m, 2H), 7.38 - 7.30 (m, 2H), 6.86 (d, J = 8.8 Hz, 1H), 5.01 (s, 2H), 3.82 (t, J = 5.8 Hz, 2H), 3.00 (t, J = 5.8 Hz, 2H), 1.28 (s, 9H), 1.22 (s, 12H) Step F. Procedure for preparation of tert-butyl 4-[2-(3-bromo-2-methyl-phenoxy)ethyl] piperidine-1-carboxylate A mixture of tert-butyl 4-(2-hydroxyethyl)piperidine-1-carboxylate (5 g, 21.80 mmol, 1 equiv.), 3-bromo-2-methyl-phenol (4.08 g, 21.80 mmol, 1 equiv.) and PPh 3 (7.43 g, 28.35 mmol, 1.3 equiv.) in THF (40 mL) was degassed and purged with N2 for three times, and then the DIAD (5.29 g, 26.16 mmol, 5.09 mL, 1.2 equiv.) was added dropwise at 0 °C. The mixture was stirred at 25 °C for 12 hours under N 2 atmosphere. The mixture solution diluted with H 2 O (100 mL) and extracted with EtOAC 180 mL (60 mL × 3). The combined organic layers were washed with H2O 180 mL (60 mL × 3), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (petroleum ether/ethyl acetate = 100/1 to 5/1). The compound tert-butyl 4-[2-(3-bromo-2- methyl-phenoxy)ethyl] piperidine-1-carboxylate (8.2 g, 18.5 mmol, 84.9% yield, 90% purity) was obtained as a brown oil. MS (ESI) m/z: 344.0 [M-56] + Step G. Procedure for preparation of 4-[2-(3-bromo-2-methyl-phenoxy)ethyl]piperidine To a solution of tert-butyl 4-[2-(3-bromo-2-methyl-phenoxy)ethyl]piperidine-1-carboxylat e (8.2 g, 20.59 mmol, 1 equiv.) was added HCl/dioxane (100 mL). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The compound 4-[2-(3-bromo-2-methyl-phenoxy)ethyl]piperidine (6 g, crude) was obtained as a white solid. Step H. Procedure for preparation of ethyl 2-[4-[2-(3-bromo-2-methyl-phenoxy)ethyl]-1- piperidyl]acetate To a solution of 4-[2-(3-bromo-2-methyl-phenoxy)ethyl]piperidine (3 g, 10.06 mmol, 1 equiv.) in CH3CN (30 mL) was added ethyl 2-bromoacetate (2.02 g, 12.07 mmol, 1.34 mL, 1.2 equiv.) and TEA (3.05 g, 30.18 mmol, 4.20 mL, 3 equiv.). The mixture was stirred at 20 °C for 12 hours. The mixture solution was diluted with H 2 O (30 mL) and extracted with ethyl acetate (60 mL × 3). The combined organic layers were washed with H2O (10mL × 3), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (petroleum ether/ethyl acetate = 100/1 to 5/1). The compound ethyl 2-[4-[2-(3-bromo-2-methyl-phenoxy)ethyl]-1-piperidyl]acetate (3 g, 7.7 mmol, 76.8% yield, 99% purity) was obtained as a white liquid. MS (ESI) m/z: 385.8 [M+H] + ( 80 Br) Step I. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[1-(2-ethoxy-2-oxo-ethy l)-4-piperidyl]ethoxy]-2- methyl-phenyl]pyridine-2-carboxylate To a solution of ethyl 2-[4-[2-(3-bromo-2-methyl-phenoxy)ethyl]-1-piperidyl]acetate (700 mg, 1.82 mmol, 1 equiv.) in 1,4-dioxane (8 mL) was added ditert- butyl(cyclopentyl)phosphane;dichloropalladium;iron (237.43 mg, 364.29 µmol, 0.2 equiv.), KF (1.5 M, 1.82 mL, 1.5 equiv.) and tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl) pyridine-2- carboxylate (1.23 g, 2.00 mmol, 1.1 equiv.). After addition, the mixture was stirred at 90 °C for 2 hours. The mixture solution diluted with H 2 O (30 mL) and extracted with ethyl acetate (60 mL × 3). The combined organic layers were washed with H2O (10 mL × 3), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (petroleum ether/ethyl acetate = 10/1 to 1/1) to give tert- butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[2-[1-(2- ethoxy-2-oxo-ethyl)-4-piperidyl]ethoxy]-2-methyl-phenyl]pyri dine-2-carboxylate (600 mg, 721.5 µmol, 39.6% yield, 95% purity) as a brown solid. MS (ESI) m/z: 790.5 [M+H] + Step J. Procedure for preparation of 2-[4-[2-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyr idyl]-2-methyl- phenoxy]ethyl]-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[2-[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]ethoxy]- 2-methyl-phenyl]pyridine-2- carboxylate (600 mg, 759.51 µmol, 1 equiv.) in THF (3 mL) was added LiOH•H2O (95.62 mg, 2.28 mmol, 3 equiv.) and H2O (1 mL). The mixture was stirred at 20 °C for 1 hour. The pH was adjusted to 5 with citric acid. The mixture solution diluted with H 2 O (10 mL) and extracted with DCM (10 mL × 3). The combined organic layers were washed with H 2 O (10 mL × 3), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The compound 2-[4-[2-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H-isoquinolin-2- yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]ethyl] -1-piperidyl]acetic acid (410 mg, crude) was obtained as a yellow solid. MS (ESI) m/z: 762.3 [M+H] + Step K. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[1-[2-[[3-(2,6-diox o-3-piperidyl)-1-methyl- indazol-7-yl]amino]-2-oxo-ethyl]-4-piperidyl]ethoxy]-2-methy l-phenyl]pyridine-2- carboxylate To a solution of 2-[4-[2-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]ethyl]-1- piperidyl]acetic acid (110 mg, 144.37 µmol, 1 equiv.) and 3-(7-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (48.47 mg, 187.68 µmol, 1.3 equiv.) in DMF (1 mL) was added HATU (71.36 mg, 187.68 µmol, 1.3 equiv.) and DIEA (55.98 mg, 433.11 µmol, 75.44 µL, 3 equiv.). The mixture was stirred at 40 °C for 2 hours. The reaction mixture was treated with H2O (10 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a crude tert- butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[2-[1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-o xo-ethyl]-4-piperidyl]ethoxy]- 2-methyl-phenyl]pyridine-2-carboxylate (120 mg, crude) as a white solid. MS (ESI) m/z: 1002.7 [M+H] + Step L. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[2-[1-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-7- yl]amino]-2-oxo-ethyl]-4-piperidyl]ethoxy]-2-methyl-phenyl]p yridine-2-carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[2-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-ind azol-7-yl]amino]-2-oxo-ethyl]-4- piperidyl]ethoxy]-2-methyl-phenyl]pyridine-2-carboxylate (70 mg, 69.85 µmol, 1 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 193.37 equiv.). The mixture was stirred at 20 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[2-[1-[2-[[3-(2,6-dioxo- 3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-oxo-ethyl]-4-pip eridyl]ethoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid (23.1 mg, 24.4 µmol, 35% yield, 99% purity) as a white solid. MS (ESI) m/z: 946.5 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 10.9 (s, 1H), 9.88 (s, 1H), 8.08 - 8.00 (m, 1H), 7.84 - 7.76 (m, 1H), 7.64 - 7.56 (m, 2H), 7.52 - 7.28 (m, 7H), 7.12 - 7.04 (m, 2H), 6.96 - 6.88 (m, 2H), 6.60 (d, J = 1.6 Hz, 1H), 4.98 (s, 2H), 4.40 - 4.32 (m, 1H), 4.12 - 4.08 (m, 3H), 4.04 - 3.96 (m, 2H), 3.92 - 3.88 (m, 2H), 3.16 - 3.08 (m, 2H), 3.04 - 3.00 (m, 2H), 2.96 - 2.92 (m, 2H), 2.72 - 2.60 (m, 1H), 2.36 - 2.32 (m, 1H), 2.24 - 2.12 (m, 4H), 1.92 (s, 3H), 1.76 - 1.68 (m, 4H), 1.56 - 1.48 (m, 1H), 1.44 - 1.36 (m, 2H) Example 118. Preparation of Compound 102 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[3-[4-(2,6- dioxo-3-piperidyl)anilino]-3-oxo-propyl]phenoxy]-2-methyl-ph enyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 2,6-dibenzyloxy-3-(4-nitrophenyl)pyridine To a solution of (2, 6-dibenzyloxy-3-pyridyl)boronic acid (4 g, 11.93 mmol, 1 equiv.) ,1- bromo-4-nitro-benzene (2.65 g, 13.13 mmol, 1.1 equiv.) in 1,4-dioxane (40 mL) and H 2 O (4 mL) was added Pd(dppf)Cl 2 (873.26 mg, 1.19 mmol, 0.1 equiv.) and Na 2 CO 3 (3.79 g, 35.80 mmol, 3 equiv.). The mixture was stirred at 100 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate = 100/1 to 50/1) to give 2,6- dibenzyloxy-3-(4-nitrophenyl)pyridine (4.6 g, 11.1 mmol, 93.4% yield) as a yellow solid. 1 H NMR (400 MHz, CDCl3) δ = 8.15 (d, J = 8.8 Hz, 2H), 7.65 (d, J = 8.8 Hz, 2H), 7.58 (d, J = 8.0 Hz, 1H), 7.39 - 7.21 (m, 10H), 6.46 (d, J = 8.0 Hz, 1H), 5.37 (s, 2H), 5.33 (s, 2H) Step B. Procedure for preparation of 3-(4-aminophenyl)piperidine-2,6-dione To a solution of 2,6-dibenzyloxy-3-(4-nitrophenyl)pyridine (4.6 g, 11.15 mmol, 1 equiv.) in THF (40 mL) and EtOH (30 mL) was added Pd(OH)2 (1.57 g, 1.12 mmol, 10% purity, 0.1 equiv.) and Pd/C (1.17 g, 1.12 mmol, 10% purity, 0.1 equiv.) under N2 atmosphere. The suspension was degassed and purged with H 2 three times. The mixture was stirred under H 2 (50 Psi or atm.) at 50 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, dichloromethane: methanol = 100/1 to 20/1) to give 3-(4-aminophenyl)piperidine-2,6-dione (900 mg, 4.4 mmol, 39.5% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ = 10.79 - 10.68 (m, 1H), 6.92 - 6.78 (m, 2H), 6.59 - 6.45 (m, 2H), 4.98 (b s, 2H), 3.66 - 3.52 (m, 1H), 2.65 - 2.55 (m, 2H), 2.12 - 1.94 (m, 2H) Step C. Procedure for preparation of methyl 3-[4-(3-bromo-2-methyl-phenoxy) phenyl]propanoate To a solution of methyl 3-(4-hydroxyphenyl) propanoate (1 g, 5.55 mmol, 1 equiv.) in NMP (10 mL) was added 1, 3-dibromo-2-methyl-benzene (4.16 g, 16.65 mmol, 3 equiv.), CuI (528.44 mg, 2.77 mmol, 0.5 equiv.), Cs2CO3 (2.17 g, 6.66 mmol, 1.2 equiv.), and dipivaloylmethane (255.65 mg, 1.39 mmol, 285.65 μL, 0.25 equiv.) at 25 °C. The reaction mixture was stirred at 120 °C for 16 hours under N 2 . The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (30 mL × 3). The combined organic layers were washed with brine (20 mL × 3), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~10% ethyl acetate/petroleum ether). The compound methyl 3-[4-(3-bromo-2-methyl- phenoxy) phenyl]propanoate (1.68 g, 4.33 mmol, 78.02% yield, 90% purity) was obtained as a yellow oil. MS (ESI) m/z: 350.7 [M+H] + 1 H NMR (400 MHz, CD3OD) δ = 7.35 (d, J = 7.6 Hz, 1H), 7.21 - 7.16 (m, 2H), 7.06 (t, J = 8.0 Hz, 1H), 6.85 - 6.77 (m, 3H), 3.67 - 3.61 (m, 3H), 2.89 (t, J = 7.6 Hz, 2H), 2.65 - 2.59 (m, 2H), 2.30 (s, 3H) Step D. Procedure for preparation of tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) - 3, 4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-(3-methoxy-3-oxo-prop yl) phenoxy]-2-methyl- phenyl]pyridine-2-carboxylate To a solution of methyl 3-[4-(3-bromo-2-methyl-phenoxy) phenyl]propanoate (1 g, 2.86 mmol, 1 equiv.) in dioxane (10 mL) was added tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-3-(4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridine- 2-carboxylate (1.93 g, 3.15 mmol, 1.1 equiv.), Ad 2 nBuP Pd G 3 (cataCXium® A Pd G 3 ) (417.09 mg, 572.71 μmol, 0.2 equiv.), and K3PO4 (1.5 M, 5.73 mL, 3 equiv.) at 25 °C. The reaction mixture was purged with N23 time and stirred under N2 at 80 °C for 16 hours. The reaction mixture was diluted with water (10 mL) and extracted with EtOAc (20 mL × 3). The combined organic layers were washed with brine (20 mL × 1), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~50% ethyl acetate/petroleum ether). The compound tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-(3- methoxy-3-oxo-propyl) phenoxy]-2-methyl-phenyl]pyridine-2-carboxylate (1 g, 1.30 mmol, 45.34% yield, 98% purity) was obtained as a yellow solid. MS (ESI) m/z: 755.3 [M+H] + . Step E. Procedure for preparation of 3-[4-[3-[6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyrid yl]-2-methyl- phenoxy]phenyl]propanoic acid To a solution of tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H- isoquinolin-2-yl]-3-[3-[4-(3-methoxy-3-oxo-propyl) phenoxy]-2-methyl-phenyl]pyridine-2- carboxylate (500 mg, 662.35 μmol, 1 eq) in THF (5 mL) was added LiOH.H 2 O (1 M, 1.99 mL, 3 eq) at 25 °C. The reaction mixture was stirred at 25 °C for 2 hours. The reaction mixture was acidified with 1N HCl to pH = 6, filtered, and concentrated under reduced pressure to give a residue. The compound 3-[4-[3-[6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]phenyl]propanoic acid (616 mg, crude) was obtained as a white solid. MS (ESI) m/z: 741.3 [M+H] + . Step F. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[4-(2,6-dioxo-3- piperidyl)anilino]-3-oxo- propyl]phenoxy]-2-methyl-phenyl]pyridine-2-carboxylate To a solution of 3-[4-[3-[6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]phenyl]propanoic acid (150 mg, 202.47 µmol, 1 equiv.) in DMF (1 mL) was added DIEA (78.50 mg, 607.40 µmol, 105.80 µL, 3 equiv.), HATU (92.38 mg, 242.96 µmol, 1.2 equiv.), and 3-(4- aminophenyl)piperidine-2,6-dione (45.48 mg, 222.71 µmol, 1.1 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was added slowly to water (10 mL). The resulting mixture was filtered, and the filtrate was concentrated under reduced pressure to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4- [3-[4-(2,6-dioxo-3-piperidyl)anilino]-3-oxo-propyl]phenoxy]- 2-methyl-phenyl]pyridine-2- carboxylate (100 mg, 107.9 µmol, 53.3% yield) as a white solid was used into the next step without further purification. MS (ESI) m/z: 927.4 [M+H] + . Step G. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[4-[3-[4-(2,6-dioxo-3-piperidyl)an ilino]-3-oxo- propyl]phenoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[4-[3-[4-(2,6-dioxo-3-piperidyl)anilino]-3-oxo-pr opyl]phenoxy]-2-methyl- phenyl]pyridine-2-carboxylate (90 mg, 97.08 µmol, 1 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 139.13 equiv.). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[4-[3-[4-(2,6-dioxo-3-piperidyl)anili no]-3-oxo-propyl]phenoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid (29.2 mg, 31.7 µmol, 32.6% yield, 94.4% purity) as a yellow solid.. MS (ESI) m/z: 871.3 [M+H] + . 1 H NMR (400 MHz, DMSO) δ = 12.92 - 12.77 (m, 1H), 10.84 - 10.77 (m, 1H), 9.89 (s, 1H), 8.04 (d, J = 8.0 Hz, 1H), 7.83 - 7.76 (m, 1H), 7.63 (d, J = 7.6 Hz, 1H), 7.55 - 7.50 (m, 3H), 7.48 - 7.44 (m, 2H), 7.40 - 7.33 (m, 2H), 7.25 - 7.09 (m, 6H), 7.03 - 6.99 (m, 1H), 6.88 (d, J = 7.6 Hz, 1H), 6.85 - 6.79 (m, 3H), 5.00 (s, 2H), 3.94 (t, J = 5.6 Hz, 2H), 3.81 - 3.77 (m, 1H), 3.04 (t, J = 5.6 Hz, 2H), 2.87 (d, J = 7.6 Hz, 2H), 2.60 (s, 2H), 2.21 - 2.09 (m, 2H), 2.06 - 1.96 (m, 2H), 1.90 (s, 3H) Example 119. Preparation of Compound 103 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[2-[4-[2-[[3- (2,6-dioxo-3-piperidyl) -1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]piperazin-1-yl]eth oxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 1-bromo-3-(2-bromoethoxy)-2-methyl-benzene To a solution of 1,2-dibromoethane (36.16 g, 192.48 mmol, 14.52 mL, 8 equiv.) in CH 3 CN (40 mL) was added Cs2CO3 (15.68 g, 48.12 mmol, 2 equiv.) and 3-bromo-2-methyl-phenol (4.5 g, 24.06 mmol, 1 equiv.). The mixture was stirred at 80 °C for 15 hours. The solution was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 50/1 to 20/1) to obtain 1-bromo-3-(2- bromoethoxy)-2-methyl-benzene (4.5 g, 15.3 mmol, 63.6% yield) as a yellow oil. 1 H NMR (400 MHz, CDCl 3 ) δ = 7.20 (d, J = 8.0 Hz, 1H), 7.01 (t, J = 8.0 Hz, 1H), 6.76 (d, J = 8.0 Hz, 1H), 4.29 (t, J = 6.0 Hz, 2H), 3.67 (t, J = 6.0 Hz, 2H), 2.36 (s, 3H). Step B. Procedure for preparation of ethyl 2-[4-[2-(3-bromo-2-methyl- phenoxy)ethyl]piperazin-1-yl]acetate To a solution of ethyl 2-piperazin-1-ylacetate (585.83 mg, 3.40 mmol, 1.00 equiv.) in DMF (10 mL) was added DIPEA (1.54 g, 11.91 mmol, 2.07 mL, 3.5 equiv.) and 1-bromo-3-(2- bromoethoxy)-2-methyl-benzene (1 g, 3.40 mmol, 1 equiv.). The mixture was stirred at 60 °C for 15 hours. The reaction mixture was partitioned between water (10 mL) and ethyl acetate (30 mL). The organic phase was separated and washed with brine (10 mL × 2), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 100/1 to 1/100). The compound ethyl 2-[4-[2-(3-bromo-2-methyl-phenoxy)ethyl]piperazin-1-yl]aceta te (2.6 g, 6.49 mmol, 95.42% yield, 96.2% purity) was obtained as a yellow oil. MS (ESI) m/z: 385.1 [M+H] + 1 H NMR (400 MHz, CDCl3) δ = 7.15 (d, J = 8.0 Hz, 1H), 6.99 (t, J = 8.0 Hz, 1H), 6.76 (d, J = 8.4 Hz, 1H), 4.19 (q, J = 7.2 Hz, 2H), 4.12 - 4.08 (m, 2H), 3.21 (s, 2H), 2.86 (t, J = 5.6 Hz, 2H), 2.72 - 2.61 (m, 8H), 2.30 (s, 3H), 1.29 - 1.26 (m, 3H). Step C. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[4-(2-ethoxy-2-oxo- ethyl)piperazin-1- yl]ethoxy]-2-methyl-phenyl]pyridine-2-carboxylate A mixture of ethyl 2-[4-[2-(3-bromo-2-methyl-phenoxy)ethyl]piperazin-1-yl]aceta te (1 g, 2.60 mmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (1.27 g, 2.08 mmol, 0.8 equiv.) , K2CO3 (1.5 M, 2.60 mL, 1.5 equiv.), and Ad2nBuP Pd G 3 (cataCXium® A Pd G 3 ) (378.03 mg, 519.08 µmol, 0.2 equiv.) in 1,4-dioxane (10 mL) was degassed and purged with N 2 three times. Then the mixture was stirred at 80 °C for 6 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , DCM/MeOH = 8:1). The compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[2-[4-(2-ethoxy-2-oxo-ethyl)piperazin-1-yl]ethoxy]- 2-methyl-phenyl]pyridine-2- carboxylate (1.3 g, 1.6 mmol, 63.3% yield) was obtained as a yellow oil. MS (ESI) m/z: 791.3 [M+H] + Step D. Procedure for preparation of 2-[4-[2-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyr idyl]-2-methyl- phenoxy]ethyl]piperazin-1-yl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[2-[4-(2-ethoxy-2-oxo-ethyl)piperazin-1-yl]ethoxy ]-2-methyl-phenyl]pyridine-2- carboxylate (1.3 g, 1.64 mmol, 1 equiv.) in THF (2 mL) was added LiOH.H 2 O (1 M, 4.93 mL, 3 equiv.). Then, the mixture was stirred at 25 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by reverse- phase HPLC. The compound 2-[4-[2-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o- 1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-meth yl-phenoxy]ethyl]piperazin-1- yl]acetic acid (250 mg, 327.6 µmol, 19.9% yield) was obtained as a white solid. MS (ESI) m/z: 763.3 [M+H] + Step E. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[4-[2-[[3-(2,6-diox o-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]piperazin-1-yl]ethoxy]-2-met hyl-phenyl]pyridine-2- carboxylate To a solution of 2-[4-[2-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]ethyl]piperazin-1- yl]acetic acid (140 mg, 183.51 umol, 1 equiv.) and 3-(6-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (56.87 mg, 220.21 umol, 1.2 equiv.) in DMF (0.1 mL) was added DIPEA (71.15 mg, 550.52 µmol, 95.89 µL, 3 equiv.) and HATU (83.73 mg, 220.21 umol, 1.2 equiv.). Then, the mixture was stirred at 25 °C for 15 hours. The reaction mixture was added to water (2 mL) slowly, and a grey solid precipitated. The combined solution was filtered, dissolved in DCM (10 mL), and concentrated under reduced pressure to give a residue. The compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[2-[4-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indaz ol-6-yl]amino]-2-oxo- ethyl]piperazin-1-yl]ethoxy]-2-methyl-phenyl]pyridine-2-carb oxylate (220 mg, crude) was obtained as a brown solid. MS (ESI) m/z: 1003.5 [M+H] + Step F. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[2-[4-[2-[[3-(2,6-dioxo-3-piperidy l) -1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]piperazin-1-yl]ethoxy]-2-methyl-phenyl ]pyridine-2-carboxylic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[2-[4-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indaz ol-6-yl]amino]-2-oxo- ethyl]piperazin-1-yl]ethoxy]-2-methyl-phenyl]pyridine-2-carb oxylate (113.46 mg, 113.10 umol, 1 equiv.) and TFA (12.90 mg, 113.10 umol, 8.37 µL, 1 equiv.) in DCM (1 mL) was stirred at 25 °C for 4 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC. The compound 6-[8-(1,3-benzothiazol- 2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[4-[ 2-[[3-(2,6-dioxo-3-piperidyl) - 1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]piperazin-1-yl]etho xy]-2-methyl-phenyl]pyridine- 2-carboxylic acid (53 mg, 56.0 µmol, 49.5% yield) was obtained as a yellow solid. MS (ESI) m/z: 947.3 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 12.99 - 12.68 (m, 1H), 10.81 (s, 1H), 9.85 - 9.73 (m, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 7.2 Hz, 1H), 7.55 (d, J = 8.4 Hz, 2H), 7.49 - 7.39 (m, 4H), 7.38 - 7.33 (m, 2H), 7.16 (d, J = 7.6 Hz, 3H), 6.98 (d, J = 8.88.9 Hz, 2H), 6.69 (d, J = 5.2 Hz, 1H), 4.98 (s, 2H), 4.44 - 4.23 (m, 2H), 3.92 (t, J = 6.0 Hz, 2H), 3.80 (dd, J = 4.4, 11.2 Hz, 1H), 3.62 - 3.54 (m, 2H), 3.28 (s, 2H), 3.04 - 3.02 (m, 2H), 2.72 - 2.61 (m, 4H), 2.54 (s, 4H), 2.24 - 2.10 (m, 2H), 2.09 - 1.96 (m, 2H), 1.94 (s, 3H) Example 120. Preparation of Compound 104 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[4-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]piperazin-1-yl]propoxy]- 2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 1-bromo-3-(3-bromopropoxy)-2-methyl-benzene A mixture of 3-bromo-2-methyl-phenol (2 g, 10.69 mmol, 1 equiv.), 1,3-dibromopropane (10.79 g, 53.47 mmol, 5.45 mL, 5 equiv.) and Cs2CO3 (10.45 g, 32.08 mmol, 3 equiv.) in CH 3 CN (25 mL) was stirred at 25 °C for 5 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~1% ethyl acetate/petroleum ether). The compound 1-bromo- 3-(3-bromopropoxy)-2-methyl-benzene (2.4 g, 7.0 mmol, 65.8% yield, 90% purity) was obtained as a white solid. Step B. Procedure for preparation of ethyl 2-[4-[3-(3-bromo-2-methyl- phenoxy)propyl]piperazin-1-yl]acetate A mixture of 1-bromo-3-(3-bromopropoxy)-2-methyl-benzene (2.15 g, 6.97 mmol, 1 equiv.), ethyl 2-piperazin-1-ylacetate (1.2 g, 6.97 mmol, 1 equiv.) and DIPEA (3.60 g, 27.87 mmol, 4.85 mL, 4 equiv.) in DMF (25 mL) was stirred at 60 °C for 3 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~10% ethyl acetate/petroleum ether) to give ethyl 2-[4-[3-(3-bromo-2-methyl-phenoxy)propyl]piperazin-1-yl]acet ate (1.3 g, 3.1 mmol, 46.0% yield, 95% purity) as a white solid. MS (ESI) m/z: 401.1 [M+H] + ( 80 Br). Step C. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[4-(2-ethoxy-2-oxo- ethyl)piperazin-1- yl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate A mixture of ethyl 2-[4-[3-(3-bromo-2-methyl-phenoxy)propyl]piperazin-1-yl]acet ate (380 mg, 951.61 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (816.07 mg, 1.33 mmol, 1.4 equiv.), [2-(2-aminophenyl)phenyl]palladium(1+);bis(1- adamantyl)-butyl-phosphane;methanesulfonate (138.61 mg, 190.32 µmol, 0.2 equiv.), and K 2 CO 3 (1.5 M, 951.61 µL, 1.5 equiv.) in dioxane (3 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 100 °C for 1 hour under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~10% DCM/ MeOH) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3- [3-[4-(2-ethoxy-2-oxo-ethyl)piperazin-1-yl]propoxy]-2-methyl -phenyl]pyridine-2- carboxylate (546 mg, 610.44 µmol, 64.15% yield, 90% purity) as a white solid. MS (ESI) m/z: 805.5 [M+H] + . 1 H NMR (400 MHz, CD3OD) δ = 7.90 (d, J = 8.0 Hz, 1H), 7.75 (d, J = 8.0 Hz, 1H), 7.58 (d, J = 7.6 Hz, 1H), 7.42 (d, J = 8.4 Hz, 1H), 7.37 - 7.34 (m, 1H), 7.14 - 7.06 (m, 2H), 7.02 (t, J = 8.0 Hz, 1H), 6.94 (d, J = 8.8 Hz, 1H), 6.89 (dd, J = 4.4, 8.0 Hz, 2H), 6.63 (d, J = 7.6 Hz, 1H), 5.02 (s, 2H), 4.17 (dq, J = 2.8, 7.2 Hz, 4H), 3.98 (t, J = 6.0 Hz, 2H), 3.25 (d, J = 3.6 Hz, 3H), 3.08 (t, J = 6.0 Hz, 2H), 2.67 - 2.63 (m, 10H), 2.28 (s, 2H), 1.93 (s, 3H), 1.80 (s, 2H), 1.08 (s, 9H) Step D. Procedure for preparation of 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyr idyl]-2-methyl- phenoxy]propyl]piperazin-1-yl]acetic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[4-(2-ethoxy-2-oxo-ethyl)piperazin-1-yl]propoxy] -2-methyl-phenyl]pyridine-2- carboxylate (230 mg, 285.72 µmol, 1 equiv.) and LiOH•H2O (35.97 mg, 857.15 µmol, 3 equiv.) in THF (1.2 mL) and H2O (0.4 mL) was stirred at 25 °C for 1.5 hour. The reaction mixture was concentrated and redissolved by H2O (2 mL), then 1M HCl was added to adjust the pH to 2, the residue was filtered and dried. The residue was used for next step without further purification. The compound 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o- 1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-meth yl-phenoxy]propyl]piperazin- 1-yl]acetic acid (206 mg, 225.3 µmol, 78.8% yield) was obtained as a white solid. Step E. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[4-[2-[[3-(2,6-diox o-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]piperazin-1-yl]propoxy]-2-me thyl-phenyl]pyridine-2- carboxylate A mixture of 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H-isoquinolin- 2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]prop yl]piperazin-1-yl]acetic acid (150 mg, 193.06 µmol, 1 equiv.), 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (59.84 mg, 231.68 µmol, 1.2 equiv.), HATU (73.41 mg, 193.06 µmol, 1 equiv.), and DIPEA (74.86 mg, 579.19 µmol, 100.88 µL, 3 equiv.) in DMF (2 mL) was stirred at 25°C for 10 hours. The reaction mixture was quenched by addition H 2 O (2 mL), and then extracted with ethyl acetate 6 mL (2 mL * 3). The combined organic layers were washed with saturated sodium chloride (3 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The reaction mixture was used for next step without other purification. The compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[4-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]piperazin-1-yl]propoxy]-2-methyl-pheny l]pyridine-2-carboxylate (150 mg, 125.3 µmol, 64.9% yield) was obtained as a white solid. MS (ESI) m/z: 1017.6 [M+H] + . Step F. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[4-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]piperazin-1-yl]propoxy]-2-methyl-pheny l]pyridine-2-carboxylic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[4-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indaz ol-6-yl]amino]-2-oxo- ethyl]piperazin-1-yl]propoxy]-2-methyl-phenyl]pyridine-2-car boxylate (120 mg, 117.97 µmol, 1 equiv.) and TFA (1.54 g, 13.51 mmol, 1 mL, 114.49 equiv.) in DCM (1 mL) was stirred at 40 °C for 3 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[4-[2- [[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]amino]-2-oxo-ethyl]piperazin-1-yl]propox y]-2-methyl-phenyl]pyridine- 2-carboxylic acid (42.32 mg, 43.3 µmol, 36.7% yield, 98.3% purity) was obtained as a white solid. MS (ESI) m/z: 961.2 [M+H] + . 1 H NMR (400 MHz, d 6 -DMSO) δ = 10.88 (s, 1H), 9.85 (s, 1H), 8.15 (s, 1H), 8.07 – 8.00 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 8.8 Hz, 2H), 7.50 – 7.41 (m, 3H), 7.40 – 7.31 (m, 2H), 7.20 (dd, J = 8.8, 1.6 Hz, 1H), 7.13 – 7.06 (m, 1H), 6.96 (d, J = 8.8 Hz, 1H), 6.88 (d, J = 8.4 Hz, 1H), 6.64 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.32 (dd, J = 9.6, 5.2 Hz, 1H), 4.00 (t, J = 6.0 Hz, 2H), 3.92 (s, 5H), 3.16 (s, 3H), 3.02 (t, J = 5.6 Hz, 2H), 2.65 – 2.52 (m, 10H), 2.41 – 2.28 (m, 2H), 2.21 – 2.14 (m, 1H), 1.95 – 1.88 (m, 5H). Example 121. Preparation of Compound 105 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[4-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-oxo-e thyl]piperazin-1-yl]propoxy]- 2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 7-bromo-3-iodo-1-methyl-1H-indazole To a solution of compound 7-bromo-3-iodo-1H-indazole (298 g, 849 mmol, 1.00 eq) in DMF (1.79 L) was added Cs 2 CO 3 (415 g, 1.27 mol, 1.50 eq) at 0 °C, then the suspension was stirred at 0 °C for 1 h. Then CH3I (181 g, 1.27 mol, 79 mL, 1.50 eq) was added dropwise to the reaction at 0 °C, after addition, the suspension was stirred at 25 °C for 2 h. The reaction suspension was poured into water (8.4 L) and stirred for 10 min. The aqueous phase was extracted with ethyl acetate (800 mL, 400 mL). The combined organic phase was washed with brine (400 mL), dried with anhydrous Na2SO4, filtered, and concentrated in vacuum. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=100/1 to 0/1) to give compound 7-bromo-3-iodo-1-methyl-1H-indazole (160 g, 52.3% yield) as an off-white solid. MS (ESI) m/z: 336.8 [M+H] + 1 H NMR: (400 MHz, DMSO-d 6 ) δ 7.73 (d, J = 6.4 Hz, 1H), 7.46 (d, J = 8.0 Hz, 1H), 7.12 (t, J = 8.0 Hz, 1H), 4.34 (s, 1H). Step B. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-7-bromo-1- methyl-1H-indazole To a solution of compound 7-bromo-3-iodo-1-methyl-1H-indazole (140 g, 415 mmol, 1.00 eq) in dioxane (980 mL) and H2O (490 mL) was added compound (2,6-bis(benzyloxy)pyridin-3- yl)boronic acid (139 g, 415 mmol, 1.00 eq), K 3 PO 4 (220 g, 1.04 mol, 2.50 eq) and Pd(PPh 3 ) 4 (14.4 g, 12.5 mmol, 0.03 eq). Then the suspension was purged with N 2 for three times and stirred at 90 °C for 24 h. The reaction was cooled to 20 °C and filtered by a pad of celite. The filtrate was poured into water (900 mL) and the aqueous phase was extracted with ethyl acetate (900 mL, 450 mL). The combined organic phase was washed with brine (400 mL), dried with anhydrous Na2SO4, filtered, and concentrated in vacuum. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=100/1 to 0/1) to give compound 3-(2,6- bis(benzyloxy)pyridin-3-yl)-7-bromo-1-methyl-1H-indazole (120 g, 240 mmol, 57.7% yield) as an off-white solid. MS (ESI) m/z: 500.1 [M+H] + 1 H NMR: (400 MHz, DMSO-d6) δ 7.86 (d, J = 8.0 Hz, 1H), 7.46 - 7.59 (m, , 2H), 7.35 - 7.39 (m, 9H), 7.27 - 7.33 (m, 7H), 6.93 (t, J = 8.0 Hz, 1H), 6.60 (d, J = 8.0 Hz, 1H), 5.43 (d, J = 1.2 Hz, 4H), 4.35 (s, 3H). Step C. Procedure for preparation of N-benzyl-3-(2,6-bis(benzyloxy)pyridin-3-yl)-1- methyl-1H-indazol-7-amine To a solution of compound 3-(2,6-bis(benzyloxy)pyridin-3-yl)-7-bromo-1-methyl-1H- indazole (120 g, 240 mmol, 1.00 eq), phenylmethanamine (38.6 g, 360 mmol, 39.2 mL, 1.50 eq), Cs 2 CO 3 (156 g, 479 mmol, 2.00 eq), BINAP (10.5 g, 16.8 mmol, 0.07 eq) and Pd 2 (dba) 3 (4.39 g, 4.80 mmol, 0.02 eq) in dioxane (1.2 L) was degassed and purged with N2 three times, and then the mixture was stirred at 110 °C for 16 h under N2 atmosphere. The reaction suspension was filtered by a pad of celite and the filter cake was washed with THF (300 mL). The filtrate was concentrated under vacuum. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=100/1 to 0/1). The crude was triturated with EtOH (250 mL) at 20 °C for 1 h. The solid was collected by filtration and dried under vacuum to give compound N-benzyl-3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H- indazol-7-amine (110 g, 209 mmol, 87.1% yield) as a yellow solid. MS (ESI) m/z: 527.2 [M+H] + 1 H NMR: (400 MHz, DMSO-d 6 ) δ 7.81 (d, J = 8.0 Hz, 1H), 7.34 - 7.46 (m, 4H), 7.26 - 7.33 (m, 12H), 6.89 (d, J = 8.0 Hz, 1H)), 6.74 (t, J = 7.2 Hz, 1H), 6.57 (d, J = 8.0 Hz, 1H), 6.30 (d, J = 7.2 Hz, 1H), 6.09 (t, J = 8.0 Hz, 1H), 5.40 (s, 4H), 4.40 (s, 5H). Step D. Procedure for preparation of 3-(7-amino-1-methyl-1H-indazol-3-yl)piperidine- 2,6-dione To a solution of N-benzyl-3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-inda zol-7-amine (60 g, 114 mmol, 1.00 eq) and AcOH (6.84 g, 114 mmol, 6.52 mL, 1.00 eq) in THF (300 mL) and EtOH (300 mL) was added Pd/C (15.0 g, 10% purity) and Pd(OH)2 (15.0 g, 21.4 mmol, 20% purity) then the suspension was stirred at 50 °C under 50 psi (H 2 ) for 12 h. The two reactions were combined for work-up. The mixture was filtered, and the filter cake was washed with THF (4.0 L). The filtrate was concentrated under reduced pressure to give a residue. The crude product was triturated with EtOAC/MTBE = 1/1 (150 mL) at 25 o C for 5 h. The solid was collected by filtration and dried under vacuum to give 3-(7-amino-1-methyl-1H-indazol- 3-yl)piperidine-2,6-dione (51.0 g, 83.9% yield) as a off-white solid. MS (ESI) m/z: 259.2 [M+H] + 1 H NMR: (400 MHz, DMSO-d6) δ 10.85 (s, 1H), 6.91 (d, J = 8.0 Hz, 1H), 6.80 (t, J = 7.6 Hz, 1H), 6.55 (dd, J = 7.2 Hz, 1H), 5.18 (s, 2H), 4.24 (dd, J = 4.4, 5.2 Hz, 1H), 4.20 (s, 3H),3.58 - 3.61 (m, 1H), 2.58 - 2.64 (m, 2H), 2.26 - 2.29 (m, 1H), 2.15 - 2.16 (m, 1H) Step E. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[4-[2-[[3-(2,6-diox o-3-piperidyl)-1-methyl- indazol-7-yl]amino]-2-oxo-ethyl]piperazin-1-yl]propoxy]-2-me thyl-phenyl]pyridine-2- carboxylate A mixture of 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H-isoquinolin- 2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]prop yl]piperazin-1-yl]acetic acid (200 mg, 257.42 µmol, 1 equiv.), 3-(7-amino-1-methyl-1H-indazol-3-yl)piperidine-2,6-dione (73.13 mg, 283.16 µmol, 1.1 equiv.), HATU (97.88 mg, 257.42 µmol, 1 equiv.), and DIPEA (99.81 mg, 772.26 µmol, 134.51 µL, 3 equiv.) in DMF (2 mL) was stirred at 25 °C for 10 hours. The reaction mixture was quenched by addition of H2O (2 mL), and then extracted with ethyl acetate 6 mL (2 mL × 3). The combined organic layers were washed with saturated sodium chloride (3 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was used for next step without other purification. The compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[4-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indaz ol-7-yl]amino]-2-oxo- ethyl]piperazin-1-yl]propoxy]-2-methyl-phenyl]pyridine-2-car boxylate (130 mg, 108.6 µmol, 42.2% yield, 85% purity) was obtained as a white solid. MS (ESI) m/z: 1017.6 [M+H] + . Step F. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[4-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-7- yl]amino]-2-oxo-ethyl]piperazin-1-yl]propoxy]-2-methyl-pheny l]pyridine-2-carboxylic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[4-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indaz ol-7-yl]amino]-2-oxo- ethyl]piperazin-1-yl]propoxy]-2-methyl-phenyl]pyridine-2-car boxylate (130 mg, 127.80 µmol, 1 equiv.) and TFA (14.57 mg, 127.80 µmol, 9.46 µL, 1 equiv.) in DCM (2 mL) was stirred at 40 °C for 2 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[3-[4-[2-[[3-(2,6-dioxo- 3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-oxo-ethyl]pipera zin-1-yl]propoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid (18.8 mg, 18.6 µmol, 14.6% yield, 95.5% purity) as a white solid. MS (ESI) m/z: 961.2 [M+H] + . 1 H NMR (400 MHz, d 6 -DMSO) δ = 12.28 - 11.84 (m, 1H), 10.95 - 10.80 (m, 2H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.64 (dd, J = 14.0, 7.6 Hz, 2H), 7.49 - 7.25 (m, 6H), 7.21 (d, J = 7.2 Hz, 1H), 7.12 (t, J = 7.6 Hz, 2H), 6.99 (d, J = 8.8 Hz, 1H), 6.91 (d, J = 8.4 Hz, 1H), 6.66 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.50 - 4.35 (m, 2H), 4.16 - 4.03 (m, 6H), 3.92 (t, J = 5.6 Hz, 2H), 3.79 - 3.65 (m, 4H), 3.56 - 3.25 (m, 5H), 3.03 (t, J = 5.6 Hz, 2H), 2.71 - 2.60 (m, 2H), 2.44 - 2.10 (m, 5H), 1.93 (s, 3H). Example 122. Preparation of Compound 107 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[3-[[3-(2,6- dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-3-oxo-propyl ]phenoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) - 3, 4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[[3-(2, 6-dioxo-3-piperidyl) -1-methyl- indazol-6-yl]amino]-3-oxo-propyl]phenoxy]-2-methyl-phenyl]py ridine-2-carboxylate To a solution of 3-[4-[3-[6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]phenyl]propanoic acid (200 mg, 269.95 μmol, 1.1 equiv.) in DMF (2 mL) was added 3-(6-amino-1-methyl-indazol-3- yl) piperidine-2, 6-dione (63.38 mg, 245.41 μmol, 1 equiv.), HATU (102.64 mg, 269.95 μmol, 1.1 equiv.), and DIPEA (95.15 mg, 736.24 μmol, 128.24 μL, 3 equiv.) at 25 °C. The reaction mixture was stirred at 25 °C for 16 hours. The reaction mixture was diluted with water (10 mL) and filtered to give a residue. The compound tert-butyl 6-[8-(1, 3-benzothiazol-2- ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[[3-(2, 6-dioxo-3-piperidyl) -1- methyl-indazol-6-yl]amino]-3-oxo-propyl]phenoxy]-2-methyl-ph enyl]pyridine-2-carboxylate (240 mg, crude) was obtained as a yellow solid. MS (ESI) m/z: 981.5 [M+H] + . Step B. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[4-[3-[[3-(2,6-dioxo-3-piperidyl)- 1-methyl-indazol-6-yl]amino]- 3-oxo-propyl]phenoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid A solution of tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin- 2-yl]-3-[3-[4-[3-[[3-(2, 6-dioxo-3-piperidyl) -1-methyl-indazol-6-yl]amino]-3-oxo- propyl]phenoxy]-2-methyl-phenyl]pyridine-2-carboxylate (240 mg, 244.62 μmol, 1 equiv.) in DCM (1 mL) and TFA (1.5 mL) was stirred at 40 °C for 2 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC. The compound 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- [3-[4-[3-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]a mino]-3-oxo-propyl]phenoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid (40.5 mg, 41.4 μmol, 16.9% yield, 99.2% purity) was obtained as a yellow solid. MS (ESI) m/z: 925.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.87 (s, 1H), 10.09 (s, 1H), 8.08 - 8.00 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.66 - 7.31 (m, 8H), 7.23 (d, J = 8.4 Hz, 2H), 7.16 (t, J = 7.6 Hz, 1H), 7.08 - 7.04 (m, 1H), 7.00 (d, J = 8.8 Hz, 1H), 6.88 (d, J = 7.2 Hz, 1H), 6.82 (d, J = 8.4 Hz, 3H), 4.99 (s, 2H), 4.31 (dd, J = 5.2, 9.6 Hz, 1H), 3.95 - 3.91 (m, 2H), 3.91 (s, 3H), 3.03 (t, J = 5.6 Hz, 2H), 2.94 - 2.87 (m, 2H), 2.69 - 2.61 (m, 4H), 2.38 - 2.28 (m, 1H), 2.20 - 2.17 (m, 1H), 1.89 (s, 3H) Example 123. Preparation of Compound 109 2-(5-(1-( (3R, 5R, 7R) -adamantan-1-ylmethyl) -5-methyl-1H-pyrazol-4-yl) -6-( (10-( (3-(2, 6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-7-yl) amino) -10-oxodecyl) carbamoyl) pyridin- 2-yl) -N-(benzo[d]thiazol-2-yl) -1, 2, 3, 4-tetrahydroisoquinoline-8-carboxamide Step A. Procedure for preparation of methyl 10-aminodecanoate To a solution of 10-aminodecanoic acid (1 g, 5.34 mmol, 1 equiv.) in MeOH (10 mL) was added SOCl 2 (952.89 mg, 8.01 mmol, 581.03 μL, 1.5 equiv.) at 25 °C. The reaction mixture was stirred at 70 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give methyl 10-aminodecanoate (1.22 g, crude) as an off-white solid. 1 H NMR (400 MHz, CD 3 OD) δ = 3.64 (s, 3H), 2.91 (t, J = 7.6 Hz, 2H), 2.32 (t, J = 7.6 Hz, 2H), 1.58-1.68 (m, 4H), 1.40 - 1.32 (m, 10H). Step B. Procedure for preparation of methyl 10-(3-(1-(adamantan-1-ylmethyl)-5-methyl- 1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-di hydroisoquinolin-2(1H)- yl)picolinamido)decanoate To a solution of 3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)picolinic acid (1 g, 1.52 mmol, 1 equiv.) in DMF (10 mL) was added methyl 10-aminodecanoate (360.90 mg, 1.52 mmol, 1 equiv., HCl), HATU (577.15 mg, 1.52 mmol, 1 equiv.), and DIPEA (588.53 mg, 4.55 mmol, 793.17 μL, 3 equiv.) at 25 °C. The reaction mixture was stirred at 25 °C for 1.5 hours. The reaction mixture was diluted with H 2 O (10 mL) and extracted with EtOAc (30 mL × 3). The combined organic layers were washed with NaHCO 3 (10 mL × 3), brine (10 mL × 3), dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~8% MeOH/DCM) to give methyl 10-(3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6- (8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)picolinamido)decanoate (861 mg, 989.7 μmol, 65.2% yield, 96.8% purity) as a yellow oil. MS (ESI) m/z: 842.9 [M+H] + . Step C. Procedure for preparation of 10-(3-(1-(adamantan-1-ylmethyl)-5-methyl-1H- pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihyd roisoquinolin-2(1H)- yl)picolinamido)decanoic acid To a solution of methyl 10-(3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6- (8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) - yl)picolinamido)decanoate(831 mg, 986.82 μmol, 1 equiv.) in THF (1 mL) and H 2 O (0.1 mL) was added LiOH•H2O (124.23 mg, 2.96 mmol, 3 equiv.) at 25 °C. The reaction mixture was stirred at 40 °C for 20 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was treated with water (4 mL). The pH was adjusted to about 3 by progressively adding 1 M HCl. The mixture was filtered and concentrated under reduced pressure to give 10-(3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6- (8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)picolinamido)decanoic acid (670 mg, 778.6 μmol, 78.9% yield, 96.2% purity) as a yellow solid. MS (ESI) m/z: 828.8 [M+H] + . 1 H NMR (400 MHz, CD3OD) δ = 7.92 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.58 (dd, J = 1.2, 7.6 Hz, 1H), 7.49 - 7.44 (m, 2H), 7.42 - 7.39 (m, 1H), 7.39 - 7.37 (m, 1H), 7.36 (d, J = 2.4 Hz, 1H), 7.33 (s, 1H), 7.03 (d, J = 8.8 Hz, 1H), 5.14 (s, 2H), 3.93 (t, J = 6.0 Hz, 2H), 3.74 (s, 2H), 3.16 (t, J = 7.2 Hz, 2H), 3.08 (t, J = 6.0 Hz, 2H), 2.23 (t, J = 7.2 Hz, 2H), 2.10 (s, 3H), 1.97 (s, 3H), 1.77 - 1.65 (m, 6H), 1.61 (d, J = 2.0 Hz, 6H), 1.57 - 1.51 (m, 2H), 1.42 (s, 2H), 1.33 - 1.23 (m, 4H), 1.16 (d, J = 3.2 Hz, 6H) Step D. Procedure for preparation of 2-(5-(1-( (3R, 5R, 7R) -adamantan-1-ylmethyl) -5- methyl-1H-pyrazol-4-yl) -6-( (10-( (3-(2, 6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-7- yl) amino) -10-oxodecyl) carbamoyl) pyridin-2-yl) -N-(benzo[d]thiazol-2-yl) -1, 2, 3, 4- tetrahydroisoquinoline-8-carboxamide To a solution of 10-(3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6- (8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)picolinamido)decanoic acid (100.00 mg, 120.76 μmol, 1 equiv.) in DMF (1 mL) was added 3-(7-amino-1-methyl-1H- indazol-3-yl)piperidine-2,6-dione (31.19 mg, 120.76 μmol, 1 equiv.), HATU (45.92 mg, 120.76 μmol, 1 equiv.) and DIPEA (46.82 mg, 362.29 μmol, 63.10 μL, 3 equiv.) at 25 °C. The reaction mixture was stirred at 25 °C for 16 hours. The reaction mixture was quenched by addition water 1 mL at 25 °C, and filtered to give a residue. The residue was purified by prep- HPLC to give compound 2-(5-(1-( (3R, 5R, 7R) -adamantan-1-ylmethyl) -5-methyl-1H- pyrazol-4-yl) -6-( (10-( (3-(2, 6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-7-yl) amino) -10- oxodecyl) carbamoyl) pyridin-2-yl) -N-(benzo[d]thiazol-2-yl) -1, 2, 3, 4- tetrahydroisoquinoline-8-carboxamide (29.2 mg, 25.6 μmol, 21.2% yield, 93.5% purity) was obtained as a yellow solid. MS (ESI) m/z: 1068.3 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 12.85 - 12.80 (m, 1H), 10.91 (s, 1H), 9.82 (s, 1H), 8.06 (s, 1H), 8.01 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 - 7.57 (m, 2H), 7.49 - 7.43 (m, 2H), 7.43 - 7.39 (m, 1H), 7.37 - 7.32 (m, 2H), 7.23 (s, 1H), 7.11 - 7.04 (m, 2H), 6.95 (d, J = 8.8 Hz, 1H), 5.00 (s, 2H), 4.37 (dd, J = 5.2, 10.0 Hz, 1H), 4.03 (s, 3H), 3.88 (t, J = 6.0 Hz, 2H), 3.67 (s, 2H), 3.01 (d, J = 4.4 Hz, 4H), 2.69 - 2.56 (m, 2H), 2.40 - 2.32 (m, 3H), 2.20 - 2.13 (m, 1H), 1.91 (s, 4H), 1.66 - 1.59 (m, 5H), 1.56 (s, 2H), 1.51 (s, 7H), 1.33 - 1.20 (m, 7H), 1.20 - 1.07 (m, 7H) Example 124. Preparation of Compound 110 2-(5-(1-((3r,5r,7r)-adamantan-1-ylmethyl)-5-methyl-1H-pyrazo l-4-yl)-6-((12-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino)-12-oxod odecyl)carbamoyl)pyridin-2- yl)-N-(benzo[d]thiazol-2-yl)-1,2,3,4-tetrahydroisoquinoline- 8-carboxamide Step A. Procedure for preparation of methyl 12-(3-(1-(adamantan-1-ylmethyl)-5-methyl- 1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-di hydroisoquinolin-2(1H)- yl)picolinamido)dodecanoate To a solution of 3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)picolinic acid (1 g, 1.52 mmol, 1 equiv.) in DMF (10 mL) was added methyl 12-aminododecanoate; hydrochloride (403.48 mg, 1.52 mmol, 1 equiv.), DIPEA (588.53 mg, 4.55 mmol, 793.17 μL, 3 equiv.) and HATU (577.15 mg, 1.52 mmol, 1 equiv.) at 25 °C. The reaction mixture was stirred at 25 °C for 1.5 hours. The reaction mixture was diluted with H 2 O (10 mL) and extracted with EtOAc (30 mL × 3). The combined organic layers were washed with NaHCO 3 (10 mL × 3), brine (10 mL × 3), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~8% MeOH/DCM) to give methyl 12-(3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6- (8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)picolinamido)dodecanoate (1 g, 1.1 mmol, 74.0% yield, 97.8% purity) as a yellow oil. MS (ESI) m/z: 871.0 [M+H] + . Step B. Procedure for preparation of 12-(3-(1-(adamantan-1-ylmethyl)-5-methyl-1H- pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihyd roisoquinolin-2(1H)- yl)picolinamido)dodecanoic acid To a solution of methyl 12-(3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6- (8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)picolinamido)dodecanoate (970 mg, 1.11 mmol, 1 equiv.) in THF (1 mL) and H 2 O (0.1 mL) was added LiOH•H 2 O (140.34 mg, 3.34 mmol, 3 equiv.) at 25 °C. The reaction mixture was stirred at 40 °C for 20 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was treated with water (4 mL). The pH was adjusted to about 3 by progressively adding 1 M HCl. The mixture was filtered and concentrated under reduced pressure to give 12-(3-(1- (adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo [d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)picolinamido)dodecanoic acid (658 mg, 732.3 μmol, 65.6% yield, 95.2% purity) as a yellow solid. MS (ESI) m/z: 856.9 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 8.06 (s, 1H), 7.97 (d, J = 8.0 Hz, 1H), 7.74 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.46 - 7.41 (m, 2H), 7.38 - 7.31 (m, 2H), 7.23 (s, 1H), 6.94 (d, J = 8.8 Hz, 1H), 5.02 (s, 2H), 3.88 (t, J = 6.0 Hz, 2H), 3.67 (s, 2H), 3.04 - 2.97 (m, 4H), 2.17 (t, J = 7.2 Hz, 2H), 2.04 (s, 3H), 1.92 (s, 3H), 1.67 - 1.55 (m, 6H), 1.52 (s, 7H), 1.49 - 1.44 (m, 2H), 1.30 - 1.25 (m, 2H), 1.24 - 1.17 (m, 7H), 1.11 (d, J = 11.6 Hz, 6H) Step C. Procedure for preparation of 2-(5-(1-((3r,5r,7r)-adamantan-1-ylmethyl)-5- methyl-1H-pyrazol-4-yl)-6-((12-((3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-7- yl)amino)-12-oxododecyl)carbamoyl)pyridin-2-yl)-N-(benzo[d]t hiazol-2-yl)-1,2,3,4- tetrahydroisoquinoline-8-carboxamide A mixture of 12-(3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6- (8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)picolinamido)dodecanoic acid (80.00 mg, 93.44 µmol, 1 equiv.), 3-(7-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (26.55 mg, 102.79 µmol, 1.10 equiv.), HATU (39.08 mg, 102.79 µmol, 1.10 equiv.), TEA (47.28 mg, 467.22 µmol, 65.03 µL, 5.00 equiv.) in DMF (1 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 20 °C for 12 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 2-(5-(1-((3r,5r,7r)-adamantan-1-ylmethyl)-5-methyl-1H- pyrazol-4-yl)-6-((12-((3-(2,6-dioxopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-12- oxododecyl)carbamoyl)pyridin-2-yl)-N-(benzo[d]thiazol-2-yl)- 1,2,3,4- tetrahydroisoquinoline-8-carboxamide (16.3 mg, 14.3 µmol, 15.3% yield, 96.3% purity) as a yellow solid. MS (ESI) m/z: 1096.3 [M+1] + . 1 H NMR (400 MHz, CD3OD) δ = 7.91 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.68 - 7.63 (m, 1H), 7.56 (d, J = 6.8 Hz, 1H), 7.49 - 7.43 (m, 2H), 7.42 - 7.38 (m, 1H), 7.37 - 7.31 (m, 3H), 7.17 - 7.09 (m, 2H), 7.01 (d, J = 8.8 Hz, 1H), 5.13 (s, 2H), 4.41 - 4.33 (m, 1H), 4.09 (s, 3H), 3.92 (t, J = 6.0 Hz, 2H), 3.73 (s, 2H), 3.17 (t, J = 7.2 Hz, 2H), 3.07 (t, J = 5.6 Hz, 2H), 2.84 - 2.65 (m, 2H), 2.52 - 2.39 (m, 3H), 2.37 - 2.26 (m, 1H), 2.10 (s, 3H), 1.97 (s, 3H), 1.78 - 1.69 (m, 5H), 1.69 - 1.64 (m, 3H), 1.61 (d, J = 1.6 Hz, 6H), 1.47 - 1.38 (m, 4H), 1.36 - 1.27 (m, 5H), 1.23 - 1.16 (s, 7H) Example 125. Preparation of Compound 111 2-(5-(1-(((3r,5r,7r)-adamantan-1-yl)methyl)-5-methyl-1H-pyra zol-4-yl)-6-((6-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-6-oxohe xyl)carbamoyl)pyridin-2-yl)- N-(benzo[d]thiazol-2-yl)-1,2,3,4-tetrahydroisoquinoline-8-ca rboxamide Step A. Procedure for preparation of methyl 6-(3-(1-(adamantan-1-ylmethyl)-5-methyl- 1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-di hydroisoquinolin-2(1H)- yl)picolinamido)hexanoate To a solution of 3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)picolinic acid (1 g, 1.52 mmol, 1 equiv.) in DMF (10 mL) and methyl 6-aminohexanoate (551.48 mg, 3.04 mmol, 2 equiv., HCl) was added HATU (692.58 mg, 1.82 mmol, 1.2 equiv.) and DIEA (588.53 mg, 4.55 mmol, 793.17 µL, 3 equiv.). The mixture was stirred at 25 °C for 1 hour. The mixture was treated dropwise with water, filtered, and concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (Eluent of 0~60% ethyl acetate/petroleum ether) to give methyl 6-(3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6-( 8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)picolinamido)hexanoate (690 mg, 816.4 µmol, 53.7% yield, 93% purity) as a yellow solid. MS (ESI) m/z: 786.9 [M+H] + Step B. Procedure for preparation of 6-(3-(1-(adamantan-1-ylmethyl)-5-methyl-1H- pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihyd roisoquinolin-2(1H)- yl)picolinamido)hexanoic acid To a solution of methyl 6-(3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6-( 8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)picolinamido)hexanoate (670 mg, 852.42 µmol, 1 equiv.) in THF (7 mL) was added LiOH•H 2 O (1 M, 2.56 mL, 3 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was added HCl (1 M) to pH= 6, filtered to give a filter cake. The crude product was triturated with CH3CN (5 mL) at 25 °C for 0.5 hour to give 6-(3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6-( 8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)picolinamido)hexanoic acid (552.07 mg, 642.20 µmol, 75.3% yield, 89.8% purity) as a yellow solid. MS (ESI) m/z: 772.1 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 8.10 (t, J = 5.2 Hz, 1H), 7.90 (d, J = 7.6 Hz, 1H), 7.66 (t, J = 6.8 Hz, 2H), 7.45 (d, J = 8.8 Hz, 1H), 7.41 - 7.26 (m, 4H), 7.26 - 7.21 (m, 2H), 6.93 (d, J = 8.8 Hz, 1H), 5.05 (s, 2H), 3.89 (t, J = 5.6 Hz, 2H), 3.04 - 2.98 (m, 4H), 2.10 (t, J = 7.2 Hz, 2H), 2.05 (s, 3H), 1.92 (s, 3H), 1.68 - 1.61 (m, 4H), 1.58 - 1.51 (m, 10H), 1.41 - 1.36 (m, 2H), 1.33 - 1.27 (m, 2H), 1.15 - 1.08 (m, 2H). Step C. Procedure for preparation of 2-(5-(1-(((3r,5r,7r)-adamantan-1-yl)methyl)-5- methyl-1H-pyrazol-4-yl)-6-((6-((3-(2,6-dioxopiperidin-3-yl)- 1-methyl-1H-indazol-6- yl)amino)-6-oxohexyl)carbamoyl)pyridin-2-yl)-N-(benzo[d]thia zol-2-yl)-1,2,3,4- tetrahydroisoquinoline-8-carboxamide To a solution of 6-(3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6-( 8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)picolinamido)hexanoic acid (80 mg, 103.63 µmol, 1 equiv.) and 3-(6-amino-1-methyl-1H-indazol-3-yl)piperidine-2,6- dione (29.44 mg, 113.99 µmol, 1.1 equiv.) in DMF (1 mL) was added HATU (47.28 mg, 124.36 µmol, 1.2 equiv.) and DIEA (40.18 mg, 310.89 µmol, 54.15 µL, 3 equiv.) .The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 2-(5-(1-(((3r,5r,7r)-adamantan- 1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-((6-((3-(2,6-dioxop iperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-6-oxohexyl)carbamoyl)pyridin-2-yl)-N-(be nzo[d]thiazol-2-yl)-1,2,3,4- tetrahydroisoquinoline-8-carboxamide (68.6 mg, 62.5 µmol, 60.3% yield, 92.1% purity) as a yellow solid. MS (ESI) m/z: 1012.4 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 12.92 - 12.80 (m, 1H), 10.88 (s, 1H), 10.03 (s, 1H), 8.12 - 8.05 (m, 2H), 8.02 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.4 Hz, 1H), 7.63 - 7.57 (m, 2H), 7.47 - 7.42 (m, 2H), 7.42 - 7.38 (m, 1H), 7.37 - 7.29 (m, 2H), 7.24 (s, 1H), 7.07 (dd, J = 1.2, 8.8 Hz, 1H), 6.94 (d, J = 8.8 Hz, 1H), 4.98 (s, 2H), 4.34 - 4.28 (m, 1H), 3.91 - 3.85 (m, 5H), 3.66 (s, 2H), 3.07 - 2.97 (m, 4H), 2.64 - 2.61 (m, 1H), 2.27 (t, J = 7.2 Hz, 2H), 2.22 - 2.13 (m, 2H), 2.04 (s, 3H), 1.89 (s, 3H), 1.64 - 1.58 (m, 3H), 1.56 - 1.46 (m, 12H), 1.38 - 1.32 (m, 2H), 1.22 - 1.14 (m, 2H). Example 126. Preparation of Compound 112 2-[5-[1-(1-adamantylmethyl)-5-methyl-pyrazol-4-yl]-6-[[10-[[ 3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]amino]-10-oxo-decyl]carbamoyl]-2-pyridyl ]-N-(1,3-benzothiazol-2-yl)- 3,4-dihydro-1H-isoquinoline-8-carboxamide Step A. Procedure for preparation of 2-[5-[1-(1-adamantylmethyl)-5-methyl-pyrazol-4- yl]-6-[[10-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl ]amino]-10-oxo- decyl]carbamoyl]-2-pyridyl]-N-(1,3-benzothiazol-2-yl)-3,4-di hydro-1H-isoquinoline-8- carboxamide To a solution of 10-[[3-[1-(1-adamantylmethyl)-5-methyl-pyrazol-4-yl]-6-[8-(1 ,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] pyridine-2- carbonyl]amino]decanoic acid (80 mg, 96.61 µmol, 1 equiv.), HATU (36.73 mg, 96.61 µmol, 1 equiv.), and DIPEA (37.46 mg, 289.83 µmol, 50.48 µL, 3 equiv.) in DMF (1 mL) was added 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (27.45 mg, 106.27 µmol, 1.1 equiv.). The mixture was stirred at 25 °C for 6 hours. The mixture was concentrated as a residue. The residue was purified by prep-TLC (ethyl acetate : MeOH = 30 : 1) to give 2-[5-[1-(1- adamantylmethyl)-5-methyl-pyrazol-4-yl]-6-[[10-[[3-(2,6-diox o-3-piperidyl)-1-methyl- indazol-6-yl]amino]-10-oxo-decyl]carbamoyl]-2-pyridyl]-N-(1, 3-benzothiazol-2-yl)-3,4- dihydro-1H-isoquinoline-8-carboxamide (25.6 mg, 22.5 µmol, 23.3% yield, 93.6% purity) as a white solid. MS (ESI) m/z: 535.1 [M+2H] + . 1 H NMR (400 MHz, CD 3 OD) δ = 8.06 (d, J = 1.2 Hz, 1H), 7.90 (d, J = 7.6 Hz, 1H), 7.77 (d, J = 8.0 Hz, 1H), 7.61 (d, J = 8.8 Hz, 1H), 7.55 (dd, J = 1.2, 7.2 Hz, 1H), 7.47 - 7.42 (m, 2H), 7.38 - 7.30 (m, 4H), 7.08 (dt, J = 1.6, 4.2 Hz, 1H), 7.00 (d, J = 8.8 Hz, 1H), 5.14 - 5.10 (m, 2H), 4.34 - 4.30 (m, 1H), 3.97 (s, 1H), 3.94 (s, 3H), 3.93 - 3.90 (m, 2H), 3.71 (s, 2H), 3.66 - 3.53 (m, 2H), 3.16 (t, J = 7.3 Hz, 2H), 3.05 (t, J = 6.0 Hz, 2H), 2.80 - 2.69 (m, 3H), 2.46 - 2.29 (m, 5H), 2.23 - 2.16 (m, 2H), 2.09 (s, 3H), 2.05 - 1.94 (m, 4H), 1.70 - 1.65 (m, 5H), 1.61 - 1.56 (m, 3H), 1.32 - 1.26 (m, 6H), 1.20 - 1.18 (m, 4H). Example 127. Preparation of Compound 113 2-(5-(1-((3r,5r,7r)-adamantan-1-ylmethyl)-5-methyl-1H-pyrazo l-4-yl)-6-((12-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-12-oxod odecyl)carbamoyl)pyridin-2- yl)-N-(benzo[d]thiazol-2-yl)-1,2,3,4-tetrahydroisoquinoline- 8-carboxamide Step A. Procedure for preparation of 2-(5-(1-((3r,5r,7r)-adamantan-1-ylmethyl)-5- methyl-1H-pyrazol-4-yl)-6-((12-((3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-6- yl)amino)-12-oxododecyl)carbamoyl)pyridin-2-yl)-N-(benzo[d]t hiazol-2-yl)-1,2,3,4- tetrahydroisoquinoline-8-carboxamide A mixture of 12-(3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6- (8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)picolinamido)dodecanoic acid (60.00 mg, 70.08 µmol, 1 equiv.), 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (18.10 mg, 70.08 µmol, 1.00 equiv.), TEA (21.27 mg, 210.24 µmol, 29.26 µL, 3.00 equiv.), HATU (29.31 mg, 77.09 µmol, 1.10 equiv.) in DMF (1 mL) was degassed and purged with N 2 and stirred at 20 °C for 3 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 2-(5- (1-((3r,5r,7r)-adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-y l)-6-((12-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-12-oxod odecyl)carbamoyl)pyridin-2- yl)-N-(benzo[d]thiazol-2-yl)-1,2,3,4-tetrahydroisoquinoline- 8-carboxamide (18.6 mg, 16.1 µmol, 23.0% yield, 95.3% purity) as an off-white solid MS (ESI) m/z: 1096.5 [M+1] + . 1 H NMR (400 MHz, CD 3 OD) δ = 8.52 (s, 1H), 8.06 (s, 1H), 7.89 (d, J = 8.0 Hz, 1H), 7.77 (d, J = 8.1 Hz, 1H), 7.62 (d, J = 8.8 Hz, 1H), 7.56 (d, J = 6.4 Hz, 1H), 7.49 - 7.42 (m, 2H), 7.41 - 7.30 (m, 4H), 7.07 (dd, J = 1.6, 8.8 Hz, 1H), 7.00 (d, J = 8.4 Hz, 1H), 5.12 (s, 2H), 4.32 (dd, J = 5.2, 8.8 Hz, 1H), 3.95 (s, 3H), 3.91 (t, J = 6.0 Hz, 2H), 3.72 (s, 2H), 3.15 (t, J = 7.2 Hz, 2H), 3.06 (t, J = 5.6 Hz, 2H), 2.83 - 2.65 (m, 2H), 2.49 - 2.36 (m, 3H), 2.34 - 2.25 (m, 1H), 2.09 (s, 3H), 1.95 (s, 3H), 1.76 - 1.68 (m, 5H), 1.66 (s, 3H), 1.60 (s, 6H), 1.42 - 1.27 (m, 9H), 1.20 - 1.14 (s, 7H) Example 128. Preparation of Compound 115 2-(5-(1-((1R,3S)-adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4 -yl)-6-((2-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2-oxoet hyl)carbamoyl)pyridin-2-yl)- N-(benzo[d]thiazol-2-yl)-1,2,3,4-tetrahydroisoquinoline-8-ca rboxamide Step A. Procedure for preparation of 1-(1-adamantylmethyl)pyrazole To a solution of 1H-pyrazole (13.50 g, 198.30 mmol, 1.00 equiv.) in DMF (200 mL) was added Cs2CO3 (129.22 g, 396.61 mmol, 2.00 equiv.) and 1-(bromomethyl)adamantane (45.44 g, 198.30 mmol, 1.00 equiv.). The mixture was stirred at 80 °C for 12 hours. The reaction was diluted with water (600 mL) and extracted with ethyl acetate (300 mL × 2). The combined organic layers were washed with brine (150 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=100/1 to 50/1) to give 1-(1- adamantylmethyl)pyrazole (42 g, 194.1 mmol, 97.9% yield) as a white solid. MS (ESI) m/z: 217.3 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ =7.51 (d, J = 0.8 Hz, 1H), 7.33 (d, J = 2.0 Hz, 1H), 6.26 (t, J = 2.0 Hz, 1H), 3.83 (s, 2H), 2.00 (s, 2H), 1.70 (s, 2H), 1.64 - 1.57 (m, 4H), 1.53 (s, 6H) Step B. Procedure for preparation of 1-(1-adamantylmethyl)-4-iodo-pyrazole To a solution of 1-(1-adamantylmethyl)pyrazole (38.00 g, 175.66 mmol, 1.00 equiv.) in DMF (400 mL) was added NIS (51.38 g, 228.36 mmol, 1.30 equiv.). The mixture was stirred at 40 °C for 10 hours. The reaction was diluted with water (1.5 L) and extracted with ethyl acetate (1L × 2). The combined organic layers were washed with brine (1500 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give 1-(1-adamantylmethyl)-4- iodo-pyrazole (60.00 g, 175.3 mmol, 99.8% yield) as a red solid. MS (ESI) m/z: 342.9 [M+H] + . Step C. Procedure for preparation of 1-(1-adamantylmethyl)-4-iodo-5-methyl-pyrazole To a solution of 1-(1-adamantylmethyl)-4-iodo-pyrazole (30.00 g, 87.66 mmol, 1.00 equiv.) in THF (300 mL) was added LDA (2 M, 78.90 mL, 1.80 equiv.) dropwise at -75°C. The reaction was stirred for 2 hours during which time the temperature raised to -20 °C. MeI (16.18 g, 113.96 mmol, 7.09 mL, 1.30 equiv.) was added dropwise. The reaction was stirred at 25 °C for 10 hours. The reaction was diluted with water (200 mL) and extracted with ethyl acetate (200 mL × 2). The combined organic layers were washed with brine (60 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , dichloromethane: methanol=100/1 to 20/1) to give a residue. The crude product was triturated with petroleum ether=100 mL at 25 °C for 30 min to give 1-(1-adamantylmethyl)-4-iodo-5-methyl-pyrazole (26 g, 72.9 mmol, 83.2% yield) as a red solid. MS (ESI) m/z: 356.9 [M+H] + . 1 H NMR (400 MHz, CDCl3) δ = 7.37 (s, 1H), 3.70 (s, 2H), 2.22 (s, 3H), 1.91 (s, 3H), 1.61 (s, 2H), 1.58 - 1.49 (m, 4H), 1.47 (d, J = 2.4 Hz, 6H) Step D. Procedure for preparation of 1-(1-adamantylmethyl)-5-methyl-4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole To a solution of 1-(1-adamantylmethyl)-4-iodo-5-methyl-pyrazole (15.00 g, 42.11 mmol, 1 equiv.) in dioxane (150 mL) was added TEA (12.78 g, 126.32 mmol, 17.58 mL, 3.00 equiv.), Pd(dppf)Cl2.CH2Cl2 (3.44 g, 4.21 mmol, 0.10 equiv.) and 4,4,5,5-tetramethyl-1,3,2- dioxaborolane (10.78 g, 84.21 mmol, 12.22 mL, 2.00 equiv.). The mixture was stirred at 100 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=100/1 to 10/1) to give 1-(1-adamantylmethyl)-5-methyl-4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole (12 g, 33.6 mmol, 79.9% yield) as a white solid. MS (ESI) m/z: 357.1 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ = 7.71 (s, 1H), 3.74 (s, 2H), 2.44 (s, 3H), 1.99 (s, 3H), 1.73 - 1.68 (m, 3H), 1.64 (s, 3H), 1.60 (d, J = 2.4 Hz, 6H), 1.33 (s, 12H). Step E. Procedure for preparation of tert-butyl 3-(1-(adamantan-1-ylmethyl)-5-methyl- 1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-di hydroisoquinolin-2(1H)- yl)picolinate A mixture of 1-(1-adamantylmethyl)-5-methyl-4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2- yl)pyrazole (4.10 g, 11.49 mmol, 1.30 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-bromopicolin ate (5.00 g, 8.84 mmol, 1.00 equiv.), Ad2nBup-Pd-G3 (643.94 mg, 884.20 µmol, 0.10 equiv.), K2CO3 (3.67 g, 26.53 mmol, 3.00 equiv.) in dioxane (50 mL) and H2O (15 mL) was degassed and purged with N2 three times and then the mixture was stirred at 100 °C for 2 hours under N 2 atmosphere. The reaction was diluted with water (120 mL) and extracted with ethyl acetate (100 mL × 3). The combined organic layers were washed with brine (150 mL × 1), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=100/1 to 1/1) to give tert-butyl 3-(1- (adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo [d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)picolinate (6.00 g, 6.6 mmol, 74.9% yield, 79% purity) as a yellow solid. MS (ESI) m/z: 715.1 [M+H] + . Step F. Procedure for preparation of 3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol- 4-yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoqui nolin-2(1H)-yl)picolinic acid To a solution of tert-butyl 3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)picolinate (12.00 g, 16.79 mmol, 1.00 equiv.) in DCM (100 mL) was added TFA (123.20 g, 1.08 mol, 80 mL, 64.37 equiv.). The mixture was stirred at 40 °C for 1 hour. The reaction was diluted with DCM (300 mL) and extracted with water (800 mL × 3). The combined organic layers were washed with brine (500 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The crude product was triturated with MeCN (100mL) at 25 °C for 12 hours to give 3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-( benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)picolinic acid (11 g, 16.70 mmol, 99.47% yield) as a yellow solid. MS (ESI) m/z: 659.1 [M+H] + . 1 H NMR (400 MHz, DMSO) δ = 12.96 - 12.77 (m, 1H), 8.04 (d, J = 8.0 Hz, 1H), 7.80 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.54 - 7.32 (m, 6H), 7.27 (s, 1H), 6.95 (d, J = 8.8 Hz, 1H), 4.96 (s, 2H), 3.92 - 3.85 (m, 2H), 3.71 (s, 2H), 3.02 (t, J = 5.2 Hz, 2H), 2.10 (s, 3H), 1.93 (s, 3H), 1.67 - 1.62 (m, 3H), 1.58 - 1.50 (m, 9H). Step G. Procedure for preparation of methyl 2-(3-(1-(adamantan-1-ylmethyl)-5-methyl- 1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-di hydroisoquinolin-2(1H)- yl)picolinamido)acetate To a solution of 3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)picolinic acid (940.00 mg, 1.43 mmol, 1.00 equiv.) in DMF (10 mL) was added HATU (651.02 mg, 1.71 mmol, 1.20 equiv.), DIEA (553.22 mg, 4.28 mmol, 745.58 uL, 3.00 equiv.) and methyl 2-aminoacetate (214.97 mg, 1.71 mmol, 1.20 equiv., HCl). The mixture was stirred at 25 °C for 1 hour. The reaction was diluted with water (80 mL) and extracted with ethyl acetate 50 mL (50 mL × 3). The combined organic layers were washed with brine 60 mL (60 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=100/1 to 1/1) to give methyl 2-(3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6-( 8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)picolinamido)ac etate (900 mg, 1.1 mmol, 82.1% yield, 95% purity) as a yellow solid. MS (ESI) m/z: 730.1 [M+H] + . Step H. Procedure for preparation of 2-(3-(1-(adamantan-1-ylmethyl)-5-methyl-1H- pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihyd roisoquinolin-2(1H)- yl)picolinamido)acetic acid To a solution of methyl 2-(3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6-( 8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)picolinamido)acetate (900.00 mg, 1.23 mmol, 1.00 equiv.) in THF (10 mL) and H2O (3 mL) was added LiOH•H2O (155.23 mg, 3.70 mmol, 3.00 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was acidified to pH=5 with 1M HCl and extracted with DCM (50 mL × 3). The combined organic layers were washed with brine (50 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The crude product was triturated with petroleum ether: ethyl acetate=1:1(10mL) at 25 °C for 30 min to give 2-(3-(1-(adamantan-1- ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)picolinamido)acetic acid (500.3 mg, 698.8 µmol, 56.6% yield) as a yellow solid. MS (ESI) m/z: 716.2 [M+H] + . 1 H NMR (400 MHz, DMSO) δ = 12.67 - 12.39 (m, 1H), 8.63 - 8.54 (m, 1H), 8.03 (d, J = 7.8 Hz, 1H), 7.80 (d, J = 8.0 Hz, 1H), 7.60 (d, J = 6.8 Hz, 1H), 7.52 - 7.32 (m, 6H), 7.29 (s, 1H), 7.04 - 6.98 (m, 1H), 5.00 (s, 2H), 3.97 (t, J = 5.6 Hz, 2H), 3.86 (d, J = 5.6 Hz, 2H), 3.68 (s, 2H), 3.02 (t, J = 5.6 Hz, 2H), 2.05 (s, 3H), 1.93 (s, 3H), 1.68 - 1.63 (m, 3H), 1.59 - 1.52 (m, 9H) Step I. Procedure for preparation of 2-(5-(1-((1R,3S)-adamantan-1-ylmethyl)-5-methyl- 1H-pyrazol-4-yl)-6-((2-((3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-6-yl)amino)-2- oxoethyl)carbamoyl)pyridin-2-yl)-N-(benzo[d]thiazol-2-yl)-1, 2,3,4- tetrahydroisoquinoline-8-carboxamide A mixture of 2-(3-(1-(adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6-( 8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)picolinamido)acetic acid (60 mg, 83.82 µmol, 1 equiv.), 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (21.65 mg, 83.82 µmol, 1 equiv.), HATU (31.87 mg, 83.82 µmol, 1 equiv.), and DIPEA (32.50 mg, 251.45 µmol, 43.80 µL, 3 equiv.) in DMF (0.6 mL) was stirred at 25 o C and 1 hour, then add 2-[[3-[1-(1-adamantylmethyl)-5-methyl-pyrazol-4-yl]-6-[8-(1, 3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]pyridine-2-carb onyl]amino]acetic acid (60 mg, 83.82 µmol, 1 equiv.) into the mixture at 25 °C and then stirred at 25 °C for 14 hours. The mixture was filtered, and the filtrate was purified by prep-HPLC to give 2-(5-(1-((1R,3S)- adamantan-1-ylmethyl)-5-methyl-1H-pyrazol-4-yl)-6-((2-((3-(2 ,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)carbamoyl)pyridin-2 -yl)-N-(benzo[d]thiazol-2- yl)-1,2,3,4-tetrahydroisoquinoline-8-carboxamide (13.9 mg, 12.8 µmol, 15.3% yield, 97.5% purity) as a yellow solid. MS (ESI) m/z: 965.5 [M+H] + . 1 H NMR (400 MHz, CD3OD) δ = 8.28 (s, 1H), 7.95 (s, 1H), 7.53 - 7.62 (m, 4H), 7.36 - 7.42 (m, 3H), 7.26 (t, J = 7.6 Hz, 1H), 7.15 (d, J = 8.8 Hz, 1H), 7.08 (t, J = 7.6 Hz, 1H), 6.98 (d, J = 7.6 Hz, 1H), 5.24 (s, 2H), 4.35 (dd, J = 9.2, 5.2 Hz, 1H), 4.21 (s, 2H), 4.00 (t, J = 6.0 Hz, 2H), 3.89 (s, 3H), 3.72 (s, 2H), 3.11 (t, J = 6.0 Hz, 2H), 2.74 - 2.84 (m, 2H), 2.44 - 2.52 (m, 1H), 2.34 (dd, J = 13.2, 6.0 Hz, 1H), 2.11 (s, 2H), 1.92 (s, 3H), 1.68-1.71 (m, 2H), 1.57-1.63 (m, 10H). Example 129. Preparation of Compound 118 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[2-[4-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-oxo-e thyl]phenoxy]ethoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 1-bromo-3-(2-bromoethoxy)-2-methylbenzene A mixture of 3-bromo-2-methylphenol (2 g, 10.69 mmol, 1 equiv.), 1,2-dibromoethane (10.00 g, 53.23 mmol, 4.02 mL, 5.00 equiv.) and K2CO3 (4.41 g, 31.94 mmol, 3 equiv.) in CH3CN (20 mL) was degassed and purged with N 2 and stirred at 80 °C for 12 hours. The reaction mixture was concentrated and diluted with water 30 mL and extracted with EtOAc (30 mL × 3). The combined organic layers were washed with brine (30 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~5% ethyl acetate/petroleum ether) to give 1-bromo-3-(2-bromoethoxy)-2-methylbenzene (1.8 g, 6.1 mmol, 57.5% yield) as a yellow oil. 1 H NMR (400 MHz, CD3OD) δ = 7.19 - 7.15 (m, 1H), 7.04 (t, J = 8.0 Hz, 1H), 6.88 (d, J = 8.0 Hz, 1H), 4.33 - 4.28 (m, 2H), 3.73 (t, J = 5.6 Hz, 2H), 2.34 (s, 3H) Step B. Procedure for preparation of methyl 2-(4-(2-(3-bromo-2- methylphenoxy)ethoxy)phenyl)acetate A mixture of 1-bromo-3-(2-bromoethoxy)-2-methylbenzene (200 mg, 680.31 µmol, 1 equiv.), methyl 2-(4-hydroxyphenyl)acetate (135.66 mg, 816.37 µmol, 1.2 equiv.), and K 2 CO 3 (282.07 mg, 2.04 mmol, 3 equiv.) in CH3CN (3 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 60 °C for 16 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , petroleum ether: ethyl acetate = 8: 1) to give methyl 2-(4-(2-(3- bromo-2-methylphenoxy)ethoxy)phenyl)acetate (150 mg, 395.5 µmol, 58.1% yield) as a colorless oil. 1 H NMR (400 MHz, CD 3 OD) δ = 7.23 - 7.16 (m, 3H), 7.07 (t, J = 8.0 Hz, 1H), 6.99 - 6.93 (m, 3H), 4.36 - 4.33 (m, 4H), 3.69 (s, 3H), 3.60 (s, 2H), 2.29 (s, 3H) Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-(2-methoxy-2-oxoethy l)phenoxy)ethoxy)-2- methylphenyl)picolinate Tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4,4,5,5- tetrameth yl-1,3,2-dioxaborolan-2-yl)picolinate (218.05 mg, 355.97 µmol, 0.9 equiv.), methyl 2-(4-(2-(3-bromo-2-methylphenoxy)ethoxy)phenyl)acetate (150 mg, 395.52 µmol, 1 equiv.), KF (1.5 M, 791.05 µL, and Ad2nBup-Pd-G3 (28.80 mg, 39.55 µmol, 0.1 equiv.) were taken up into a microwave tube in 1,4-dioxane (4 mL). The sealed tube was heated at 100 °C for 2 hours under microwave. The reaction mixture was diluted with water (30 mL) and extracted with EtOAc (30 mL× 3). The combined organic layers were washed with brine (30 mL × 2), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , petroleum ether: ethyl acetate= 1.5: 1) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- (3-(2-(4-(2-methoxy-2-oxoethyl)phenoxy)ethoxy)-2-methylpheny l)picolinate (220 mg, 280.2 µmol, 70.8% yield) as a yellow oil. MS (ESI) m/z: 785.4 [M+H] + Step D. Procedure for preparation of 2-(4-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyri din-3-yl)-2- methylphenoxy)ethoxy)phenyl)acetic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(2-(4-(2-methoxy-2-oxoethyl)phenoxy)ethoxy)-2-methy lphenyl)picolinate (220 mg, 280.28 µmol, 1 equiv.) and LiOH•H2O (35.29 mg, 840.85 µmol, 3 equiv.) in THF (1.6 mL) and H2O (0.4 mL) was degassed and purged with N2 and stirred at 25 °C for 1 hour under N2 atmosphere. The reaction mixture was diluted with water (30 mL) and extracted with EtOAc (30 mL× 3). The combined organic layers were washed with brine (30 mL× 3), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 2-(4-(2-(3- (6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)ethoxy)phenyl)a cetic acid (380 mg, crude) as a yellow solid. MS (ESI) m/z: 771.6 [M+H] + Step E. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[4-[2-[[3-(2,6-diox o-3-piperidyl)-1-methyl- indazol-7-yl]amino]-2-oxo-ethyl]phenoxy]ethoxy]-2-methyl-phe nyl]pyridine-2- carboxylate A mixture of 2-[4-[2-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H-isoquinolin- 2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]etho xy]phenyl]acetic acid (180 mg, 233.50 µmol, 1 equiv.), 3-(7-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (60.31 mg, 233.50 µmol, 1 equiv.), DIEA (90.53 mg, 700.50 µmol, 122.01 µL, 3 equiv.) in DMF (3 mL) was degassed and purged with N2 and stirred at 25°C for 5 minutes. After 5 minutes, added HATU (106.54 mg, 280.20 µmol, 1.2 equiv.), the mixture was stirred at 40 °C for 1 hour under N 2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (25 mL × 3). The combined organic layers were washed with brine (15 mL × 3), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was used in the next step without purification. The compound tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[2-[4-[2-[[3-(2,6-dioxo- 3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-oxo-ethyl]phenox y]ethoxy]-2-methyl- phenyl]pyridine-2-carboxylate (200 mg, crude) was obtained as a yellow oil. MS (ESI) m/z: 1011.3 [M+H] + . Step F. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[2-[4-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-7- yl]amino]-2-oxo-ethyl]phenoxy]ethoxy]-2-methyl-phenyl]pyridi ne-2-carboxylic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[2-[4-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indaz ol-7-yl]amino]-2-oxo- ethyl]phenoxy]ethoxy]-2-methyl-phenyl]pyridine-2-carboxylate (200 mg, 197.79 µmol, 1 equiv.) in DCM (1 mL) and TFA (1 mL) was degassed and purged with N2 and stirred at 40 °C for 3 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC. The compound 6- [8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinol in-2-yl]-3-[3-[2-[4-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-oxo-e thyl]phenoxy]ethoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid (25.65 mg, 26.6 µmol, 13.4% yield, 99.2% purity) was obtained as a yellow solid. MS (ESI) m/z: 956.2 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.88 - 12.80 (m, 1H), 10.87 (s, 1H), 9.99 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.4 Hz, 1H), 7.64 - 7.55 (m, 2H), 7.49 - 7.42 (m, 3H), 7.42 - 7.24 (m, 5H), 7.14 - 7.04 (m, 3H), 7.00 - 6.94 (m, 4H), 6.67 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.37 - 4.30 (m, 5H), 3.91 (t, J = 5.6 Hz, 2H), 3.87 (s, 3H), 3.65 (s, 2H), 3.02 (s, 2H), 2.70 - 2.64 (m, 2H), 2.19 - 2.09 (m, 2H), 1.89 (s, 3H) Example 130. Preparation of Compound 123 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[3-[[3-(2,6- dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]propyl]phenox y]-2-methyl-phenyl]pyridine- 2-carboxylic acid Step A. Procedure for preparation of ethyl 3-[4-(3-bromo-2-methyl- phenoxy)phenyl]propanoate A mixture of ethyl 3-(4-hydroxyphenyl)propanoate (5 g, 25.74 mmol, 1 equiv.), 1,3-dibromo- 2-methyl-benzene (12.87 g, 51.49 mmol, 2 equiv.), Cs 2 CO 3 (10.07 g, 30.89 mmol, 1.2 equiv.) and 2,2,6,6-tetramethylheptane-3,5-dione (1.19 g, 6.44 mmol, 1.33 mL, 0.25 equiv.) in NMP (50 mL) was degassed and purged with N2 and then CuI (2.45 g, 12.87 mmol, 0.5 equiv.) was added to the mixture and the mixture was stirred at 120 °C for 12 hours under N 2 atmosphere. The reaction mixture was filtered, and the filtrate was diluted with water 30 mL and extracted with ethyl acetate (40 mL × 2). The combined organic layers were washed with brine (20 mL × 2), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~30% ethyl acetate/petroleum ether). The compound ethyl 3-[4-(3-bromo-2-methyl- phenoxy)phenyl]propanoate (7.6 g, 20.9 mmol, 81.2% yield) was obtained as a colorless oil. 1 H NMR (400 MHz, CDCl 3 ) δ = 7.35 (d, J = 8.0 Hz, 1H), 7.15 (d, J = 8.4 Hz, 2H), 7.01 (t, J = 8.0 Hz, 1H), 6.86 - 6.80 (m, 3H), 4.14 (q, J = 7.2 Hz, 2H), 2.93 (t, J = 8.0 Hz, 2H), 2.61 (t, J = 8.0 Hz, 2H), 2.34 (s, 3H), 1.25 (t, J = 7.2 Hz, 3H) Step B. Procedure for preparation of 3-[4-(3-bromo-2-methyl-phenoxy)phenyl]propan-1- ol A mixture of ethyl 3-[4-(3-bromo-2-methyl-phenoxy)phenyl]propanoate (7.6 g, 20.92 mmol, 1 equiv.) in THF (70 mL) was slowly added LiAlH4 (794.01 mg, 20.92 mmol, 1 equiv.) at 0 °C, then the mixture was stirred at 25 °C for 2 hours under N2 atmosphere The mixture was poured into ice-water (150 mL) slowly and the pH was acidized to pH 4-5, extracted with EtOAc (60 mL ×3), washed with brine (30 mL ×2), dried by sodium sulfate, filtered and concentrated under reduced pressure. The compound 3-[4-(3-bromo-2-methyl- phenoxy)phenyl]propan-1-ol (6 g, 18.6 mmol, 89.2% yield) was obtained as a yellow oil. 1 H NMR (HNMR, 400 MHz, CDCl3) δ = 7.32 (d, J = 8.0 Hz, 1H), 7.13 (d, J = 8.4 Hz, 2H), 6.98 (t, J = 8.0 Hz, 1H), 6.85 - 6.79 (m, 3H), 3.67 (t, J = 6.4 Hz, 2H), 2.70 - 2.66 (m, 2H), 2.33 (s, 3H), 1.91 - 1.83 (m, 2H) Step C. Procedure for preparation of 3-[4-(3-bromo-2-methyl-phenoxy)phenyl]propanal To a solution of oxalyl dichloride (2.37 g, 18.68 mmol, 1.64 mL, 2 equiv.) in DCM (5 mL) was added the mixture of DMSO (2.92 g, 37.36 mmol, 2.92 mL, 4 equiv.) in DCM (5 mL) under - 70 °C and stirred for 0.5 hours. And 3-[4-(3-bromo-2-methyl-phenoxy)phenyl]propan-1-ol (3 g, 9.34 mmol, 1 equiv.) in DCM (5 mL) was stirred for 10 mins and added into the mixture for 20 mins. TEA (5.67 g, 56.04 mmol, 7.80 mL, 6 equiv.) was added into the mixture and stirred at -70 °C for 2 hours. The reaction mixture was quenched by addition of water 60 mL at 0 °C, and then extracted with DCM 90 mL (30 mL × 2). The combined organic layers were washed with brine 40 mL (20 mL × 2), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~15% ethyl acetate/petroleum ether). The compound 3-[4-(3-bromo-2-methyl- phenoxy)phenyl]propanal (2.6 g, 8.1 mmol, 87.21% yield) was obtained as a yellow oil. 1 H NMR (HNMR, 400 MHz, CDCl 3 ) δ = 9.84 (s, 1H), 7.35 (d, J = 8.0 Hz, 1H), 7.15 (d, J = 8.4 Hz, 2H), 7.02 (t, J = 8.0 Hz, 1H), 6.86 - 6.81 (m, 3H), 2.97 - 2.91 (m, 2H), 2.82 - 2.76 (m, 2H), 2.34 (s, 3H) Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[2-methyl-3-[4-(3-oxoprop yl)phenoxy]phenyl] pyridine-2-carboxylate A mixture of 3-[4-(3-bromo-2-methyl-phenoxy)phenyl]propanal (100 mg, 313.29 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-2-carb oxylate (153.52 mg, 250.63 µmol, 0.8 equiv.), Ad 2 nBuP Pd G 3 (cataCXium® A Pd G 3 ) (45.63 mg, 62.66 µmol, 0.2 equiv.), K2CO3 (1.5 M, 313.29 µL, 1.5 equiv.) in 1,4-dioxane (1 mL) was degassed and purged with N2 and stirred at 80 °C for 2 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , DCM: MeOH = 10:1). The compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[2-methyl-3-[4-(3-oxopropyl)phenoxy]p henyl] pyridine-2-carboxylate (100 mg, 107.3 µmol, 11.4% yield, 77.7% purity) was obtained as a yellow oil. MS (ESI) m/z: 725.2 [M+H] + Step E. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[[3-(2,6-dioxo-3 -piperidyl)-1-methyl- indazol-6-yl]amino]propyl]phenoxy]-2-methyl-phenyl]pyridine- 2-carboxylate A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[2-methyl-3-[4-(3-oxopropyl)phenoxy]phenyl] pyridine-2-carboxylate (100 mg, 137.96 µmol, 1 equiv.), 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (35.63 mg, 137.96 µmol, 1 equiv.), HCOOH (6.63 mg, 137.96 µmol, 1 equiv.), and NaBH(OAc)3 (43.86 mg, 206.94 µmol, 1.5 equiv.) in DCM (1 mL) was stirred at 25 °C for 15 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- [3-[4-[3-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]a mino]propyl]phenoxy]-2- methyl-phenyl]pyridine-2-carboxylate (120 mg, crude) was obtained as yellow solid and used into the next step without further purification. MS (ESI) m/z: 969.4 [M+H] + . Step F. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[4-[3-[[3-(2,6-dioxo-3-piperidyl)- 1-methyl-indazol-6- yl]amino]propyl]phenoxy]-2-methyl-phenyl]pyridine-2-carboxyl ic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[4-[3-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol- 6-yl]amino]propyl]phenoxy]-2- methyl-phenyl]pyridine-2-carboxylate (120 mg, 124.08 µmol, 1 equiv.), TFA (42.44 mg, 372.23 µmol, 27.56 µL, 3 equiv.) in DCM (1 mL) was stirred at 25 °C for 44 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]propyl]phenoxy]-2-methyl-phenyl]pyridine-2-carboxyl ic acid (29.69 mg, 22.8 µmol, 18.4% yield, 69.9% purity) as a white solid. MS (ESI) m/z: 911.4 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.86 (s, 1H), 8.08 (d, J = 8.0 Hz, 1H), 7.84 (d, J =8.0 Hz, 1H), 7.68 (d, J = 7.6 Hz, 1H), 7.57 (d, J = 8.8 Hz, 1H), 7.55 - 7.48 (m, 2H), 7.45 - 7.37 (m, 3H), 7.34 - 7.15 (m, 4H), 7.04 (d, J = 8.4 Hz, 1H), 6.94 (d, J = 7.6 Hz, 1H), 6.89 (d, J = 8.0 Hz, 3H), 6.59 (d, J = 8.4 Hz, 1H), 6.32 (s, 1H), 6.00 (s, 1H), 5.05 (s, 2H), 4.23 (dd, J = 5.2, 8.4 Hz, 1H), 3.98 (t, J = 5.6 Hz, 2H), 3.84 (s, 3H), 3.17 - 3.12 (m, 2H), 3.09 (t, J = 5.2 Hz, 2H), 2.76 - 2.72 (m, 2H), 2.66 (t, J = 6.4 Hz, 2H), 2.36 - 2.25 (m, 2H), 2.24 - 2.10 (m, 2H), 1.96 (s, 3H) Example 131. Preparation of Compound 124a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(3- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino )-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of ethyl 4-(3-bromo-2-methyl- phenoxy)cyclohexanecarboxylate To a solution of ethyl 4-hydroxycyclohexanecarboxylate (10 g, 58.06 mmol, 1 equiv.), 3- bromo-2-methyl-phenol (11.95 g, 63.87 mmol, 1.1 equiv.), and PPh 3 (24.37 g, 92.90 mmol, 1.6 equiv.) in THF (100 mL) was added DIAD (18.79 g, 92.90 mmol, 18.06 mL, 1.6 equiv.) under 0 °C. Then the mixture was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 10% ethyl acetate / petroleum ether) to give ethyl 4-(3-bromo- 2-methyl-phenoxy)cyclohexanecarboxylate (3.6 g, 10.55 mmol, 18.17% yield) as a white solid. 1 H NMR (400 MHz, CDCl3) δ = 7.14 (d, J = 8.0 Hz, 1H), 6.97 (t, J = 8.0 Hz, 1H), 6.80 (d, J = 8.0 Hz, 1H), 4.20 - 4.11 (m, 3H), 2.39 - 2.35 (m, 2H), 2.30 (s, 3H), 2.19 - 2.14 (m, 2H), 2.09 - 2.04 (m, 2H), 1.62 - 1.52 (m, 5H), 1.26 (t, J = 6.8 Hz, 3H). Step B. Procedure for preparation of [4-(3-bromo-2-methyl- phenoxy)cyclohexyl]methanol To a solution of ethyl 4-(3-bromo-2-methyl-phenoxy)cyclohexanecarboxylate (3.6 g, 10.55 mmol, 1 equiv.) in THF (15 mL) was added LiAlH4 (480.49 mg, 12.66 mmol, 1.2 equiv.) under 0 °C. Then the mixture was stirred at 25 °C for 1.5 hours. The reaction mixture was quenched by addition Na 2 SO 4 •10H 2 O (1 g), and then extracted with ethyl acetate (10 mL × 3). The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give compound [4-(3-bromo-2-methyl-phenoxy)cyclohexyl]methanol (2.96 g, crude) as a white solid. 1 H NMR (400 MHz, CDCl 3 ) δ = 7.13 (d, J = 8.0 Hz, 1H), 6.97 (t, J = 8.0 Hz, 1H), 6.81 (d, J = 8.4 Hz, 1H), 4.13 - 4.08(m, 1H), 3.50 (d, J = 6.4 Hz, 2H), 2.30 (s, 3H), 2.20 - 2.14 (m, 2H), 1.95 - 1.86 (m, 2H), 1.53 - 1.45 (m, 3H), 1.14 - 1.04 (m, 2H). Step C. Procedure for preparation of 4-(3-bromo-2-methyl- phenoxy)cyclohexanecarbaldehyde To a solution of DMSO (3.09 g, 39.57 mmol, 3.09 mL, 4 equiv.) in DCM (10 mL) was added dropwise a solution of (COCl) 2 (2.51 g, 19.79 mmol, 1.73 mL, 2 equiv.) in DCM (2 mL) at - 70 °C under N2 atmosphere. The mixture was stirred at -70 °C for 1 hour. After that, [4-(3- bromo-2-methyl-phenoxy)cyclohexyl]methanol (2.96 g, 9.89 mmol, 1 equiv.) in DCM (10 mL) was added dropwise at -70 °C. The solution was stirred for 1 hour at -70 °C. Then TEA (6.01 g, 59.36 mmol, 8.26 mL, 6 equiv.) was added into the solution. The solution was stirred at -70 °C for 0.5 hour under N2 atmosphere. The reaction mixture was quenched by addition H2O (10 mL), and then diluted with DCM (10 mL) and extracted with DCM (10 mL × 3). The combined organic layers were washed with H 2 O 5 mL × 3, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give compound 4-(3-bromo-2-methyl- phenoxy)cyclohexanecarbaldehyde (3 g, crude) as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ = 9.62 - 9.59 (m, 1H), 7.16 - 7.01 (m, 3H), 4.36 - 4.23 (m, 1H), 2.39 - 2.31 (m, 1H), 2.25 - 2.18 (m, 3H), 2.03 - 1.92 (m, 4H), 1.50 - 1.38 (m, 4H). Step D. Procedure for preparation of ethyl (E)-3-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]prop-2-enoate To a solution of NaH (56.52 mg, 1.41 mmol, 60% purity, 2.1 equiv.) in THF (3 mL) was added ethyl 2-diethoxyphosphorylacetate (301.75 mg, 1.35 mmol, 267.04 µL, 2 equiv.) under 0 °C. Then 4-(3-bromo-2-methyl-phenoxy)cyclohexanecarbaldehyde (200 mg, 672.98 µmol, 1 equiv.) was added and the mixture was stirred at 25 °C for 6 hours. The reaction mixture was quenched by addition sat. NH4Cl (5 mL) at 0 °C, and then diluted with ethyl acetate (5 mL) and extracted with ethyl acetate (10 mL × 3). The combined organic layers were washed with H 2 O (10 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~10% ethyl acetate / petroleum ether) to give ethyl (E)-3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]prop- 2-enoate (140 mg, 362.1 µmol, 53.8% yield, 95% purity) as a colorless oil. 1 H NMR (400 MHz, CDCl 3 ) δ = 7.14 (d, J = 8.0 Hz, 1H), 7.01 - 6.87 (m, 2H), 6.80 (d, J = 8.4 Hz, 1H), 5.82 (dd, J = 1.2, 15.6 Hz, 1H), 4.21 - 4.15 (m, 2H), 4.16 - 4.07 (m, 1H), 2.30 (s, 3H), 2.26 - 2.16 (m, 3H), 1.97 - 1.89 (m, 2H), 1.58 - 1.48 (m, 2H), 1.33 - 1.26 (m, 5H). Step E. Procedure for preparation of ethyl 3-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]propanoate A mixture of ethyl (E)-3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]prop-2-enoate (1.6 g, 4.36 mmol, 1 equiv.) and PtO 2 (98.92 mg, 435.64 µmol, 0.1 equiv.) in EtOH (15 mL) was degassed and purged with H2 three times, and then the mixture was stirred at 25 °C for 3 hours under H2 (15 Psi) atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give ethyl 3-[4- (3-bromo-2-methyl-phenoxy)cyclohexyl]propanoate (1.4 g, 3.7 mmol, 87.0% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.17 - 7.10 (m, 1H), 7.06 (t, J = 8.0 Hz, 1H), 7.03 - 6.99 (m, 1H), 4.27 - 4.17 (m, 1H), 4.11 - 4.04 (m, 2H), 2.33 - 2.26 (m, 2H), 2.20 (s, 3H), 2.03 (d, J = 10.0 Hz, 2H), 1.75 (d, J = 12.0 Hz, 2H), 1.55 - 1.42 (m, 2H), 1.39 - 1.21 (m, 3H), 1.17 (t, J = 7.2 Hz, 3H), 1.11 - 0.97 (m, 2H). Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-ethoxy-3-ox opropyl)cyclohexyl)oxy)-2- methylphenyl)picolinate A mixture of ethyl 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]propanoate (200 mg, 541.58 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (546.40 mg, 758.21 µmol, 85% purity, 1.4 equiv.), [2-(2- aminophenyl)phenyl]palladium(1 + );bis(1-adamantyl)-butyl-phosphane;methanesulfonate (78.88 mg, 108.32 µmol, 0.2 equiv.), and K2CO3 (1.5 M, 541.58 µL, 1.5 equiv.) in dioxane (2.5 mL) was degassed and purged with N2 and stirred at 100 °C for 1 hour under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The mixture was purified by flash silica gel chromatography (Eluent of 0 ~ 50% ethyl acetate/petroleum ether) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-ethoxy-3-ox opropyl)cyclohexyl)oxy)-2- methylphenyl)picolinate (440 mg, crude) as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ = 8.02 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.60 (d, J = 7.5 Hz, 1H), 7.50 - 7.40 (m, 3H), 7.40 - 7.30 (m, 2H), 7.12 - 7.03 (m, 1H), 6.99 - 6.89 (m, 2H), 6.59 - 6.49 (m, 1H), 5.03 - 4.90 (m, 2H), 4.27 - 4.14 (m, 1H), 4.03 (d, J = 6.8 Hz, 2H), 3.87 (t, J = 5.6 Hz, 2H), 3.29 (s, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.29 (t, J = 7.6 Hz, 2H), 2.05 (d, J = 11.2 Hz, 1H), 1.84 (s, 3H), 1.79 - 1.70 (m, 2H), 1.50 - 1.42 (m, 2H), 1.23 (d, J = 8.4 Hz, 2H), 1.19 - 1.17 (m, 3H), 1.07 (s, 2H), 1.00 (s, 9H). Step G. Procedure for preparation of 3-((1r,4r)-4-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)cyclohexyl)propanoic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1r,4r)-4-(3-ethoxy-3-oxopropyl)cyclohexyl)oxy)-2 -methylphenyl)picolinate (440 mg, 567.77 µmol, 1 equiv.), LiOH•H 2 O (71.47 mg, 1.70 mmol, 3 equiv.) in THF (1.2 mL) and H 2 O (0.4 mL) was stirred at 25 °C for 1.5 hours. The mixture was concentrated and redissolved by H 2 O (2 mL), and then 1M HCl was added to adjust the pH to 2. The residue was filtered and dried to give 3-((1r,4r)-4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)cyclohexyl)propanoic acid (380 mg, 508.7 µmol, 89.6% yield) was obtained as a gray solid. Step H. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-((3-(2,6-di oxopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-3-oxopropyl)cyclohexyl)oxy)-2-methylp henyl)picolinate A mixture of 3-((1r,4r)-4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-di hydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)cyclohexyl)propanoic acid (150 mg, 200.83 µmol, 1 equiv.), 3-(7-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (57.06 mg, 220.91 µmol, 1.1 equiv.), HATU (76.36 mg, 200.83 µmol, 1 equiv.), and DIPEA (77.86 mg, 602.48 µmol, 104.94 µL, 3 equiv.) in DMF (0.5 mL) was stirred at 25 °C for 12 hours. The reaction mixture was diluted with H2O (1 mL) and extracted with ethyl acetate (1 mL × 3). The combined organic layers were washed with H2O (1 mL × 3), dried over anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1r,4r)-4-(3-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino)-3- oxopropyl)cyclohexyl)oxy)-2- methylphenyl)picolinate (130 mg, crude) as a white solid. Step I. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-((3-(2,6-di oxopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-3-oxopropyl)cyclohexyl)oxy)-2-methylp henyl)picolinic acid Tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1r,4r)-4-(3-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-ind azol-7-yl)amino)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (130 mg, 131.69 µmol, 1 equiv.) in TFA (0.5 mL) and DCM (0.5 mL) was stirred at 40 °C for 6 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)- 4-(3-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl) amino)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (47.2 mg, 50.2 µmol, 38.1% yield, 99.03% purity) as a white solid. MS (ESI) m/z: 931.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.88 (s, 1H), 9.88 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.61 (dd, J = 7.6, 12.0 Hz, 2H), 7.52 - 7.30 (m, 6H), 7.16 - 7.02 (m, 3H), 6.96 (dd, J = 8.8, 13.2 Hz, 2H), 6.62 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.37 (dd, J = 4.4, 8.8 Hz, 1H), 4.27 - 4.16 (m, 2H), 4.05 (s, 3H), 3.91 (s, 2H), 3.03 (s, 2H), 2.74 - 2.60 (m, 2H), 2.47 - 2.32 (m, 4H), 2.20 - 2.06 (m, 3H), 1.88 (s, 3H), 1.59 (d, J = 6.4 Hz, 2H), 1.45 - 1.30 (m, 3H), 1.21 - 1.13 (m, 2H). Example 132. Preparation of Compound 128 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(4-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino)-4-oxobu tyl)phenoxy)-2- methylphenyl)picolinic acid Step A. Procedure for preparation of ethyl 4-(2-(3-bromo-2- methylphenoxy)phenyl)butanoate A mixture of ethyl 4-(2-hydroxyphenyl)butanoate (1.8 g, 8.64 mmol, 1 equiv.), 1,3-dibromo- 2-methylbenzene (4.32 g, 17.29 mmol, 2 equiv.), 2,2,6,6-tetramethylheptane-3,5-dione (398.19 mg, 2.16 mmol, 444.90 µL, 0.25 equiv.), CuI (823.06 mg, 4.32 mmol, 0.5 equiv.), and Cs2CO3 (3.38 g, 10.37 mmol, 1.2 equiv.) in NMP (20 mL) was degassed and purged with N2, and the mixture was stirred at 120 °C for 12 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure (3 g, crude) to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~1% ethyl acetate/petroleum ether) to give ethyl 4-(2-(3-bromo-2-methylphenoxy)phenyl)butanoate (1.9 g, 5.0 mmol, 63.3% yield) as a blackish green oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.37 (d, J = 8.0 Hz, 1H), 7.31 (d, J = 7.2 Hz, 1H), 7.23 - 7.16 (m, 1H), 7.14 - 7.06 (m, 2H), 6.74 - 6.66 (m, 2H), 3.99 (q, J = 7.2 Hz, 2H), 2.60 (t, J = 7.6 Hz, 2H), 2.33 - 2.26 (m, 5H), 1.88 -1.78 (m, 2H), 1.12 (t, J = 7.2 Hz, 3H) Step B. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-ethoxy-4-oxobutyl)ph enoxy)-2- methylphenyl)picolinate Ethyl 4-(2-(3-bromo-2-methylphenoxy)phenyl)butanoate (190.93 mg, 506.09 µmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)picolinate (310 mg, 506.09 µmol, 1 equiv.), K 2 CO 3 (209.83 mg, 1.52 mmol, 3 equiv.), and Ad 2 nBuP Pd G 3 (cataCXium® A Pd G 3 ) (36.86 mg, 50.61 µmol, 0.1 equiv.) were taken up into a microwave tube in dioxane (5 mL). The sealed tube was heated at 100 °C for 120 minutes under microwave. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc 25 mL × 3. The combined organic layers were washed with brine (15 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, petroleum ether: ethyl acetate = 1:1) to give compound tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-eth oxy-4-oxobutyl)phenoxy)-2- methylphenyl)picolinate (250 mg, 319.31 µmol, 63.09% yield) as a yellow solid. MS (ESI) m/z: 783.2 [M+H] + . Step C. Procedure for preparation of 4-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)phenyl)butanoic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(2-(4-ethoxy-4-oxobutyl)phenoxy)-2-methylphenyl)pic olinate (250 mg, 319.31 µmol, 1 equiv.) and LiOH·H 2 O (40.20 mg, 957.92 µmol, 3 equiv.) in THF (2 mL) and H 2 O (0.5 mL) was degassed and purged with N 2 and stirred at 25 °C for 1 hour under N 2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (25 mL × 3). The combined organic layers were washed with brine (15 mL × 3), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 4-(2-(3-(6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)phenyl)butanoic acid (150 mg, crude) as a yellow solid. MS (ESI) m/z: 755.5 [M+H] + Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-((3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H- indazol-7-yl)amino)-4-oxobutyl)phenoxy)-2-methylphenyl)picol inate A mixture of 4-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroiso quinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)phe nyl)butanoic acid (90 mg, 119.22 µmol, 1 equiv.), 3-(7-amino-1-methyl-1H-indazol-3-yl)piperidine-2,6-dione (33.87 mg, 131.14 µmol, 1.1 equiv.), DIEA (46.23 mg, 357.67 µmol, 62.30 uL, 3 equiv.), and HATU (54.40 mg, 143.07 µmol, 1.2 equiv.) in DMF (1.2 mL) was degassed and purged with N2 and stirred at 25 °C for 1 hour under N 2 atmosphere. The mixture was concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(4-((3-(2,6-dioxopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-4- oxobutyl)phenoxy)-2-methylphenyl)picolinate (100 mg, 100.5 µmol, 84.3% yield) as a yellow oil. MS (ESI) m/z: 995.2 [M+H] + . Step E. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-((3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H- indazol-7-yl)amino)-4-oxobutyl)phenoxy)-2-methylphenyl)picol inic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(2-(4-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-ind azol-7-yl)amino)-4- oxobutyl)phenoxy)-2-methylphenyl)picolinate (100 mg, 100.49 µmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL) was degassed and purged with N 2 and stirred at 35 °C for 1 hour under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-((3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H-indazol- 7-yl)amino)-4-oxobutyl)phenoxy)-2-methylphenyl)picolinic acid (24.52 mg, 25.9 µmol, 25.8% yield, 99.4% purity) as a yellow solid. MS (ESI) m/z: 939.4 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 12.86 - 12.68 (m, 1H), 10.89 (s, 1H), 9.86 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 7.6 Hz, 1H), 7.63 (d, J = 7.6 Hz, 1H), 7.57 (d, J = 6.4 Hz, 1H), 7.55 - 7.27 (m, 7H), 7.20 - 7.14 (m, 2H), 7.09 - 7.02 (m, 3H), 6.98 (d, J = 8.4 Hz, 1H), 6.88 (d, J = 7.2 Hz, 1H), 6.77 (d, J = 8.0 Hz, 1H), 6.63 (d, J = 8.0 Hz, 1H), 5.00 (s, 2H), 4.39 -4.32 (m, 1H), 3.98 (s, 3H), 3.93 (t, J = 5.6 Hz, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.75 (t, J = 7.6 Hz, 2H), 2.66 -2.59 (m, 2H), 2.45 (t, J = 7.2 Hz, 2H), 2.38 - 2.31 (m, 1H), 2.19 - 2.12 (m, 1H), 2.04 - 1.94 (m, 5H). Example 133. Preparation of Compound 129 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(4-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-4-oxobu tyl)phenoxy)-2- methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-((3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H- indazol-6-yl)amino)-4-oxobutyl)phenoxy)-2-methylphenyl)picol inate A mixture of 4-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroiso quinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)phe nyl)butanoic acid (100 mg, 105.98 µmol, 80% purity, 1 equiv.), 3-(6-amino-1-methyl-1H-indazol-3-yl)piperidine-2,6- dione (38.32 mg, 148.37 µmol, 1.4 equiv.), DIEA (41.09 mg, 317.93 µmol, 55.38 µL, 3 equiv.), and HATU (48.35 mg, 127.17 µmol, 1.2 equiv.) in DMF (1.4 mL) was degassed and purged with N2 and stirred at 25 °C for 2 hours under N2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with DCM (25 mL × 3). The combined organic layers were washed with brine (15 mL × 3), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-((3 -(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-4-oxobutyl)phenoxy)-2-methylph enyl)picolinate (100 mg, crude) as a brown oil. MS (ESI) m/z: 995.4 [M+H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-((3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H- indazol-6-yl)amino)-4-oxobutyl)phenoxy)-2-methylphenyl)picol inic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(2-(4-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-ind azol-6-yl)amino)-4- oxobutyl)phenoxy)-2-methylphenyl)picolinate (100 mg, 100.49 µmol, 1 equiv.) in TFA (0.5 mL) and DCM (0.5 mL) was degassed and purged with N 2 and stirred at 40 °C for 1 hour under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The crude product was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-((3 -(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-4-oxobutyl)phenoxy)-2-methylph enyl)picolinic acid (20.0 mg, 20.4 µmol, 20.3% yield, 95.7% purity) as a yellow solid. MS (ESI) m/z: 939.5 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.85 - 12.58 (m, 2H), 10.84 (s, 1H), 10.06 (s, 1H), 8.08 - 8.00 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.65 - 7.56 (m, 2H), 7.51 - 7.42 (m, 3H), 7.40 - 7.30 (m, 3H), 7.18 - 7.12 (m, 2H), 7.09 - 7.02 (m, 2H), 7.00 - 6.95 (m, 1H), 6.86 (d, J = 7.6 Hz, 1H), 6.77 (d, J = 8.0 Hz, 1H), 6.65 - 6.59 (m, 1H), 5.00 (s, 2H), 4.34 - 4.25 (m, 1H), 3.96 - 3.90 (m, 2H), 3.88 (s, 2H), 3.04 (t, J = 5.6 Hz, 2H), 2.75 - 2.57 (m, 4H), 2.41 (t, J = 7.6 Hz, 3H), 2.33 - 2.14 (m, 3H), 1.99 - 1.93 (m, 4H) Example 134. Preparation of Compound 132 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(4-(4-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-4-oxobu tyl)phenoxy)-2- methylphenyl)picolinic acid Step A. Procedure for preparation of methyl 4-(4-hydroxyphenyl)butanoate To a solution of 4-(4-hydroxyphenyl)butanoic acid (2 g, 11.10 mmol, 1 equiv.) in MeOH (25 mL) was added H2SO4 (23.55 g, 240.13 mmol, 12.8 mL, 21.64 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was diluted with H 2 O (50 mL) and extracted with ethyl acetate (50 mL × 2). The combined organic layers were washed with brine (20mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~20 % ethyl acetate/petroleum ether) to give methyl 4-(4-hydroxyphenyl)butanoate (1.7 g, 7.8 mmol, 70.9% yield, 90% purity) as a colorless oil. 1 H NMR (400 MHz, CDCl3) δ = 7.04 (d, J = 6.8 Hz, 2H), 6.78 - 6.75 (m, 2H), 3.69 - 3.67 (m, 3H), 2.58 (t, J = 7.2 Hz, 2H), 2.33 (dt, J = 1.6, 6.0 Hz, 2H), 1.98 - 1.91 (m, 2H). Step B. Procedure for preparation of methyl 4-(4-(3-bromo-2- methylphenoxy)phenyl)butanoate A mixture of methyl 4-(4-hydroxyphenyl)butanoate (500 mg, 2.57 mmol, 1 equiv.), 1,3- dibromo-2-methylbenzene (1.93 g, 7.72 mmol, 3 equiv.), CuI (245.14 mg, 1.29 mmol, 0.5 equiv.), Cs2CO3 (1.01 g, 3.09 mmol, 1.2 equiv.), and 2,2,6,6-tetramethylheptane-3,5-dione (118.60 mg, 643.58 µmol, 132.51 uL, 0.25 equiv.) in NMP (20 mL) was degassed and purged with N2 and stirred at 120 °C for 12 hours under N2 atmosphere. The reaction mixture was diluted with H2O (30 mL) and extracted with ethyl acetate (20 mL × 4). The combined organic layers were washed with brine (15mL × 2), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~7 % ethyl acetate/petroleum ether) to give methyl 4-(4-(3-bromo-2- methylphenoxy)phenyl)butanoate (900 mg, 2.2 mmol, 86.6% yield, 90% purity) as a yellow oil. 1 H NMR (400 MHz, CDCl3) δ = 7.36 - 7.32 (m, 1H), 7.13 (d, J = 8.4 Hz, 2H), 7.01 (t, J = 8.4 Hz, 1H), 6.88 - 6.82 (m, 3H), 3.68 (s, 3H), 2.63 (t, J = 7.6 Hz, 2H), 2.38 - 2.33 (m, 5H), 1.95 (q, J = 7.6 Hz, 2H). Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(4-methoxy-4-oxobutyl)p henoxy)-2- methylphenyl)picolinate A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)picolinat e (850 mg, 1.39 mmol, 1 equiv.) and methyl 4-(4-(3-bromo-2-methylphenoxy)phenyl)butanoate (672.08 mg, 1.67 mmol, 90% purity, 1.2 equiv.) in 1,4-dioxane (10 mL) was added Ad2nBuP Pd G3(cataCXium® A Pd G3 ) (101.06 mg, 138.77 µmol, 0.1 equiv.) and KF (1.5 M, 2.78 mL, 3 equiv.). After addition, the mixture was degassed and purged with N 2 and stirred at 100 °C for 2 hours under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~30 % Dichloromethane/Methanol) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(4-methoxy-4-oxobutyl)p henoxy)-2- methylphenyl)picolinate (520 mg, 635.7 µmol, 45.8% yield, 94% purity) as a yellow solid. MS (ESI) m/z: 769.9 [M+H] + . Step D. Procedure for preparation of 4-(4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2-methylphenoxy) phenyl)butanoic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(4-(4-methoxy-4-oxobutyl)phenoxy)-2-methylphe nyl)picolinate (520 mg, 635.70 µmol, 94% purity, 1 equiv.) in THF (5 mL) was added LiOH•H 2 O (80.03 mg, 1.91 mmol, 3 equiv.) and H2O (1.25 mL) at 25 °C. The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove THF. The aqueous phase was adjusted to pH = 4~5 with 1M HCl. The reaction mixture was filtered and diluted in ethyl acetate. The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give 4-(4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2-methylphenoxy) phenyl)butanoic acid (500 mg, crude) as a yellow solid. MS (ESI) m/z: 755.5 [M+H] + . Step E. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(4-((3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H- indazol-6-yl)amino)-4-oxobutyl)phenoxy)-2-methylphenyl)picol inate A mixture of 4-(4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3, 4-dihydroisoquinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)phe nyl)butanoic acid (100 mg, 132.47 µmol, 1 equiv.), HATU (55.41 mg, 145.72 µmol, 1.1 equiv.), and DIEA (51.36 mg, 397.41 µmol, 69.22 µL, 3 equiv.) in DMF (1 mL) was added 3-(6-amino-1-methyl-1H-indazol- 3-yl)piperidine-2,6-dione (34.21 mg, 132.47 µmol, 1 equiv.), and then the mixture was stirred at 25 °C for 2 hours. The reaction mixture was treated with water (2 mL), filtered, washed with CH2Cl2 (10 mL), and concentrated under the reduced pressure to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(4-(4-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-4-oxobu tyl)phenoxy)-2- methylphenyl)picolinate (103 mg, 83.8 µmol, 63.3% yield, 81% purity) as a yellow solid. MS (ESI) m/z: 995.5 [M+H] + . Step F. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(4-((3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H- indazol-6-yl)amino)-4-oxobutyl)phenoxy)-2-methylphenyl)picol inic acid tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(4-(4- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino )-4-oxobutyl)phenoxy)-2- methylphenyl)picolinate (100 mg, 100.49 µmol, 1 equiv.) in CH 2 Cl 2 (1.5 mL) and TFA (0.5 mL) was stirred at 40 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(4-(4-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-4-oxobu tyl)phenoxy)-2- methylphenyl)picolinic acid (28.7 mg, 29.9 µmol, 29.7% yield, 97.7% purity) as a white solid. MS (ESI) m/z: 939.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.88 (s, 1H), 10.08 (s, 1H), 8.08 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.64 - 7.58 (m, 2H), 7.52 ( d, J = 8.6 Hz, 1H), 7.49 - 7.44 (m, 2H), 7.39 - 7.33 (m, 2H), 7.24 - 7.11 (m, 4H), 7.07 (d, J = 8.8 Hz, 1H), 6.99 ( d, J = 8.4 Hz, 1H), 6.89 (d, J = 7.6 Hz, 1H), 6.83 ( d, J = 8.4 Hz, 3H), 4.99 ( s, 2H), 4.30 (dd, J = 4.8, 9.6 Hz, 1H), 3.96 - 3.92 (m, 2H), 3.90 (s, 3H), 3.03 ( t, J = 5.4 Hz, 2H), 2.70 - 2.64 (m, 2H), 2.40 - 2.35 (m, 2H), 2.35 (s, 2H), 2.24 - 2.08 (m, 2H), 1.93 - 1.88 (m, 5H) Example 135. Preparation of Compound 134 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(3-((4-(2,6- dioxopiperidin-3-yl)phenyl)amino)-3-oxopropyl)phenoxy)-2-met hylphenyl)picolinic acid Step A. Procedure for preparation of ethyl 3-(2-(3-bromo-2- methylphenoxy)phenyl)propanoate A mixture of ethyl 3-(2-hydroxyphenyl)propanoate (2 g, 10.30 mmol, 1 equiv.), 1,3-dibromo- 2-methylbenzene (5.15 g, 20.59 mmol, 2 equiv.), CuI (980.56 mg, 5.15 mmol, 0.5 equiv.), Cs 2 CO 3 (4.03 g, 12.36 mmol, 1.2 equiv.), and 2,2,6,6-tetramethylheptane-3,5-dione (474.38 mg, 2.57 mmol, 530.04 uL, 0.25 equiv.) in NMP (20 mL) was degassed and purged with N 2 and stirred at 120 °C for 12 hours under N2 atmosphere. The reaction mixture was diluted with H2O (50 mL) and extracted with EtOAc (100 mL × 3). The combined organic layers were washed with saturated NaHCO 3 solution (100 mL × 3), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~10% ethyl acetate/petroleum ether) to give ethyl 3-(2-(3- bromo-2-methylphenoxy)phenyl)propanoate (1.7 g, crude) as a green oil. 1 H NMR (400 MHz,CD 3 OD) δ = 7.37 - 7.27 (m, 2H), 7.22 - 7.13 (m, 1H), 7.10 - 7.01 (m, 2H), 6.75 - 6.63 (m, 2H), 4.11 - 4.04 (m, 2H), 2.96 (t, J = 7.6 Hz, 2H), 2.68 - 2.60 (m, 2H), 2.41 - 2.34 (m, 3H), 1.21 - 1.17 (m, 3H). Step B. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(3-ethoxy-3-oxopropyl)p henoxy)-2- methylphenyl)picolinate A mixture of ethyl 3-(2-(3-bromo-2-methylphenoxy)phenyl)propanoate (400 mg, 1.10 mmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)- 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)picolinate (674.53 mg, 1.10 mmol, 1 equiv.), di-tert-butyl(cyclopentyl)phosphane;dichloropalladium;iron (71.77 mg, 110.12 µmol, 0.1 equiv.), and K2CO3 (1.5 M, 2.20 mL, 3 equiv.) in dioxane (5.5 mL) was degassed and purged with N2 and stirred at 80 °C for 12 hours under N2 atmosphere. The reaction mixture was diluted with H 2 O (10 mL) and extracted with solvent (20 mL × 3). The combined organic layers were washed with brine (60 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~35% ethyl acetate/petroleum ether) to give tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(3-eth oxy-3-oxopropyl)phenoxy)-2- methylphenyl)picolinate (520 mg, 517.8 µmol, 47.0% yield, 76.5% purity) as a yellow oil. 1 H NMR (400 MHz, CDCl 3 ) δ = 7.34 (d, J = 8.0 Hz, 1H), 7.17 - 7.13 (m, 2H), 7.01 (t, J = 8.0 Hz, 1H), 6.86 - 6.81 (m, 3H), 3.69 (t, J = 6.4 Hz, 2H), 2.72 - 2.68 (m, 2H), 2.36 (s, 3H), 1.92 - 1.87 (m, 2H) Step C. Procedure for preparation of 3-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)phenyl)propanoic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(3-ethoxy-3-oxopropyl)phenoxy)-2-methylphe nyl)picolinate (520 mg, 517.89 µmol, 76.58% purity, 1 equiv.) in THF (3 mL) was added LiOH•H2O (1 M, 2.5 mL, 4.83 equiv.). The mixture was stirred at 25 °C for 12 hours. The mixture was concentrated under reduced pressure to remove THF, treated with HCl (1 M) to pH = 6, and filtered to give 3-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroiso quinolin-2(1H)-yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)phenyl)propanoi c acid (380 mg, crude) as a yellow solid. 1 H NMR (400 MHz, CDCl 3 ) δ = 9.87 - 9.80 (m, 1H), 7.38 - 7.31 (m, 1H), 7.16 - 7.13 (m, 2H), 7.05 -6.97 (m, 1H), 6.86 - 6.81 (m, 3H), 2.98 - 2.92 (m, 2H), 2.81 - 2.76 (m, 2H), 2.34 (s, 3H) Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(3-((4-(2,6-dioxopiperi din-3-yl)phenyl)amino)-3- oxopropyl)phenoxy)-2-methylphenyl)picolinate To a solution of 3-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroiso quinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)phe nyl)propanoic acid (90 mg, 121.48 µmol, 1 equiv.) in DMF (1 mL) was added HATU (69.29 mg, 182.22 µmol, 1.5 equiv.), DIEA (47.10 mg, 364.44 µmol, 63.48 µL, 3 equiv.), and 3-(4-aminophenyl)piperidine-2,6- dione (37.21 mg, 182.22 µmol, 1.5 equiv.). The mixture was stirred at 40 °C for 1 hour. The reaction was added into H2O (5 mL) and filtered to give tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(3-((4 -(2,6-dioxopiperidin-3- yl)phenyl)amino)-3-oxopropyl)phenoxy)-2-methylphenyl)picolin ate (100 mg, crude) as a yellow solid. MS (ESI) m/z: 927.7 [M+H] + Step E. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(3-((4-(2,6-dioxopiperi din-3-yl)phenyl)amino)-3- oxopropyl)phenoxy)-2-methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(3-((4-(2,6-dioxopiperidin-3-yl)phenyl)ami no)-3-oxopropyl)phenoxy)-2- methylphenyl)picolinate (100 mg, 107.87 µmol, 1 equiv.) in DCM (0.5 mL) was added TFA (770.00 mg, 6.75 mmol, 0.5 mL, 62.61 equiv.). The mixture was stirred at 25 °C for 12 hours. The mixture was concentrated under reduced pressure to remove DCM. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(3-((4-(2,6-dioxopiperidin-3-yl)phenyl)ami no)-3-oxopropyl)phenoxy)-2- methylphenyl)picolinic acid (23.9 mg, 26.7 µmol, 24.8% yield, 97.58% purity) as a yellow solid. MS (ESI) m/z: 871.4 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.91 - 12.82 (m, 1H), 12.80 - 12.58 (m, 1H), 10.80 (s, 1H), 9.94 - 9.89 (m, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.82 - 7.76 (m, 1H), 7.64 - 7.60 (m, 1H), 7.55 - 7.50 (m, 3H), 7.48 - 7.43 (m, 2H), 7.40 - 7.31 (m, 3H), 7.20 - 7.11 (m, 4H), 7.04 - 6.99 (m, 2H), 6.88 (d, J = 6.8 Hz, 1H), 6.81 (d, J = 7.6 Hz, 1H), 6.58 (d, J = 8.0 Hz, 1H), 5.00 (s, 2H), 3.95 - 3.91 (m, 2H), 3.80 - 3.77 (m, 1H), 3.07 - 3.01 (m, 4H), 3.01 - 2.95 (m, 4H), 2.70 - 2.69 (m, 1H), 2.17 - 2.12 (m, 1H), 1.95 (s, 3H) Example 136. Preparation of Compound 139 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(4-(3-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-3-oxopr opyl)phenoxy)-2- methylphenyl)picolinic acid Step A. Procedure for preparation of methyl 3-(4-(4-bromo-3- methylphenoxy)phenyl)propanoate To a solution of (4-bromo-3-methylphenyl)boronic acid (2.00 g, 9.31 mmol, 1 equiv.) in DCM (20 mL) was added methyl 3-(4-hydroxyphenyl)propanoate (2.01 g, 11.17 mmol, 1.2 equiv.) , TEA (941.94 mg, 9.31 mmol, 1.30 mL, 1 equiv. ) , 4A MS (20 mg, 465.44 umol), and Cu(OAc)2 (1.69 g, 9.31 mmol, 1 equiv. ) at 25 °C. The reaction mixture was purged with O2 and stirred at 25 °C for 40 hours under O2 (15 Psi). The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~10% Ethylacetate/petroleum ether). Methyl 3-(4-(4-bromo- 3-methylphenoxy)phenyl)propanoate (360 mg, 1.0 mmol, 11.0% yield) was obtained as an colorless oil. 1 H NMR (400 MHz, CDCl 3 ) δ = 7.45 (d, J = 8.8 Hz, 1H), 7.17 (d, J = 8.4 Hz, 2H), 6.95 - 6.90 (m, 2H), 6.89 (d, J = 2.8 Hz, 1H), 6.70 (dd, J = 2.8, 8.8 Hz, 1H), 3.69 (s, 3H), 2.95 (t, J = 8.0 Hz, 2H), 2.68 - 2.60 (m, 2H), 2.36 (s, 3H) Step B. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(4-(3-methoxy-3-oxopropyl) phenoxy)-2- methylphenyl)picolinate Methyl 3-(4-(4-bromo-3-methylphenoxy)phenyl)propanoate (330 mg, 944.97 µmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)picolinate (578.84 mg, 944.97 µmol, 1 equiv. ), KF (1.5 M, 1.89 mL, 3 equiv.), and [2-(2-aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-buty l- phosphane;methanesulfonate (137.64 mg, 188.99 µmol, 0.2 equiv. ) were taken up into a microwave tube in dioxane (3.3 mL). The sealed tube was heated at 100 °C for 1 hour under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~20% ethyl acetate/petroleum ether). tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(4-(3-methoxy-3-oxopropyl) phenoxy)-2- methylphenyl)picolinate (328 mg, 427.9 µmol, 45.2% yield, 98.5% purity) was obtained as a yellow oil. MS (ESI) m/z: 755.5[M+H] +. Step C. Procedure for preparation of 3-(4-(4-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-3- methylphenoxy)phenyl)propanoic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4-(4-(3-methoxy-3-oxopropyl)phenoxy)-2-methylphenyl)p icolinate (298 mg, 394.76 µmol, 1 equiv. ) , LiOH.H2O (82.83 mg, 1.97 mmol, 5 equiv. ) and H 2 O (1 mL) was added THF (3 mL). The mixture was stirred at 25 °C for 2 hours . The reaction mixture was concentrated under reduced pressure to give a residue. The residue was treated with water (4 mL). The pH was adjusted to around 3 by progressively adding diluted HCl. The mixture was filtered to give a residue. 3-(4-(4-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-3- methylphenoxy)phenyl)propanoic acid (278 mg, 339.2 µmol, 85.9% yield, 90.4% purity) was obtained as a yellow solid. MS (ESI) m/z: 741.4[M+H] +. Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(4-(3-((3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H- indazol-6-yl)amino)-3-oxopropyl)phenoxy)-2-methylphenyl)pico linate A mixture of 3-(4-(4-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroiso quinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-3-methylphenoxy)phe nyl)propanoic acid (119 mg, 160.62 µmol, 1 equiv.), 3-(6-amino-1-methyl-1H-indazol-3-yl)piperidine-2,6-dione (49.78 mg, 192.75 µmol, 1.2 equiv. ) in pyridine (2 mL) was added EDCI (46.19 mg, 240.93 µmol, 1.5 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was added into H 2 O (5 mL), filtered, and concentrated under reduced pressure to give a residue. The crude product tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- (4-(4-(3-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6 -yl)amino)-3- oxopropyl)phenoxy)-2-methylphenyl)picolinate (180 mg, crude) was obtained as a yellow oil and used into the next step without further purification. MS (ESI) m/z: 982.5[M+H] +. Step E. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(4-(3-((3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H- indazol-6-yl)amino)-3-oxopropyl)phenoxy)-2-methylphenyl)pico linic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(4-(3-((3-(2,6-dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-3- oxopropyl)phenoxy)-2-methylphenyl)picolinate (160 mg, 163.08 µmol, 1 equiv. ) in CH2Cl2 (1.5 mL) was added TFA (2.05 g, 18.01 mmol, 1.33 mL, 110.43 equiv.). The mixture was stirred at 40 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC.6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(4-(4-(3-((3-(2,6-dioxopi peridin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-3-oxopropyl)phenoxy)-2-methylphenyl)pico linic acid (76.1 mg, 80.0 µmol, 49.1% yield, 97.4% purity) was obtained as a yellow solid. MS (ESI) m/z: 925.5[M+H] +. 1 HNMR (400 MHz, DMSO-d 6 ) δ = 13.02 - 12.46 (m, 2H), 10.90 - 10.84 (m, 1H), 10.11 (s, 1H), 8.10 - 8.00 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.65 - 7.58 (m, 2H), 7.50 - 7.43 (m, 3H), 7.40 - 7.33 (m, 2H), 7.29 (d, J = 8.4 Hz, 2H), 7.07 (dd, J = 1.2, 8.8 Hz, 1H), 7.01 - 6.93 (m, 4H), 6.84 (d, J = 2.4 Hz, 1H), 6.73 (dd, J = 2.4, 8.4 Hz, 1H), 4.98 (s, 2H), 4.30 (dd, J = 5.2, 9.6 Hz, 1H), 3.95 - 3.87 (m, 5H), 3.02 (t, J = 5.6 Hz, 2H), 2.93 (t, J = 7.6 Hz, 2H), 2.71 - 2.61 (m, 4H), 2.36 - 2.30 (m, 1H), 2.20 - 2.14 (m, 1H), 2.01 (s, 3H) Example 137. Preparation of Compound 147a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pip erazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of 3-((1r, 4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propan-1-ol To a solution of ethyl 3-((1r, 4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propanoate (300 mg, 812.36 µmol, 1 equiv.) in THF (5 mL) was added LiAlH 4 (37.00 mg, 974.84 µmol, 1.2 equiv.) under 0 °C. Then the mixture was stirred at 25 °C for 1.5 hours. The reaction mixture was quenched by addition H2O 0.5 mL, 15% NaOH 0.5 mL, H2O 1.5 mL, and then extracted with EtOAc 3 mL (1 mL × 3). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give 3-((1r, 4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propan-1-ol (277 mg, crude) as a white solid. Step B. Procedure for preparation of 3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propanal To a solution of DMSO (264.53 mg, 3.39 mmol, 264.53 µL, 4 equiv.) in DCM (2 mL) was added dropwise a solution of (COCl)2 (214.88 mg, 1.69 mmol, 148.19 µL, 2 equiv.) in DCM (2 mL) at -70 °C under N2 atmosphere. The mixture was stirred at -70 °C for 1 hour. After which time, 3-((1r, 4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propan-1-ol (277 mg, 846.43 µmol, 1 equiv.) in DCM (2 mL) was added dropwise at -70 °C. The solution was stirred for 1 hour at -70 °C. Then TEA (513.90 mg, 5.08 mmol, 706.88 µL, 6 equiv.) was added into the solution. The solution was stirred at -70 °C for 0.5 hour under N 2 atmosphere. The reaction mixture was quenched by addition of H2O (10 mL), and then diluted with DCM (10 mL) and extracted with DCM (10 mL × 3). The combined organic layers were washed with H2O (5 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a yellow residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 10 % ethyl acetate / petroleum ether) to give 3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propanal (220 mg, 676.42 µmol, 79.91% yield) as a yellow solid. Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(2-methyl-3-(((1r,4r)-4-(3 oxopropyl)cyclohexyl)oxy)phenyl)picolinate A mixture of 3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propanal (220 mg, 676.42 µmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pic olinate (497.21 mg, 811.71 µmol, 1.2 equiv.), [2-(2-aminophenyl)phenyl]palladium(1 + );bis(1-adamantyl)-butyl- phosphane;methanesulfonate (98.52 mg, 135.28 µmol, 0.2 equiv.), and K2CO3 (1.5 M, 676.42 µL, 1.5 equiv.) in dioxane (5 mL) was degassed and purged with N2 and stirred at 100 °C for 1 hour under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~10% ethyl acetate/petroleum ether) to give tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(2-methyl-3- (((1r,4r)-4-(3 oxopropyl)cyclohexyl)oxy)phenyl)picolinate (280 mg, 383.08 µmol, 56.63% yield) as a white solid. MS (ESI) m/z: 731.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.86 - 12.82 (m, 1H), 9.94 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.69 - 7.64 (m, 1H), 7.57 (d, J = 8.0 Hz, 1H), 7.50 - 7.44 (m, 2H), 7.41 - 7.32 (m, 4H), 7.20 (d, J = 7.2 Hz, 1H), 7.06 (d, J = 8.4 Hz, 1H), 5.07 - 4.89 (m, 3H), 2.98 (t, J = 6.0 Hz, 2H), 1.99 (s, 3H), 1.36 (s, 2H), 1.33 (s, 4H), 1.24 (s, 9H), 1.16 (s, 9H). Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)propyl)cyclohexyl)oxy) -2- methylphenyl)picolinate A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(2-methyl-3-(((1r,4r)-4-(3 oxopropyl)cyclohexyl)oxy)phenyl)picolinate (80 mg, 109.45 µmol, 1 equiv.), 3-(1-methyl-7-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (43.00 mg, 131.34 µmol, 1.2 equiv.), and NaBH(OAc) 3 (69.59 mg, 328.36 µmol, 3 equiv.) in DCM (2 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 0 °C for 1.5 hour under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-7- yl)piperazin-1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl)pico linate (100 mg, 95.9 µmol, 87.6% yield) as a white solid. Step E. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)propyl)cyclohexyl)oxy) -2-methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-7- yl)piperazin-1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl)pico linate (80 mg, 76.75 µmol, 1 equiv.) in DCM (1 mL) and TFA (1 mL). The mixture was stirred at 40 °C for 3 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-7- yl)piperazin-1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl)pico linic acid (45.5 mg, 44.3 µmol, 57.7% yield, 95.9% purity) as a white solid. MS (ESI) m/z: 986.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.91 - 12.83 (m, 1H), 10.89 (s, 1H), 8.14 (s, 1H), 8.04 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 8.0 Hz, 1H), 7.51 - 7.34 (m, 7H), 7.08 - 7.03 (m, 3H), 6.95 (d, J = 15.6 Hz, 2H), 6.62 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.34 (d, J = 9.6 Hz, 1H), 4.25 (s, 3H), 3.93 (t, J = 5.6 Hz, 2H), 3.03 (t, J = 5.6 Hz, 4H), 2.95 - 2.81 (m, 3H), 2.70 - 2.61 (m, 4H), 2.36 - 2.32 (m, 3H), 2.20 - 2.16 (m, 1H), 2.10 - 2.08 (m, 2H), 1.88 (s, 3H), 1.81 (d, J = 11.2 Hz, 2H), 1.53 (d, J = 8.0 Hz, 2H), 1.39 - 1.24 (m, 6H), 1.17 - 1.06 (m, 3H). Example 138. Preparation of Compound 156 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-((7-(2-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2-oxoet hyl)-7-azaspiro[3.5]nonan-2- yl)methoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 2-((3-bromo-2-methylphenoxy)methyl)-7- azaspiro[3.5]nonane-7-carboxylate A mixture of tert-butyl 2-(hydroxymethyl)-7-azaspiro[3.5]nonane-7-carboxylate (1 g, 3.92 mmol, 1.2 equiv.), 3-bromo-2-methylphenol (610.38 mg, 3.26 mmol, 1 equiv.), and 2-(tributyl- λ 5 -phosphanylidene)acetonitrile (1.18 g, 4.90 mmol, 1.5 equiv.) in toluene (10 mL) was stirred at 120 °C for 2 h under N2 atmosphere. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 8% ethyl acetate / petroleum ether) to give tert-butyl 2-((3- bromo-2-methylphenoxy)methyl)-7-azaspiro[3.5]nonane-7-carbox ylate (1.34 g, 3.16 mmol, 96.76% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.16 (d, J = 8.0 Hz,1H), 7.09 (t, J = 8.0 Hz, 1H), 6.96 (d, J = 8.0 Hz, 1H), 3.95 ( d, J = 6.2 Hz, 2H), 3.29 - 3.25 (m, 2H), 3.22 - 3.15 (m, 2H), 2.77 - 2.64 (m, 1H), 2.24 (s, 3H), 1.94 - 1.86 (m, 2H), 1.69 - 1.62 (m, 2H), 1.56 - 1.50 (m, 2H), 1.44 - 1.40 (m, 2H), 1.38 (s, 9H) Step B. Procedure for preparation of 2-((3-bromo-2-methylphenoxy)methyl)-7- azaspiro[3.5]nonane A mixture of tert-butyl 2-((3-bromo-2-methylphenoxy)methyl)-7-azaspiro[3.5]nonane-7- carboxylate (1.34 g, 3.16 mmol, 1 equiv.) in HCl / dioxane (15 mL) was stirred at 25 °C for 1 h. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was used in the next step without other purification. The compound 2- ((3-bromo-2-methylphenoxy)methyl)-7-azaspiro[3.5]nonane (1.32 g, crude) was obtained as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ = 9.0 - 8.75(m, 2H), 7.15 (d, J = 7.2 Hz, 1H), 7.09 (t, J = 8.0 Hz, 1H), 6.96 (d, J = 8.0 Hz, 1H), 3.95 (d, J = 6.2 Hz, 2H), 3.02 - 2.93(m, 2H), 2.93 - 2.84 (m, 2H), 2.74 - 2.63 (m, 1H), 2.23 (s, 3H), 2.00 - 1.92 (m, 2H), 1.82 - 1.77 (m, 2H), 1.74 - 1.66 (m, 4H) Step C. Procedure for preparation of ethyl 2-(2-((3-bromo-2-methylphenoxy)methyl)-7- azaspiro[3.5]nonan-7-yl)acetate A mixture of 2-((3-bromo-2-methylphenoxy)methyl)-7-azaspiro[3.5]nonane (1.5 g, 4.63 mmol, 1.0 equiv.), ethyl 2-bromoacetate (772.54 mg, 4.63 mmol, 511.62 µL, 1.0 equiv.), KI (383.96 mg, 2.31 mmol, 0.5 equiv.), and K 2 CO 3 (1.92 g, 13.88 mmol, 3.0 equiv.) in CH 3 CN (15 mL) was stirred at 60 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The mixture was purified by flash silica gel chromatography (Eluent of 0 ~ 18% ethyl acetate / petroleum ether) to give ethyl 2-(2-((3- bromo-2-methylphenoxy)methyl)-7-azaspiro[3.5]nonan-7-yl)acet ate (1.8 g, 4.4 mmol, 94.8% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.14 (d, J = 7.6 Hz, 1H), 7.07 (t, J = 8.0 Hz, 1H), 6.94 (d, J = 7.6 Hz, 1H), 4.06 (q, J = 7.2 Hz, 2H), 3.92 (d, J = 6.0 Hz, 2H), 3.14 (s, 2H), 2.70 - 2.61 (m, 1H), 2.45 - 2.40 (m, 2H), 2.36 - 2.31 (m, 2H), 2.23 (s, 3H), 1.89 - 1.83 (m, 2H), 1.64 - 1.57 (m, 4H), 1.50 - 1.47 (m, 2H), 1.18 (t, J = 7.2 Hz, 3H) Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-((7-(2-ethoxy-2-oxoethyl)- 7-azaspiro[3.5]nonan-2- yl)methoxy)-2-methylphenyl)picolinate A mixture of ethyl 2-(2-((3-bromo-2-methylphenoxy)methyl)-7-azaspiro[3.5]nonan- 7- yl)acetate (750 mg, 1.83 mmol, 1.0 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3, 2-dioxaborolan-2-yl)pyridine-2- carboxylate (1.34 g, 2.19 mmol, 1.2 equiv.), [2-(2-aminophenyl)phenyl]palladium(1 + );bis(1- adamantyl)-butyl-phosphane;methanesulfonate (133.11 mg, 182.77 µmol, 0.1 equiv.), and K2CO3 (1.5 M, 1.83 mL, 1.5 equiv.) in dioxane (7.5 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 100 °C for 1 hour under microwave. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The mixture was purified by flash silica gel chromatography (Eluent of 48% ethyl acetate / petroleum ether) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-((7-(2-ethoxy-2-oxoethyl)- 7-azaspiro[3.5]nonan-2- yl)methoxy)-2-methylphenyl)picolinate (2.5 g, 3.0 mmol, 83.8% yield) as a yellow solid. MS (ESI) m/z: 816.4[M+H] + . Step E. Procedure for preparation of 2-(2-((3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)methyl)-7-azaspiro[3.5]nonan-7-yl)acetic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-((7-(2-ethoxy-2-oxoethyl)-7-azaspiro[3.5]nonan-2-yl )methoxy)-2- methylphenyl)picolinate (1.9 g, 2.33 mmol, 1.0 equiv.) and LiOH•H 2 O (488.50 mg, 11.64 mmol, 5 equiv.) in H 2 O (7 mL) and THF (14 mL) was stirred at 25 °C for 16 hours. The reaction mixture was diluted with H2O (5 mL) and concentrated as a turbid liquid. Then the pH of the turbid liquid was adjusted to 3 with 1 N HCl (5 mL), the residue was extracted with DCM / MeOH(20:1). The combined organic layers were filtered and concentrated to give 2-(2-((3-(6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2( 1H)-yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)methyl)-7-azasp iro[3.5]nonan-7-yl)acetic acid (2.1 g, crude) was obtained as a yellow solid. MS (ESI) m/z: 788.3 [M+H] + . Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-((7-(2-((3-(2,6-dioxopiper idin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)-7-azaspiro[3.5]nonan-2-yl)me thoxy)-2- methylphenyl)picolinate To a solution of 2-(2-((3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydrois oquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)methyl)-7- azaspiro[3.5]nonan-7-yl)acetic acid (80 mg, 101.53 µmol, 1 equiv.) and 3-(6-amino-1-methyl- indazol-3-yl)piperidine-2,6-dione (31.47 mg, 121.83 umol, 1.2 equiv.) in pyridine (0.5 mL) was added EDCI (29.19 mg, 152.29 µmol, 1.5 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was quenched by the addition of water (1 mL). The organic phase was separated, extracted with ethyl acetate (1 mL × 3), and concentrated under reduced pressure to give a residue to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-((7-(2-((3-(2,6-dioxopiper idin-3-yl)-1-methyl-1H-indazol- 6-yl)amino)-2-oxoethyl)-7-azaspiro[3.5]nonan-2-yl)methoxy)-2 -methylphenyl)picolinate (150 mg, crude) as a brown oil. MS (ESI) m/z: 1028.7 [M+H] + Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-((7-(2-((3-(2,6-dioxopiper idin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)-7-azaspiro[3.5]nonan-2-yl)me thoxy)-2- methylphenyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-((7-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-in dazol-6-yl)amino)-2-oxoethyl)- 7-azaspiro[3.5]nonan-2-yl)methoxy)-2-methylphenyl)picolinate (80 mg, 77.80 umol, 1 equiv.) in DCM (1 mL) and TFA (1.54 g, 13.51 mmol, 1 mL, 173.59 equiv.) was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-((7-(2-((3-(2,6-dioxopiper idin-3-yl)-1-methyl-1H-indazol- 6-yl)amino)-2-oxoethyl)-7-azaspiro[3.5]nonan-2-yl)methoxy)-2 -methylphenyl)picolinic acid (34.54 mg, 35.14 µmol, 45.16% yield, 98.9% purity) as a white solid. MS (ESI) m/z: 972.7 [M+H] + . 1 HNMR (400 MHz, DMSO-d 6 ) δ = 10.88 (s, 1H), 9.83 (s, 1H), 8.15 (s, 1H), 8.04 - 8.01 (m, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 8.4 Hz, 2H), 7.49 - 7.40 (m, 3H), 7.40 - 7.32 (m, 2H), 7.24 - 7.05 (m, 2H), 6.95 (d, J = 8.8 Hz, 1H), 6.88 (d, J = 8.4 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 1H), 4.32 (dd, J = 5.6, 9.6 Hz, 1H), 3.95 - 3.88 (m, 7H), 3.10 (s, 2H), 3.02 (t, J = 5.5 Hz, 2H), 2.70 - 2.60 (m, 4H), 2.39 - 2.30 (m, 4H), 2.21 - 2.13 (m, 1H), 1.94 - 1.86 (m, 5H), 1.73 - 1.64 (m, 4H), 1.60 (s, 2H). Example 139. Preparation of Compound 157 2-(5-(1-(((3r,5r,7r)-adamantan-1-yl)methyl)-5-methyl-1H-pyra zol-4-yl)-6-((8-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-8-oxooc tyl)carbamoyl)pyridin-2-yl)- N-(benzo[d]thiazol-2-yl)-1,2,3,4-tetrahydroisoquinoline-8-ca rboxamide Step A. Procedure for preparation of 2-(5-(1-(((3r,5r,7r)-adamantan-1-yl)methyl)-5- methyl-1H-pyrazol-4-yl)-6-((8-((3-(2,6-dioxopiperidin-3-yl)- 1-methyl-1H-indazol-6- yl)amino)-8-oxooctyl)carbamoyl)pyridin-2-yl)-N-(benzo[d]thia zol-2-yl)-1,2,3,4- tetrahydroisoquinoline-8-carboxamide A mixture of 8-[[3-[1-(1-adamantylmethyl)-5-methyl-pyrazol-4-yl]-6-[8-(1, 3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]pyridine-2-carb onyl]amino]octanoic acid (110 mg, 137.50 µmol, 1 equiv.), 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (42.61 mg, 165.00 µmol, 1.2 equiv.), and HATU (62.74 mg, 165.00 µmol, 1.2 equiv.) in DMF (1.4 mL) was degassed and purged with N2 and stirred at 25 °C for 5 minutes. After 5 minutes, to the mixture was added DIPEA (53.31 mg, 412.49 µmol, 71.85 µL, 3 equiv.). The mixture was stirred at 25 °C for 1 hour under N2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with DCM (25 mL × 3). The combined organic layers were washed with brine (15 mL × 3), dried over anhydrous sodium sulfate filtered, and concentrated under reduced pressure to give a residue. The compound 2-(5-(1-(((3r,5r,7r)-adamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-((8-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-8-oxooctyl)carbamoyl)pyridin-2-yl)-N-(be nzo[d]thiazol-2-yl)-1,2,3,4- tetrahydroisoquinoline-8-carboxamide (139.8 mg, 128.6 µmol, 93.5% yield, 95.7% purity) was obtained as a yellow solid. MS (ESI) m/z: 520.9 [M+H] + 1 H NMR (400 MHz, CD3OD) δ = 8.33 (t, J = 6.4 Hz, 1H), 8.05 (s, 1H), 7.91 (d, J = 8.0 Hz, 1H), 7.79 - 7.76 (m, 1H), 7.60 (d, J = 8.8 Hz, 1H), 7.60 -7.53 (m, 1H), 7.47 - 7.43 (m, 2H), 7.33 (t, J = 4.0 Hz, 3H), 7.05 (dd, J =8.4, 1.2 Hz, 1H), 6.99 (d, J = 8.8 Hz, 1H), 5.09 (s, 2H), 4.33 (q, J = 5.2 Hz, 1H), 3.93 (s, 3H), 3.90 (t, J = 6.4 Hz, 2H), 3.72 (s, 2H), 3.21 - 3.11 (m, 3H), 3.04 (t, J = 6.0 Hz, 2H), 2.81 - 2.70 (m, 2H), 2.37 (t, J = 7.6 Hz, 2H), 2.09 (s, 3H), 1.98 - 1.92 (m, 4H), 1.74 - 1.69 (m, 4H), 1.65 (s, 4H), 1.62 -1.61 (m, 2H), 1.60 -1.57 (m, 6H), 1.47 - 1.43 (m, 2H), 1.31- 1.28 (m, 4H) Example 140. Preparation of Compound 158 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-((3- (2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-7-yl)amino)-2-oxoethyl)piperidin- 4- yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 4-(3-(3-bromo-2- methylphenoxy)propyl)piperidine-1-carboxylate A mixture of 3-bromo-2-methyl-phenol (2 g, 10.69 mmol, 1 equiv.), K2CO3 (4.43 g, 32.08 mmol, 3 equiv.), and tert-butyl 4-(3-hydroxypropyl)piperidine-1-carboxylate (3.27 g, 10.69 mmol, 1 equiv.) in CH3CN (20 mL) was degassed and purged with N2 and stirred at 60 °C for 12 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~7% ethyl acetate/petroleum ether) to give tert-butyl 4-(3-(3-bromo-2- methylphenoxy)propyl)piperidine-1-carboxylate (1.8 g, 4.3 mmol, 40.8% yield) as a colorless oil 1 H NMR (400 MHz, CDCl 3 ) δ = 7.09 - 7.07 (m, 1H), 6.91 (t, J = 8.0 Hz, 1H), 6.68 (s, 1H), 3.86 (t, J = 6.4 Hz, 2H), 2.63 (t, J = 12.0 Hz, 2H), 2.23 (s, 3H), 1.79 - 1.69 (m, 2H), 1.69 - 1.50 (m, 4H), 1.39 (s, 9H), 1.38 - 1.30 (m, 3H), 1.11 - 0.98 (m, 2H) Step B. Procedure for preparation of 4-(3-(3-bromo-2-methylphenoxy)propyl)piperidine A mixture of tert-butyl 4-(3-(3-bromo-2-methylphenoxy)propyl)piperidine-1-carboxylat e (1.8 g, 4.37 mmol, 1 equiv.) in HCl/EtOAc (20 mL) was stirred at 20 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give 4-(3-(3-bromo-2- methylphenoxy)propyl)piperidine (1.5 g, crude) as a white solid. MS (ESI) m/z: 313.9 [M+3] + . Step C. Procedure for preparation of ethyl 2-(4-(3-(3-bromo-2- methylphenoxy)propyl)piperidin-1-yl)acetate A mixture of 4-(3-(3-bromo-2-methylphenoxy)propyl)piperidine (1 g, 3.20 mmol, 1 equiv.), ethyl 2-bromoacetate (534.84 mg, 3.20 mmol, 354.20 µL, 1 equiv.) and K 2 CO 3 (885.24 mg, 6.41 mmol, 2 equiv.) in CH3CN (15 mL) was degassed and purged with N2 and stirred at 60 °C for 2 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~45% ethyl acetate/petroleum ether) to give ethyl 2-(4-(3-(3-bromo-2- methylphenoxy)propyl)piperidin-1-yl)acetate (0.5 g, 1.2 mmol, 39.1% yield) as a yellow oil MS (ESI) m/z: 399.9 [M+3] + . 1 H NMR (400 MHz, CD 3 OD) δ = 7.14 (d, J = 8.0 Hz, 1H), 6.98 (t, J = 8.0 Hz, 1H), 6.75 (d, J = 8.0 Hz, 1H), 4.19 (q, J = 7.2 Hz, 2H), 3.93 (t, J = 6.4 Hz, 2H), 3.23 - 3.18 (m, 2H), 2.95 (d, J = 11.6 Hz, 2H), 2.31 (s, 3H), 2.18 - 2.11 (m, 2H), 1.84 - 1.79 (m, 2H), 1.72 - 1.64 (m, 4H), 1.59 - 1.53 (m, 2H), 1.46 - 1.40 (m, 4H), 1.35 (dd, J = 3.2, 10.8 Hz, 2H), 1.28 (t, J = 7.2 Hz, 3H) Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-ethoxy-2-oxoethyl )piperidin-4-yl)propoxy)-2- methylphenyl)picolinate A mixture of ethyl 2-(4-(3-(3-bromo-2-methylphenoxy)propyl)piperidin-1-yl)aceta te (500 mg, 1.26 mmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)picolinate (768.89 mg, 1.26 mmol, 1 equiv.), [2-(2-aminophenyl)phenyl]palladium(1+);bis(1- adamantyl)-butyl-phosphane;methanesulfonate (91.41 mg, 125.52 µmol, 0.1 equiv.) and K3PO4 (1.5 M, 2.51 mL, 3 equiv.) in 1,4-dioxane (5 mL) was degassed and purged with N2 and stirred at 80 °C for 12 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~5% ethyl acetate/petroleum ether) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-(1-(2-ethoxy-2- oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinate (800 mg, 696.5 µmol, 55.4% yield, 70% purity) as a yellow solid. MS (ESI) m/z: 804.5 [M+H] + . Step E. Procedure for preparation of 2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyri din-3-yl)-2- methylphenoxy)propyl)piperidin-1-yl)acetic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-ethoxy-2-oxoethyl)piperidin-4-yl)pro poxy)-2- methylphenyl)picolinate (342.86 mg, 298.51 µmol, 70% purity, 1 equiv.) in THF (3 mL) was added LiOH (1 M, 895.52 µL, 3 equiv.).The mixture was stirred at 20 °C for 1 hour. The reaction mixture was adjusted to pH = 5, filtered, and concentrated under reduced pressure to give 2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin-2(1H)-yl)-2- (tert-butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)propyl)pi peridin-1-yl)acetic acid (150 mg, 193.3 µmol, 64.7% yield) as a yellow solid. MS (ESI) m/z: 776.6 [M+H] + Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-7-yl)amino)-2-oxoethyl)piperidin-4-yl)propoxy)-2-met hylphenyl)picolinate A mixture of 2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)pro pyl)piperidin-1-yl)acetic acid (70 mg, 90.21 µmol, 1 equiv.), 3-(7-amino-1-methyl-1H-indazol-3-yl)piperidine-2,6-dione (23.30 mg, 90.21 µmol, 1 equiv.), HATU (37.73 mg, 99.23 µmol, 1.1 equiv.), and TEA (27.39 mg, 270.64 µmol, 37.67 µL, 3 equiv.) in DMF (1 mL) was degassed and purged with N2 and stirred at 40 °C for 12 hours under N 2 atmosphere. The solution was poured into water (2 mL) and filtered to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-meth yl-1H-indazol-7-yl)amino)-2- oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinate (60 mg, 59.0 µmol, 65.4% yield) as a yellow solid. MA (ESI) m/z: 508.9 [M/2+H] + Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-((3-(2,6-dioxopip eridin -3-yl)-1-methyl-1H- indazol-7-yl)amino)-2-oxoethyl)piperidin-4-yl)propoxy)-2-met hylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-meth yl-1H-indazol-7-yl)amino)-2- oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinate (60 mg, 59.04 µmol, 1 equiv.) in DCM (0.5 mL) was added TFA (0.5 mL).The mixture was stirred at 40 °C for 0.5 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-((3-(2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-7-yl)amino)-2- oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinic acid (29.1 mg, 29.4 µmol, 49.8% yield, 96.8% purity) as a yellow solid MS (ESI) m/z: 960.6 [M+1] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.15 - 12.41 (m, 1H), 10.94 - 10.81 (m, 1H), 9.92 - 9.73 (m, 1H), 8.03 (d, J = 7.2 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 6.4 Hz, 1H), 7.60 - 7.54 (m, 1H), 7.52 - 7.20 (m, 7H), 7.13 - 7.05 (m, 2H), 7.01 - 6.93 (m, 1H), 6.88 (d, J = 8.0 Hz, 1H), 6.64 (d, J = 7.2 Hz, 1H), 4.99 (s, 2H), 4.38 (dd, J = 4.8, 9.2 Hz, 1H), 4.12 (s, 3H), 4.03 - 3.85 (m, 4H), 3.03 (s, 4H), 2.98 - 2.94 (m, 2H), 2.69 - 2.62 (m, 2H), 2.22 - 2.17 (m, 2H), 1.90 (s, 3H), 1.83 - 1.67 (m, 5H), 1.46 - 1.26 (m, 6H) Example 141. Preparation of Compound 159a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-((3R,5S)-4- (2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)am ino)-2-oxoethyl)-3,5- dimethylpiperazin-1-yl)ethoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of ethyl 2-[(2R, 6S)-4-[2-(3-bromo-2-methyl- phenoxy)ethyl]-2,6-dimethyl-piperazin-1-yl]acetate A mixture of (3R,5S)-1-[2-(3-bromo-2-methyl-phenoxy)ethyl]-3,5-dimethyl-p iperazine (2.7 g, 8.25 mmol, 1 equiv.), ethyl 2-bromoacetate (3.44 g, 20.63 mmol, 2.28 mL, 2.5 equiv.), and Et 3 N (3.34 g, 33.00 mmol, 4.59 mL, 4 equiv.) in DMF (10 mL) was degassed and purged with N2 and stirred at 50 °C for 4 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , dichloromethane: methanol=1/0 to 5/1). The compound ethyl 2-[(2R, 6S)-4-[2-(3-bromo-2-methyl-phenoxy)ethyl]-2,6-dimethyl-piper azin-1-yl]acetate (1.2 g, 2.9 mmol, 35.1% yield) was obtained as a white solid. MS (ESI) m/z: 414.9 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ = 7.20 (d, J = 8.0 Hz, 1H), 7.02 (t, J = 8.0 Hz, 1H), 6.79 (d, J = 8.0 Hz, 1H), 4.38 (s, 2H), 4.20 (q, J = 6.8 Hz,, 2H), 3.56 (s, 2H), 3.42 - 3.40 (m, 2H), 3.24 - 3.18 (m, 4H), 2.53 - 2.46 (m, 2H), 2.30 (s, 3H), 1.31 (t, J = 7.2 Hz, 3H), 1.14 (d, J = 6.0 Hz, 6H). Step B. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[(3S,5R)-4-(2-ethox y-2-oxo-ethyl)-3,5-dimethyl- piperazin-1-yl]ethoxy]-2-methyl-phenyl]pyridine-2-carboxylat e To a solution of ethyl 2-[(2R, 6S)-4-[2-(3-bromo-2-methyl-phenoxy)ethyl]-2,6-dimethyl- piperazin-1-yl]acetate (1.2 g, 2.90 mmol, 1 equiv.) and tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tet ramethyl-1,3,2-dioxaborolan-2- yl)pyridine-2-carboxylate (1.96 g, 3.19 mmol, 1.1 equiv.) in 1,4-dioxane (8 mL) was added KF (1.5 M, 5.81 mL, 3 equiv.) and ditert-butyl(cyclopentyl)phosphane;dichloropalladium;iron (189.21 mg, 290.31 µmol, 0.1 equiv.). The mixture was stirred at 100 °C for 1 hour under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=1/1). The compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[2-[(3S,5R)-4-(2-ethoxy-2-oxo-ethy l)-3,5-dimethyl-piperazin-1- yl]ethoxy]-2-methyl-phenyl] pyridine-2-carboxylate (2 g, 2.4 mmol, 84.1% yield) was obtained as a yellow solid. MS (ESI) m/z: 819.3 [M+H] + . Step C. Procedure for preparation of 2-((2R,6S)-4-(2-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)ethyl)-2,6-dimethylpiperazin-1-yl)acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[2-[(3S,5R)-4-(2-ethoxy-2-oxo-ethyl)-3,5-dimethyl -piperazin-1-yl]ethoxy]-2- methyl-phenyl]pyridine-2-carboxylate (2 g, 2.44 mmol, 1 equiv.) in THF (10 mL) was added LiOH•H2O (1 M, 7.33 mL, 3 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to remove THF, and then acidified with HCl (1M), filtered, and concentrated under reduced pressure to give a residue. The compound 2- [(2S,6R)-4-[2-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-2- tert-butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy] ethyl]-2,6-dimethyl-piperazin-1-yl]acetic acid (1.9 g, 2.4 mmol, 98.3% yield) was obtained as a brown solid. MS (ESI) m/z: 791.5 [M+H] + Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((3R,5S)-4-(2-((3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)-3,5-dimethylpipera zin-1-yl)ethoxy)-2- methylphenyl)picolinate A mixture of 2-((2R,6S)-4-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4 - dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2-methylphenoxy)ethyl)- 2,6-dimethylpiperazin-1-yl)acetic acid (160 mg, 202.28 µmol, 1.00 equiv.), 3-(6-amino-1- methyl-1H-indazol-3-yl)piperidine-2,6-dione (62.69 mg, 242.74 µmol, 1.20 equiv.), and DIPEA (78.43 mg, 606.85 µmol, 105.70 µL, 3.00 equiv.) in DMF (2 mL) was degassed and purged with N2 three times, the mixture was stirred at 25 °C for 5 mins. After 5 mins, to the mixture was added HATU (92.30 mg, 242.74 µmol, 1.20 equiv.), and the mixture was stirred at 25 °C for 16 hours under N2 atmosphere. The reaction mixture was diluted with water (30 mL) and extracted with DCM (30 mL × 3). The combined organic layers were dried over anhydrous sodium sulfate filtered and concentrated under reduced pressure to give a crude tert- butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2- ((3R,5S)-4-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-inda zol-6-yl)amino)-2-oxoethyl)- 3,5-dimethylpiperazin-1-yl)ethoxy)-2-methylphenyl)picolinate (280 mg, crude) as a brown solid. MS (ESI) m/z: 1031.6[M+H] + Step E. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((3R,5S)-4-(2-((3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)-3,5-dimethylpipera zin-1-yl)ethoxy)-2- methylphenyl)picolinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(2-((3R,5S)-4-(2-((3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)amino)-2- oxoethyl)-3,5-dimethylpiperazin-1-yl)ethoxy)-2-methylphenyl) picolinate (280 mg, 271.52 µmol, 1.00 equiv.) in DCM (1.4 mL) and TFA (1.4 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 25 °C for 16 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by reverse-phase HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-((3R,5S)-4-(2-((3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-6- yl)amino)-2-oxoethyl)-3,5-dimethylpiperazin-1-yl)ethoxy)-2-m ethylphenyl)picolinic acid (81.8 mg, 78.0 µmol, 28.7% yield, 92.4% purity) as a pink solid. MS (ESI) m/z: 957.7 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 13.02 - 12.70 (m, 1H), 10.88 (s, 1H), 9.80 (s, 1H), 8.17 - 8.11 (m, 1H), 8.02 (s, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.68 - 7.58 (m, 2H), 7.50 - 7.30 (m, 6H), 7.22 (d, J = 7.2 Hz, 1H), 7.16 - 7.08 (m, 1H), 7.01 - 6.89 (m, 2H), 6.66 (d, J = 6.8 Hz, 1H), 4.98 (s, 2H), 4.37 - 4.29 (m, 1H), 4.23 - 4.06 (m, 2H), 3.92 (s, 4H), 3.21 - 2.77 (m, 9H), 2.74 - 2.58 (m, 4H), 2.44 - 2.27 (m, 2H), 2.26 - 2.07 (m, 2H), 1.91 (s, 3H), 1.02 (s, 6H) Example 142. Preparation of Compound 166 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-1,1-dim ethyl-2-oxo-ethyl]-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of methyl 2-[4-[3-(3-bromo-2-methyl- phenoxy)propyl]-1-piperidyl]-2-methyl-propanoate To a solution of 4-[3-(3-bromo-2-methyl-phenoxy)propyl]piperidine (500 mg, 1.60 mmol, 1 equiv.) and methyl 2-bromo-2-methyl-propanoate (347.86 mg, 1.92 mmol, 248.47 µL, 1.2 equiv.) in CH 3 CN(2 mL) was added K 2 CO 3 (663.95 mg, 4.80 mmol, 3 equiv.) and KI (132.91 mg, 800.65 µmol, 0.5 equiv.). The mixture was stirred at 80 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate= 0/1 to 1/1) to give methyl 2-[4-[3-(3-bromo-2-methyl-phenoxy)propyl]-1-piperidyl]-2-met hyl-propanoate (340 mg, 824.5 µmol, 51.4% yield) as a yellow oil. MS (ESI) m/z: 412.2 [M+H] + Step B. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-(2-methoxy-1,1-d imethyl-2-oxo-ethyl)-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate A mixture of methyl 2-[4-[3-(3-bromo-2-methyl-phenoxy)propyl]-1-piperidyl]-2-met hyl- propanoate (340 mg, 824.52 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3, 2-dioxaborolan-2-yl)pyridine-2- carboxylate (505.06 mg, 824.52 µmol, 1 equiv.), Ad 2 nBuP Pd G 3 (cataCXium® A Pd G 3 ) (60.05 mg, 82.45 µmol, 0.1 equiv.), and KF (1.5 M, 549.68 uL, 1 equiv.) in dioxane (2 mL) was degassed and purged with N2 and stirred at 100 °C for 1 hour under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=1/0 to 1/1) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[1-(2-methoxy-1,1-dimethyl-2-oxo-ethyl)-4-piperi dyl]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (370 mg, 452.3 µmol, 54.8% yield) as a yellow oil. MS (ESI) m/z: 818.7 [M+H] + 1 HNMR (400 MHz, CDCl3) δ = 11.11 - 10.17 (m, 1H), 7.87 - 7.82 (m, 1H), 7.56 (d, J = 7.6 Hz, 1H), 7.48 (d, J = 7.2 Hz, 1H), 7.38 (d, J = 8.4 Hz, 1H), 7.35 - 7.31 (m, 2H), 7.31 - 7.27 (m, 1H), 7.25 - 7.22 (m, 1H), 7.09 (t, J = 8.0 Hz, 1H), 6.90 (d, J = 8.8 Hz, 1H), 6.80 (d, J = 8.4 Hz, 1H), 6.68 (d, J = 7.2 Hz, 1H), 5.12 - 4.95 (m, 2H), 4.11 - 4.08 (m, 2H), 3.96 (t, J = 6.4 Hz, 2H), 3.71 (s, 3H), 3.06 (t, J = 6.0 Hz, 2H), 2.91 (d, J = 10.0 Hz, 2H), 2.16 - 2.08 (m, 2H), 1.98 (s, 3H), 1.85 - 1.77 (m, 2H), 1.73 (d, J = 8.0 Hz, 2H), 1.45 - 1.38 (m, 2H), 1.35 - 1.28 (m, 9H), 1.15 (s, 9H). Step C. Procedure for preparation of 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyr idyl]-2-methyl- phenoxy]propyl]-1-piperidyl]-2-methyl-propanoic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[3-[1-(2-methoxy-1,1-dimethyl-2-oxo-ethyl)-4-pipe ridyl]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (370 mg, 452.30 µmol, 1 equiv.) in H2O (1 mL), MeOH (1 mL) and THF (1 mL) was added LiOH•H2O (189.80 mg, 4.52 mmol, 10 equiv.). The mixture was stirred at 50 °C for 2 hours. The pH was adjusted to 6 with 1M HCl. The mixture was extracted with DCM (2mL × 3). The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH = 10:1) to give compound 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]propyl]-1-piperidyl]-2- methyl-propanoic acid (166 mg, 206.4 µmol, 45.6% yield) as a yellow oil. MS (ESI) m/z: 804.7 [M+H] + Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-diox o-3-piperidyl)-1-methyl- indazol-6-yl]amino]-1,1-dimethyl-2-oxo-ethyl]-4-piperidyl]pr opoxy]-2-methyl- phenyl]pyridine-2-carboxylate To a solution of 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]propyl]-1-piperidyl]-2- methyl-propanoic acid (100 mg, 124.38 µmol, 1 equiv.), 3-(6-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (48.19 mg, 186.57 µmol, 1.5 equiv.) in DMF (1 mL) was added HATU (118.23 mg, 310.94 µmol, 2.5 equiv.) and DIEA (48.22 mg, 373.13 µmol, 64.99 µL, 3 equiv.). The mixture was stirred at 40 °C for 4 hours. The reaction mixture was quenched by water (2 mL) at 25 °C filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, ethyl acetate) to give tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-[2- [[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]amino]-1,1-dimethyl-2-oxo-ethyl]-4-piper idyl]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (50 mg, 47.8 µmol, 38.5% yield) as a colorless oil. MS (ESI) m/z: 1044.9 [M+H] + Step E. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6- yl]amino]-1,1-dimethyl-2-oxo-ethyl]-4-piperidyl]propoxy]-2-m ethyl-phenyl]pyridine-2- carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-ind azol-6-yl]amino]-1,1-dimethyl-2- oxo-ethyl]-4-piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-c arboxylate (50 mg, 47.88 µmol, 1 equiv.) in DCM (1 mL) was added TFA (1 mL). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol-6- yl]amino]-1,1-dimethyl-2-oxo-ethyl]-4-piperidyl]propoxy]-2-m ethyl-phenyl]pyridine-2- carboxylic acid (27.5 mg, 27.8 µmol, 58.1% yield, 99.9% purity) as a white solid. MS (ESI) m/z: 988.8[M+H] + 1 HNMR (400 MHz, DMSO-d6,) δ = 13.49 - 11.83 (m, 1H), 10.88 (s, 1H), 9.74 (s, 1H), 8.16 (s, 1H), 8.08 - 7.99 (m, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 8.4 Hz, 2H), 7.49 - 7.41 (m, 3H), 7.39 - 7.32 (m, 2H), 7.23 (d, J = 8.8 Hz, 1H), 7.11 - 7.06 (m, 1H), 6.95 (d, J = 8.8 Hz, 1H), 6.87 (d, J = 8.0 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.32 (dd, J = 5.2, 9.6 Hz, 1H), 3.95 (t, J = 6.4 Hz, 2H), 3.92 (s, 5H), 3.18 - 3.17 (m, 1H), 3.17 (s, 2H), 3.02 (t, J = 5.6 Hz, 2H), 2.74 (d, J = 10.0 Hz, 2H), 2.67 - 2.59 (m, 2H), 2.15 (d, J = 5.6 Hz, 2H), 1.90 (s, 3H), 1.81 - 1.70 (m, 4H), 1.45 - 1.39 (m, 2H), 1.32 (s, 3H), 1.19 (s, 6H). Example 143. Preparation of Compound 167a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[(3R)-3-[1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-o xo-ethyl]-4-piperidyl]butoxy]- 2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 4-[(E)-3-ethoxy-1-methyl-3-oxo-prop-1- enyl]piperidine-1-carboxylate To a solution of NaH (2.51 g, 62.69 mmol, 60% purity, 1.5 equiv.) in THF (100 mL) was added ethyl 2-diethoxyphosphorylacetate (11.24 g, 50.15 mmol, 9.95 mL, 1.2 equiv.) at 0 °C slowly. The reaction mixture was stirred at 0 °C for 1 hour, after which tert-butyl 4-acetylpiperidine- 1-carboxylate (9.5 g, 41.80 mmol, 1 equiv.) was added at 0 °C slowly. The mixture was warmed to 25 °C and then stirred at 25 °C for 12 hours. The reaction mixture was cooled to 0 °C, and saturated NH4Cl solution was added dropwise. Then the mixture was diluted with H2O (100 mL) and extracted with EtOAc (200 mL × 3). The combined organic layers were washed with brine (300 mL × 1), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=20/1 to 10/1) to give tert-butyl 4-[(E)-3-ethoxy-1-methyl-3-oxo-prop-1- enyl]piperidine-1-carboxylate (5.7 g, 15.4 mmol, 36.9% yield, 80.6% purity) as a yellow oil. MS (ESI) m/z: 198.0 [M-100+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 5.68 - 5.60 (m, 1H), 4.09 - 4.03 (m, 2H), 4.03 - 3.90 (m, 2H), 2.80 - 2.59 (m, 2H), 2.25 - 2.15 (m, 1H), 2.08 (s, 3H), 1.69 - 1.59 (m, 2H), 1.39 (s, 9H), 1.32 - 1.21 (m, 2H), 1.19 (t, J = 7.2 Hz, 3H) Step B. Procedure for preparation of tert-butyl 4-(3-ethoxy-1-methyl-3-oxo- propyl)piperidine-1-carboxylate To a solution of tert-butyl 4-[(E)-3-ethoxy-1-methyl-3-oxo-prop-1-enyl]piperidine-1- carboxylate (5.7 g, 19.17 mmol, 1 equiv.) in EtOH (60 mL) was added PtO2 (870.47 mg, 3.83 mmol, 0.2 equiv.) under N 2 . The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H2 (15 psi) at 25 °C for 12 hours. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give tert-butyl 4-(3-ethoxy-1-methyl-3-oxo-propyl)piperidine-1-carboxylate (5.7 g, crude) as a colorless oil. MS (ESI) m/z: 200.2 [M-100+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 4.09 - 4.02 (m, 2H), 3.97 (d, J = 11.6 Hz, 2H), 2.60 (s, 2H), 2.36 (dd, J = 5.2, 14.8 Hz, 1H), 2.10-2.00 (m, 1H), 1.82 - 1.71 (m, 1H), 1.55 (d, J = 12.0 Hz, 2H), 1.38 (s, 9H), 1.35 - 1.26 (m, 1H), 1.17 (t, J = 7.2 Hz, 3H), 1.10 - 0.96 (m, 2H), 0.84 (d, J = 6.8 Hz, 3H) Step C. Procedure for preparation of tert-butyl 4-(3-hydroxy-1-methyl- propyl)piperidine-1-carboxylate To a solution of tert-butyl 4-(3-ethoxy-1-methyl-3-oxo-propyl)piperidine-1-carboxylate (5.7 g, 19.04 mmol, 1 equiv.) in THF (60 mL) was added LiAlH 4 (722.57 mg, 19.04 mmol, 1 equiv.) at 0 °C. The mixture was then degassed and purged with N 2 and stirred at 0 °C for 2 hours. The reaction mixture was quenched by addition Na2SO4•10H2O (2 g) at 0 °C and filtered to give a filtrate. The filtrate was extracted with ethyl acetate (100 mL×3).The combined organic phase was washed with brine (100 mL×2), dried with anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~30% ethyl acetate/petroleum ether) to give tert-butyl 4-(3- hydroxy-1-methyl-propyl)piperidine-1-carboxylate (4.2 g, 15.6 mmol, 81.7% yield, 95.6% purity) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 4.30 (t, J = 5.2 Hz, 1H), 4.02 - 3.92 (m, 2H), 3.49 - 3.41 (m, 1H), 3.41 - 3.34 (m, 1H), 2.73 - 2.55 (m, 2H), 1.56 - 1.52 (m, 1H), 1.52 - 1.47 (m, 2H), 1.38 (s, 9H), 1.34 - 1.28 (m, 1H), 1.28 - 1.10 (m, 2H), 1.10 - 0.95 (m, 2H), 0.78 (d, J = 6.8 Hz, 3H) Step D. Procedure for preparation of tert-butyl 4-[3-(3-bromo-2-methyl-phenoxy)-1- methyl-propyl]piperidine-1-carboxylate A mixture of tert-butyl 4-(3-hydroxy-1-methyl-propyl)piperidine-1-carboxylate (2 g, 7.77 mmol, 1 equiv.), 3-bromo-2-methyl-phenol (1.45 g, 7.77 mmol, 1 equiv.), 3-bromo-2-methyl- phenol (1.45 g, 7.77 mmol, 1 equiv.), and 2-(tributyl-λ5-phosphanylidene)acetonitrile (2.25 g, 9.33 mmol, 1.2 equiv.) in toluene (20 mL) was degassed and purged with N2 and stirred at 120 °C for 4 hours under N2 atmosphere. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by reverse-phase HPLC (with 0.1% HCOOH) to give tert-butyl 4-[3-(3-bromo-2-methyl-phenoxy)-1-methyl-propyl]piperidine-1 -carboxylate (2.5 g, 5.8 mmol, 75.0% yield, 99.5% purity) as a brown oil. MS (ESI) m/z: 328.3 [M-100+3] + ( 80 Br). 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.17 - 7.12 (m, 1H), 7.12 - 7.06 (m, 1H), 6.98 (d, J = 7.6 Hz, 1H), 4.08 - 3.93 (m, 4H), 2.71 - 2.54 (m, 2H), 2.22 (s, 3H), 1.88 - 1.80 (m, 1H), 1.60 - 1.48 (m, 4H), 1.39 (s, 10H), 1.16 - 1.00 (m, 2H), 0.87 (d, J = 6.4 Hz, 3H) Step E. Procedure for preparation of tert-butyl 4-[(1R)-3-(3-bromo-2-methyl-phenoxy)- 1-methyl-propyl]piperidine-1-carboxylate The compound tert-butyl 4-[3-(3-bromo-2-methyl-phenoxy)-1-methyl-propyl]piperidine-1 - carboxylate (2.5 g, 5.86 mmol, 1 equiv.) was purified by prep-HPLC to give tert-butyl 4-[(1R)- 3-(3-bromo-2-methyl-phenoxy)-1-methyl-propyl]piperidine-1-ca rboxylate (0.9 g, 2.0 mmol, 35.2% yield, 97.7% purity) as a brown oil and tert-butyl 4-[(1S)-3-(3-bromo-2-methyl- phenoxy)-1-methyl-propyl]piperidine-1-carboxylate (0.7 g, 1.4 mmol, 25.0% yield, 89.3% purity) as a brown oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.17 - 7.12 (m, 1H), 7.12 - 7.06 (m, 1H), 6.98 (d, J = 7.6 Hz, 1H), 4.08 - 3.90 (m, 4H), 2.77 - 2.54 (m, 2H), 2.22 (s, 3H), 1.90 - 1.79 (m, 1H), 1.61 - 1.47 (m, 4H), 1.38 (s, 10H), 1.17 - 1.05 (m, 2H), 0.87 (d, J = 6.8 Hz, 3H) Step F. Procedure for preparation of 4-[(1R)-3-(3-bromo-2-methyl-phenoxy)-1-methyl- propyl]piperidine A mixture of tert-butyl 4-[(1R)-3-(3-bromo-2-methyl-phenoxy)-1-methyl-propyl]piperid ine-1- carboxylate (0.9 g, 2.11 mmol, 1 equiv.) and HCl/ dioxane (4 M, 527.69 µL, 1 equiv.) was stirred at 25 °C for 2 hours. The mixture was filtered to give a filter cake. The crude product was triturated with EtOAc 5 mL at 25 °C for 10 min to give 4-[(1R)-3-(3-bromo-2-methyl- phenoxy)-1-methyl-propyl]piperidine (620 mg, 1.6 mmol, 78.9% yield, 97.4% purity) as a white solid. MS (ESI) m/z: 326.0 [M+H] + SFC Method details: Column: Chiralpak IH-350 × 4.6mm I.D., 3um; Mobile phase: Phase A for Hexane (0.05%IPAm), and Phase B for IPA (0.05%IPAm); Gradient elution: 10% B in A; Flow rate: 1mL/min; Detector: PDA; Column Temp: 35 o C; Step G. Procedure for preparation of ethyl 2-[4-[(1R)-3-(3-bromo-2-methyl-phenoxy)-1- methyl-propyl]-1-piperidyl]acetate To a solution of 4-[(1R)-3-(3-bromo-2-methyl-phenoxy)-1-methyl-propyl]piperid ine (620 mg, 1.71 mmol, 1 equiv., HCl) and ethyl 2-bromoacetate (256.90 mg, 1.54 mmol, 170.13 uL, 0.9 equiv.) in MeCN (7 mL) was added K2CO3 (708.71 mg, 5.13 mmol, 3 equiv.). The mixture was stirred at 60 °C for 2 hours. The mixture was filtered to give a filtrate, which was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~10% methanol/ dichloromethane) to give ethyl 2-[4-[(1R)-3-(3- bromo-2-methyl-phenoxy)-1-methyl-propyl]-1-piperidyl]acetate (530 mg, 1.2 mmol, 74.5% yield, 99.1% purity) as a yellow oil. MS (ESI) m/z: 414.0 [M+3] + ( 80 Br). 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.17 - 7.13 (m, 1H), 7.12 - 7.06 (m, 1H), 6.98 (d, J = 7.6 Hz, 1H), 4.09 - 4.04 (m, 2H), 4.04 - 3.91 (m, 2H), 3.14 (s, 2H), 2.84 (d, J = 11.2 Hz, 2H), 2.22 (s, 3H), 2.13 - 2.04 (m, 2H), 1.90 - 1.80 (m, 1H), 1.60 - 1.46 (m, 4H), 1.33 - 1.20 (m, 2H), 1.20 - 1.10 (m, 4H), 0.88 (d, J = 6.4 Hz, 3H) Step H. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[(3R)-3-[1-(2-ethoxy-2 -oxo-ethyl)-4- piperidyl]butoxy]-2-methyl-phenyl]pyridine-2-carboxylate Ethyl 2-[4-[(1R)-3-(3-bromo-2-methyl-phenoxy)-1-methyl-propyl]-1-p iperidyl]acetate (400 mg, 970.03 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (594.19 mg, 970.03 µmol, 1 equiv.), KF (169.07 mg, 2.91 mmol, 68.17 µL, 3 equiv.), and Ad2nBuP Pd G3 (70.64 mg, 97.00 µmol, 0.1 equiv.) were taken up into a microwave tube in dioxane (4 mL) and H2O (0.4 mL). The sealed tube was heated at 100 °C for 1 hour under microwave. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~50% ethyl acetate/petroleum ether) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[(3R)-3-[1-(2-ethoxy-2-oxo-ethyl)- 4-piperidyl]butoxy]-2-methyl- phenyl]pyridine-2-carboxylate (470 mg, 529.1 µmol, 54.5% yield, 92.1% purity) as a yellow solid. MS (ESI) m/z: 818.8 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.86 (s, 1H), 8.01 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.60 (d, J = 7.6 Hz, 1H), 7.50 - 7.39 (m, 3H), 7.39 - 7.31 (m, 2H), 7.12 - 7.06 (m, 1H), 6.96 - 6.88 (m, 2H), 6.57 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.09 - 4.03 (m, 2H), 4.02 - 3.92 (m, 2H), 3.87 (t, J = 5.6 Hz, 2H), 3.14 (s, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.83 (d, J = 10.0 Hz, 2H), 2.13 - 2.03 (m, 2H), 1.86 (s, 3H), 1.84 - 1.76 (m, 1H), 1.61 - 1.44 (m, 4H), 1.33 - 1.21 (m, 2H), 1.18 - 1.14 (m, 4H), 1.03 - 0.97 (m, 9H), 0.86 (d, J = 5.6 Hz, 3H) Step I. Procedure for preparation of 2-[4-[(1R)-3-[3-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-2- methyl-phenoxy]-1-methyl-propyl]-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[(3R)-3-[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]but oxy]-2-methyl-phenyl]pyridine- 2-carboxylate (470 mg, 574.55 µmol, 1 equiv.) in THF (4 mL) was added LiOH (1 M, 1.72 mL, 3 equiv.).The mixture was stirred at 25 °C for 12 hours. The mixture was concentrated under reduced pressure to removed THF, added HCl (1 M) to pH = 6, and filtered to give 2-[4- [(1R)-3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H-isoquinolin-2-yl]-2-tert- butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]-1-methyl-propyl] -1-piperidyl]acetic acid (370 mg, crude) as a yellow solid. MS (ESI) m/z: 713.2 [M+H] + Step J. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[(3R)-3-[1-[2-[[3-(2,6 -dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-4-piperidyl]butoxy]-2-methy l-phenyl]pyridine-2- carboxylate To a solution of 2-[4-[(1R)-3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]-1-methyl-propyl]-1- piperidyl]acetic acid (100 mg, 126.59 µmol, 1 equiv.) and HATU (72.20 mg, 189.88 µmol, 1.5 equiv.) in DMF (1 mL) was added DIEA (49.08 mg, 379.76 µmol, 66.15 µL, 3 equiv.) and 3- (6-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (39.23 mg, 151.90 µmol, 1.2 equiv.). The mixture was stirred at 25 °C for 1 hour. The mixture was added into H 2 O (5 mL), filtered to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- [3-[(3R)-3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazo l-6-yl]amino]-2-oxo-ethyl]-4- piperidyl]butoxy]-2-methyl-phenyl]pyridine-2-carboxylate (110 mg, crude) as a pink solid. MS (ESI) m/z: 1030.7 [M+H] + Step K. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[(3R)-3-[1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]butoxy]-2-methyl-phenyl]p yridine-2-carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[(3R)-3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methy l-indazol-6-yl]amino]-2-oxo- ethyl]-4-piperidyl]butoxy]-2-methyl-phenyl]pyridine-2-carbox ylate (110 mg, 106.77 µmol, 1 equiv.) in DCM (0.5 mL) was added TFA (770.00 mg, 6.75 mmol, 0.5 mL, 63.25 equiv.). The mixture was stirred at 25 °C for 12 hours. The mixture was concentrated under reduced pressure to remove DCM. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[(3R)-3-[ 1-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]-4-piper idyl]butoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid (34.3 mg, 33.6 µmol, 31.5% yield, 95.4% purity) as a yellow solid. MS (ESI) m/z: 974.5 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.16 - 11.97 (m, 2H), 10.88 (s, 1H), 9.87 (s, 1H), 8.14 (s, 1H), 8.07 - 8.00 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 8.8 Hz, 2H), 7.49 - 7.43 (m, 3H), 7.39 - 7.32 (m, 2H), 7.21 (dd, J = 1.2, 8.8 Hz, 1H), 7.10 (t, J = 8.0 Hz, 1H), 6.96 (d, J = 8.8 Hz, 1H), 6.90 (d, J = 8.4 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.32 (dd, J = 5.2, 9.6 Hz, 1H), 4.04 - 3.97 (m, 2H), 3.94 - 3.89 (m, 5H), 3.17 (s, 2H), 3.02 (t, J = 5.6 Hz, 2H), 2.99 - 2.93 (m, 2H), 2.66 - 2.60 (m, 2H), 2.42 - 2.24 (m, 2H), 2.19 - 2.14 (m, 2H), 1.89 (s, 3H), 1.65 - 1.54 (m, 4H), 1.46 - 1.35 (m, 2H), 1.30 - 1.18 (m, 2H), 0.93 (d, J = 6.4 Hz, 3H) Example 144. Preparation of Compound 167b 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[(3S)-3-[1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-o xo-ethyl]-4-piperidyl]butoxy]- 2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 4-[(1S)-3-(3-bromo-2-methyl-phenoxy)-1-methyl- propyl]piperidine A mixture of tert-butyl 4-[(1S)-3-(3-bromo-2-methyl-phenoxy)-1-methyl-propyl]piperid ine-1- carboxylate (700 mg, 1.64 mmol, 1 equiv.) and HCl/EtOAc (4 M, 7.00 mL, 17.06 equiv.) was stirred at 25 °C for 2 hours. The mixture was concentrated under reduced pressure to give a residue. The residue was triturated with EtOAc (3 mL) at 25 °C for 10 min to give 4-[(1S)-3- (3-bromo-2-methyl-phenoxy)-1-methyl-propyl]piperidine (550 mg, 1.5 mmol, 91.4% yield, 98.9% purity, HCl) as a yellow solid. MS (ESI) m/z: 327.8 [M+3] + ( 80 Br). Step B. Procedure for preparation of ethyl 2-[4-[(1S)-3-(3-bromo-2-methyl-phenoxy)-1- methyl-propyl]-1-piperidyl]acetate To a solution of 4-[(1S)-3-(3-bromo-2-methyl-phenoxy)-1-methyl-propyl]piperid ine (550 mg, 1.52 mmol, 1 equiv., HCl) and ethyl 2-bromoacetate (227.90 mg, 1.36 mmol, 150.93 µL, 0.9 equiv.) in CH3CN (6 mL) was added K2CO3 (628.67 mg, 4.55 mmol, 3 equiv.) The mixture was stirred at 60 °C for 1 hour. The mixture was filtered to give a filtrate, which was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~10% methanol / dichloromethane) to give ethyl 2-[4-[(1S)- 3-(3-bromo-2-methyl-phenoxy)-1-methyl-propyl]-1-piperidyl]ac etate (480 mg, 1.2 mmol, 76.3% yield, 99.5% purity) as a yellow oil. MS (ESI) m/z: 414.0 [M+3] + ( 80 Br). 1 H NMR (400 MHz, DMSO-d6) δ = 7.17 - 7.12 (m, 1H), 7.12 - 7.06 (m, 1H), 6.98 (d, J = 8.0 Hz, 1H), 4.10 - 4.04 (m, 2H), 4.04 - 3.92 (m, 2H), 3.14 (s, 2H), 2.84 (d, J = 11.2 Hz, 2H), 2.22 (s, 3H), 2.14 - 2.03 (m, 2H), 1.90 - 1.80 (m, 1H), 1.60 - 1.46 (m, 4H), 1.34 - 1.20 (m, 1H), 1.18 (t, J = 7.2 Hz, 4H), 0.88 (d, J = 6.4 Hz, 3H) Step C. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[(3S)-3-[1-(2-ethoxy-2 -oxo-ethyl)-4- piperidyl]butoxy]-2-methyl-phenyl]pyridine-2-carboxylate Ethyl 2-[4-[(1S)-3-(3-bromo-2-methyl-phenoxy)-1-methyl-propyl]-1-p iperidyl]acetate (400 mg, 970.03 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (594.19 mg, 970.03 µmol, 1 equiv.), Ad 2 nBuP Pd G 3 (70.64 mg, 97.00 µmol, 0.1 equiv.), and KF (169.07 mg, 2.91 mmol, 68.17 µL, 3 equiv.) were taken up into a microwave tube in dioxane (4 mL) and H 2 O (0.4 mL). The sealed tube was heated at 100 °C for 1 hour under microwave. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~50% ethyl acetate/petroleum ether) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3- [(3S)-3-[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]butoxy]-2-meth yl-phenyl]pyridine-2- carboxylate (530 mg, 595.7 µmol, 61.4% yield, 91.9% purity) as a yellow solid. MS (ESI) m/z: 818.7 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 12.85 (s, 1H), 8.01 (d, J = 8.0 Hz, 1H), 7.77 (d, J = 8.0 Hz, 1H), 7.60 (d, J = 7.6 Hz, 1H), 7.50 - 7.39 (m, 3H), 7.38 - 7.31 (m, 2H), 7.13 - 7.06 (m, 1H), 6.96 - 6.89 (m, 2H), 6.57 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.08 - 4.03 (m, 2H), 4.02 - 3.92 (m, 2H), 3.87 (t, J = 5.6 Hz, 2H), 3.13 (s, 2H), 3.02 (t, J = 5.6 Hz, 2H), 2.83 (d, J = 10.0 Hz, 2H), 2.06 (t, J = 10.4 Hz, 2H), 1.86 (s, 3H), 1.84 - 1.75 (m, 1H), 1.60 - 1.44 (m, 4H), 1.34 - 1.20 (m, 2H), 1.18 - 1.14 (m, 4H), 1.03 - 0.98 (m, 9H), 0.88 - 0.82 (m, 3H) Step D. Procedure for preparation of 2-[4-[(1S)-3-[3-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-2- methyl-phenoxy]-1-methyl-propyl]-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[(3S)-3-[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]but oxy]-2-methyl-phenyl]pyridine- 2-carboxylate (530 mg, 647.90 µmol, 1 equiv.) in THF (4 mL) was added LiOH (1 M, 1.94 mL, 3 equiv.). The mixture was stirred at 25 °C for 12 hours. The mixture was concentrated under reduced pressure to remove THF, treated with HCl (1 M) to pH = 6, and filtered to give 2-[4- [(1S)-3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H-isoquinolin-2-yl]-2-tert- butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]-1-methyl-propyl] -1-piperidyl]acetic acid (500 mg, crude) as a yellow solid. MS (ESI) m/z: 790.3 [M+H] + Step E. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[(3S)-3-[1-[2-[[3-(2,6 -dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-4-piperidyl]butoxy]-2-methy l-phenyl]pyridine-2- carboxylate To a solution of 2-[4-[(1S)-3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]-1-methyl-propyl]-1- piperidyl]acetic acid (150 mg, 189.88 µmol, 1 equiv.) and HATU (86.64 mg, 227.85 µmol, 1.2 equiv.) in DMF (1.5 mL) was added DIEA (73.62 mg, 569.64 µmol, 99.22 µL, 3 equiv.) and 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (68.66 mg, 265.83 µmol, 1.4 equiv.). The mixture was stirred at 25 °C for 1 hour. The mixture was added into H2O (5 mL) and filtered to give a filter cake, which was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH = 10:1) to give tert-butyl 6-[8- (1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin- 2-yl]-3-[3-[(3S)-3-[1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-4-piperidyl]butoxy]-2- methyl-phenyl]pyridine-2-carboxylate (100 mg, 95.0 µmol, 50.1% yield, 97.9% purity) as a yellow solid. MS (ESI) m/z: 1030.6 [M+H] + Step F. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[(3S)-3-[1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]butoxy]-2-methyl-phenyl]p yridine-2-carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[(3S)-3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methy l-indazol-6-yl]amino]-2-oxo- ethyl]-4-piperidyl]butoxy]-2-methyl-phenyl]pyridine-2-carbox ylate (100 mg, 97.06 µmol, 1 equiv.) in DCM (0.5 mL) was added TFA (770.00 mg, 6.75 mmol, 0.5 mL, 69.57 equiv.). The mixture was stirred at 25 °C for 12 hours. The mixture was concentrated under reduced pressure to remove DCM. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[(3S)-3-[ 1-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]-4-piper idyl]butoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid (33.4 mg, 31.4 µmol, 32.4% yield, 96.1% purity) as an off- white solid. MS (ESI) m/z: 974.5 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 13.01 - 12.69 (m, 1H), 10.89 (s, 1H), 8.07 - 7.97 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.69 - 7.58 (m, 2H), 7.50 - 7.41 (m, 3H), 7.40 - 7.32 (m, 2H), 7.17 (d, J = 8.8 Hz, 1H), 7.13 - 7.07 (m, 1H), 6.97 (d, J = 8.8 Hz, 1H), 6.91 (d, J = 8.4 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.36 - 4.30 (m, 1H), 4.06 - 3.97 (m, 2H), 3.93 (s, 5H), 3.11 - 2.92 (m, 4H), 2.69 - 2.60 (m, 2H), 2.40 - 2.27 (m, 2H), 2.25 -2.04 (m, 2H), 1.90 (s, 5H), 1.78 - 1.21 (m, 8H), 0.93 (d, J = 6.4 Hz, 3H) Example 145. Preparation of Compound 171a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pip erazin-1-yl)-2,2-difluoro-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-2,2- difluoro-propanoic acid A mixture of ethyl 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-2,2-difluoro-prop anoate (800 mg, 1.97 mmol, 1 equiv.), LiOH•H2O (248.51 mg, 5.92 mmol, 3 equiv.), in THF (6 mL) and H 2 O (2 mL) was stirred at 40 °C for 16 hours. The reaction mixture was diluted with H 2 O (2 mL) and concentrated as a turbid liquid. Then the pH of the turbid liquid was adjusted to 3 with 1 N HCl (0.5 mL), and the residue was extracted with DCM (10 mL), The combined organic layers were filtered and concentrated. The residue was used in the next step without other purification. The compound 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-2,2-difluoro- propanoic acid (800 mg, crude) was obtained as a yellow oil. Step B. Procedure for preparation of 3-(7-(4-(3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2,2-difluoropropanoyl)piperazin-1- yl)-1-methyl-1H-indazol- 3-yl)piperidine-2,6-dione A mixture of 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-2,2-difluoro-prop anoic acid (100 mg, 265.10 µmol, 1 equiv.), HATU (251.99 mg, 662.74 µmol, 2.5 equiv.), and DIEA (102.79 mg, 795.29 µmol, 138.53 uL, 3 equiv.) in DMF (1 mL) was stirred at 40 °C for 0.25 h.3-(1- Methyl-7-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dione (104.15 mg, 318.12 µmol, 1.2 equiv.) was then added to the mixture, and the mixture was stirred at 40 °C for 15.75 hours. The reaction mixture was diluted with H2O (4 mL × 3) and extracted with ethyl acetate (8 mL × 3). The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The mixture was purified by flash silica gel chromatography (Eluent of 0 ~ 70% ethyl acetate / Petroleum) to give 3-(7-(4-(3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2,2-difluoropropanoyl)piperazin-1- yl)-1-methyl-1H-indazol-3- yl)piperidine-2,6-dione (125 mg, 182.1 µmol, 68.7% yield) as a yellow oil. MS (ESI) m/z: 686.2 [M+H] + . Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)-2,2-difluoro-3-oxopro pyl)cyclohexyl)oxy)-2- methylphenyl)picolinate A mixture of 3-(7-(4-(3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2 ,2- difluoropropanoyl)piperazin-1-yl)-1-methyl-1H-indazol-3-yl)p iperidine-2,6-dione (125 mg, 182.06 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (133.82 mg, 218.47 µmol, 1.2 equiv.), [2-(2-aminophenyl)phenyl]palladium(1 + );bis(1- adamantyl)-butyl-phosphane;methanesulfonate (26.52 mg, 36.41 µmol, 0.2 equiv.), and KF (1.5 M, 182.06 µL, 1.5 equiv.) in dioxane (1.5 mL) and H 2 O (0.5 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 100 °C for 1 h under microwave. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The mixture was purified by flash silica gel chromatography (Eluent of 0 ~ 70% ethyl acetate / petroleum ether) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)piperazin-1-yl)-2,2-difluoro-3-oxopropyl)cyc lohexyl)oxy)-2- methylphenyl)picolinate (170 mg, 155.6 µmol, 85.5% yield) as a yellow oil. MS (ESI) m/z: 1092.4 [M+H] + . Step D. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)-2,2-difluoro-3-oxopro pyl)cyclohexyl)oxy)-2- methylphenyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1r,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-7-yl)piperazin-1- yl)-2,2-difluoro-3-oxopropyl)cyclohexyl)oxy)-2-methylphenyl) picolinate (170 mg, 155.64 µmol, 1 equiv.) in DCM (0.8 mL) and TFA (0.8 mL) was stirred at 40 °C for 4 h. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)piperazin-1-yl)-2,2-difluoro-3-oxopropyl)cyc lohexyl)oxy)-2- methylphenyl)picolinic acid (34.9 mg, 32.4 µmol, 20.8% yield, 96.1% purity) as a white solid. MS (ESI) m/z: 1036.3 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.25 - 12.24 (m, 2H), 10.88 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.49 - 7.43 (m, 4H), 7.39 - 7.33 (m, 2H), 7.09 - 7.01 (m, 3H), 6.97 - 6.91 (m, 2H), 6.62 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.35 (dd, J = 4.8, 9.6 Hz, 1H), 4.28 (s, 3H), 4.25 - 4.13 (m, 2H), 3.93 - 3.90 (m, 2H), 3.64 - 3.49 (m, 1H), 3.29 - 3.09 (m, 4H), 3.04 - 3.01 (m, 2H), 2.90 - 2.71 (m, 2H), 2.68 - 2.62 (m, 2H), 2.36 - 2.31 (m, 1H), 2.21 - 2.06 (m, 5H), 1.92 - 1.85 (m, 5H), 1.74 - 1.65 (m, 1H), 1.45 - 1.34 (m, 2H), 1.31 - 1.23 (m, 2H) 19 F NMR (400 MHz, DMSO-d 6 ) δ = -95.763 ppm. Example 146. Preparation of Compound 174a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(((1R,3r,5S)- 8-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl) amino)-2-oxoethyl)-8- azabicyclo[3.2.1]octan-3-yl)oxy)ethoxy)-2-methylphenyl)picol inic acid Step A. Procedure for preparation of tert-butyl (1R, 5S)-3-(2-ethoxy-2-oxo-ethoxy)-8- azabicyclo [3.2.1] octane-8-carboxylate A mixture of (1R, 3r, 5S)-tert-butyl 3-hydroxy-8-azabicyclo[3.2.1]octane-8-carboxylate (2.0 g, 8.80 mmol, 1.0 equiv.), rhodium (II) acetate dimer (194.45 mg, 439.95 µmol, 0.05 equiv.) in DCM (30 mL), ethyl 2-diazoacetate (1.71 g, 14.96 mmol, 1.57 mL, 1.7 equiv.) was stirred at 20 °C for 2 hours under N 2 , and then treated with additional ethyl 2-diazoacetate (1.51 g, 13.20 mmol, 1.38 mL, 1.5 equiv.). The mixture was stirred at 20 °C for another 2 hours under N 2 , and to the mixture was added ethyl 2-diazoacetate (501.99 mg, 4.40 mmol, 460.54 µL, 0.5 equiv.). The mixture was stirred at 20 °C for 12 hours under N2. The reaction mixture was filtered under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~15% ethyl acetate/petroleum ether) to give tert-butyl (1R, 5S)-3- (2-ethoxy-2-oxo-ethoxy)-8-azabicyclo [3.2.1] octane-8-carboxylate (2.46 g, 7.85 mmol, 89.18% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 4.12 - 4.06 (m, 4H), 3.99 (s, 2H), 3.63 (s, 1H), 2.03 (d, J = 6.4 Hz, 2H), 1.88 - 1.78 (m, 5H), 1.39 (s, 8H), 1.24 - 1.14 (m, 5H) Step B. Procedure for preparation of tert-butyl (1R, 5S)-3-(2-hydroxyethoxy)-8- azabicyclo [3.2.1]octane-8-carboxylate To a solution of tert-butyl (1R, 5S)-3-(2-ethoxy-2-oxo-ethoxy)-8-azabicyclo [3.2.1] octane-8- carboxylate (800 mg, 2.55 mmol, 1.0 equiv.) in THF (8 mL) was purged with N 2 and added in portions LiAlH 4 (96.89 mg, 2.55 mmol, 1.0 equiv.) at 0 °C. The mixture was stirred under N 2 at 0 °C for 2 hours. The reaction mixture was quenched with Na2SO4•10H2O and slowly warmed to 20 °C. The mixture was filtered and the filter cake was washed with DCM (8 mL). Then the organic layer were concentrated under reduced pressure to give tert-butyl (1R, 5S)-3- (2-hydroxyethoxy)-8-azabicyclo [3.2.1]octane-8-carboxylate (553 mg, crude) as a yellow oil. Step C. Procedure for preparation of tert-butyl (1R, 5S)-3-[2-(3-bromo-2-methyl- phenoxy) ethoxy]-8-azabicyclo[3.2.1]octane-8-carboxylate To a solution of tert-butyl (1R, 5S)-3-(2-hydroxyethoxy)-8-azabicyclo [3.2.1]octane-8- carboxylate (1.40 g, 5.16 mmol, 1.0 equiv.) and 3-bromo-2-methyl-phenol (1.93 g, 10.32 mmol, 2.0 equiv.) in toluene (20 mL) was added 2-(tributyl-λ 5 -phosphanylidene) acetonitrile (1.87 g, 7.74 mmol, 1.5 equiv.). The mixture was stirred at 120 °C for 12 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by flash silica gel chromatography (Eluent of 0~30 % ethyl acetate/petroleum ether) to give compound tert-butyl (1R, 5S)-3-[2-(3-bromo-2-methyl- phenoxy) ethoxy]-8-azabicyclo[3.2.1]octane-8-carboxylate (1.80 g, 3.9 mmol, 76.2% yield, 96.2% purity) as a light yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.18 - 7.15 (m, 1H), 7.09 (t, J = 8.0 Hz, 1H), 7.00 (d, J = 8.0 Hz, 1H), 4.14 - 4.09 (m, 2H), 3.99 (s, 2H), 3.71 – 3.67 (m, 3H), 2.24 (s, 3H), 2.04 - 1.98 (m, 2H), 1.86 - 1.73 (m, 6H), 1.39 (s, 9H) Step D. Procedure for preparation of ethyl 2-[(1R, 5S)-3-[2-(3-bromo-2-methyl- phenoxy)ethoxy]-8-azabicyclo[3.2.1]octan-8-yl]acetate To a solution of tert-butyl (1R, 5S)-3-[2-(3-bromo-2-methyl-phenoxy) ethoxy]-8- azabicyclo[3.2.1]octane-8-carboxylate (1.80 g, 4.09 mmol, 1.0 equiv.) was added HCl/EtOAc (15 mL). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give (1R, 5S)-3-[2-(3-bromo-2-methyl-phenoxy) ethoxy]-8- azabicyclo [3.2.1] octane (1.19 g, crude, HCl) as a white solid. MS (ESI) m/z: 342.0 [M+H] + . Step E. Procedure for preparation of ethyl 2-[(1R, 5S)-3-[2-(3-bromo-2-methyl- phenoxy)ethoxy]-8-azabicyclo[3.2.1]octan-8-yl]acetate To a solution of (1R, 5S)-3-[2-(3-bromo-2-methyl-phenoxy) ethoxy]-8-azabicyclo [3.2.1] octane (1.19 g, 3.16 mmol, 1.0 equiv. HCl) and ethyl 2-bromoacetate (474.78 mg, 2.84 mmol, 314.43 µL, 0.9 equiv.) in CH3CN (20 mL) was added K2CO3 (2.18 g, 15.79 mmol, 5.0 equiv.). The mixture was stirred at 60 °C for 2 hours. The reaction mixture was quenched by addition H 2 O (30 mL) at 10 °C, and then diluted with ethyl acetate (30 mL) and extracted with ethyl acetate (25 mL × 2). The combined organic layers were washed with brine (25 mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 30~60% ethyl acetate/petroleum ether) to give ethyl 2-[(1R, 5S)-3-[2-(3-bromo-2-methyl-phenoxy)ethoxy]-8- azabicyclo[3.2.1]octan-8-yl]acetate (912 mg, 2.1 mmol, 67.0% yield, 98.9% purity) as a colorless oil. MS (ESI) m/z: 428.0 [M+H] + . Step F. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[[(1R, 5S)-8-(2-methoxy-2-oxo-ethyl)-8- azabicyclo[3.2.1]octan-3-yl]oxy]ethoxy]-2-methyl-phenyl]pyri dine-2-carboxylate Ethyl 2-[(1R, 5S)-3-[2-(3-bromo-2-methyl-phenoxy)ethoxy]-8-azabicyclo[3.2. 1]octan-8- yl]acetate (250 mg, 606.33 µmol, 1.0 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tet ramethyl-1,3,2-dioxaborolan-2- yl)pyridine-2-carboxylate (408.54 mg, 666.96 µmol, 1.1 eq), [2-(2- aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-butyl-phos phane;methanesulfonate (44.16 mg, 60.63 µmol, 0.1 equiv.) and K 3 PO 4 (386.11 mg, 1.82 mmol, 3.0 equiv.) were taken up into a microwave tube in H 2 O (1 mL) and dioxane (10 mL).The sealed tube was heated at 100 °C for 2 hours under microwave. The reaction mixture was filtered under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~5% ethyl acetate/Methanol) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[2-[[(1R, 5S)-8-(2-methoxy-2-oxo-ethyl)-8-azabicyclo[3.2.1]octan- 3-yl]oxy]ethoxy]-2-methyl-phenyl]pyridine-2-carboxylate (212.5 mg, 201.7 µmol, 33.2% yield, 77.6% purity) as a yellow solid. MS (ESI) m/z: 832.5 [M+H] + . Step G. Procedure for preparation of 2-[(1R, 5S)-3-[2-[3-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-2- methyl-phenoxy]ethoxy]-8-azabicyclo[3.2.1]octan-8-yl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[2-[[(1R, 5S)-8-(2-methoxy-2-oxo-ethyl)-8-azabicyclo[3.2.1]octan-3- yl]oxy]ethoxy]-2-methyl-phenyl]pyridine-2-carboxylate (1.4 g, 1.68 mmol, 1.0 equiv.) in MeOH (10 mL) and H2O (5 mL) was added LiOH•H2O (353.05 mg, 8.41 mmol, 5.0 equiv.). The reaction mixture was stirred at 40 °C for 1 hour. The reaction mixture was treated with H 2 O (5 ml) and adjusted to pH = 4~5 with 1M HCl. Then the reaction mixture concentrated under reduced pressure to give a residue. The compound 2-[(1R, 5S)-3-[2-[3-[6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -2-tert-butoxycarbonyl-3- pyridyl]-2-methyl-phenoxy]ethoxy]-8-azabicyclo[3.2.1]octan-8 -yl]acetic acid (767 mg, crude) was obtained as a yellow solid. MS (ESI) m/z: 804.2 [M+H] + . Step H. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[[(1R,5S)-8-[2-[[3- (2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]amino]-2-oxo-ethyl]-8-azabicyclo[3.2.1]o ctan-3-yl]oxy]ethoxy]-2- methyl-phenyl]pyridine-2-carboxylate To a solution of 2-[(1R, 5S)-3-[2-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihyd ro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]ethoxy]-8- azabicyclo[3.2.1]octan-8-yl]acetic acid (120 mg, 149.26 µmol, 1.0 equiv.) and 3-(6-amino-1- methyl-indazol-3-yl)piperidine-2,6-dione (46.26 mg, 179.11 µmol, 1.2 equiv.) in pyridine (2 mL) was added EDCI (42.92 mg, 223.89 µmol, 1.5 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was added into H 2 O (5 mL). The mixture was triturated with water (5 mL) at 25°C for 30 minutes and filtered to give tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[[(1R, 5S)-8-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]-8-azabi cyclo[3.2.1]octan-3- yl]oxy]ethoxy]-2-methyl-phenyl]pyridine-2-carboxylate (137 mg, 112.9 µmol, 75.6% yield, 86.4% purity) as a brown solid. MS (ESI) m/z: 1044.9 [M+H] + Step I. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(((1R,3r,5S)-8-(2-((3-( 2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)-8-azabicyclo[3.2.1 ]octan-3-yl)oxy)ethoxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[2-[[(1R,5S)-8-[2-[[3-(2,6-dioxo-3-piperidyl)-1-m ethyl-indazol-6-yl]amino]-2-oxo- ethyl]-8-azabicyclo[3.2.1]octan-3-yl]oxy]ethoxy]-2-methyl-ph enyl]pyridine-2-carboxylate (137 mg, 131.20 µmol, 1.0 equiv. ) in DCM (1 mL) and TFA (1 mL), the mixture was stirred at 25 °C for 16 hours. The mixture was concentrated to remove DCM. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(2-(((1R,3r,5S)-8-(2-((3-(2,6-dioxopiperidin-3-yl)- 1-methyl-1H-indazol-6- yl)amino)-2-oxoethyl)-8-azabicyclo[3.2.1]octan-3-yl)oxy)etho xy)-2-methylphenyl)picolinic acid (32.8 mg, 32.2 µmol, 24.5% yield, 96.9% purity) as a white solid. MS (ESI) m/z: 988.6 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.86 (s, 1H), 9.92 (s, 1H), 8.16 (s, 1H), 8.05 - 7.98 (m, 2H), 7.77 (d, J = 8.0 Hz, 1H), 7.65 - 7.60 (m, 2H), 7.47 - 7.41 (m, 3H), 7.38 - 7.31 (m, 2H), 7.24 (dd, J = 1.6, 8.8 Hz, 1H), 7.09 (t, J = 8.0 Hz, 1H), 6.96 - 6.88 (m, 2H), 6.65 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.32 (dd, J = 5.2, 9.6 Hz, 1H), 4.08 (s, 2H), 3.93 - 3.89 (m, 5H), 3.69 (d, J = 4.4 Hz, 2H), 3.62 (s, 2H), 3.08 (s, 2H), 3.02 (t, J = 5.6 Hz, 2H), 2.70 - 2.60 (m, 2H), 2.35 - 2.30 (m, 1H), 2.20 - 2.10 (m, 1H), 2.07 - 1.99 (m, 5H), 1.91 (s, 3H), 1.83 (d, J = 14.0 Hz, 4H). Example 147. Preparation of Compound 176a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(2- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1-yl)-2- oxoethyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of methyl 2-(4-hydroxycyclohexyl) acetate To a solution of 2-(4-hydroxycyclohexyl) acetic acid (2 g, 12.64 mmol, 1.0 equiv.) in MeOH (20 mL) was added H2SO4 (124.00 mg, 1.26 mmol, 67.39 µL, 0.1 equiv.). The mixture was stirred at 70 °C for 3 hours under N2 atmosphere. The reaction was diluted with water (20 mL) and extracted with DCM (50 mL × 2). The combined organic layers were washed with brine (50 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. It was used in the next step without further purification to give methyl 2-(4- hydroxycyclohexyl) acetate (2 g, 11.6 mmol, 91.8 % yield) as a white oil. Step B. Procedure for preparation of methyl 2-(4-(3-bromo-2-methylphenoxy) cyclohexyl) acetate To a solution of methyl 2-(4-hydroxycyclohexyl) acetate (2 g, 11.61 mmol, 1.0 equiv.) in toluene (20 mL) was added 2-(tributyl-λ5-phosphanylidene) acetonitrile (4.20 g, 17.42 mmol, 1.5 equiv.) and 3-bromo-2-methylphenol (2.61 g, 13.94 mmol, 1.2 equiv.). The mixture was degassed and purged with N 2 and stirred at 120 °C for 2 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 15 % ethyl acetate / petroleum ether) to give methyl 2-(4-(3-bromo-2-methylphenoxy) cyclohexyl) acetate (3.8 g, 10.9 mmol, 94.0 % yield, 98.0 % purity) as a white oil. 1 H NMR (400 MHz, CDCl3-d) δ = 7.08 - 7.03 (m, 1H), 6.89 (t, J = 8.0 Hz, 1H), 6.75 - 6.66 (m, 1H), 4.47 (s, 1H), 3.61 (s, 3H), 2.28 - 2.15 (m, 5H), 2.09 - 1.93 (m, 2H), 1.83 - 1.77 (m, 1H), 1.51 (d, J = 13.2 Hz, 4H), 1.44 - 1.32 (m, 2H) Step C. Procedure for preparation of methyl 2-((1r,4r)-4-(3-bromo-2-methylphenoxy) cyclohexyl) acetate The methyl 2-(4-(3-bromo-2-methylphenoxy) cyclohexyl) acetate residue was purified by prep-HPLC to give methyl 2-((1r,4r)-4-(3-bromo-2-methylphenoxy) cyclohexyl) acetate (200 mg, 586.10 µmol, 11.11 % yield) as a yellow oil. 1 H NMR (400 MHz, CDCl 3 -d) δ = 7.06 (d, J = 8.0 Hz, 1H), 6.89 (t, J = 8.0 Hz, 1H), 6.72 (d, J = 8.0 Hz, 1H), 4.02 (tt, J = 4.4, 10.6 Hz, 1H), 3.61 (s, 3H), 2.22 (s, 3H), 2.17 (d, J = 6.4 Hz, 2H), 2.11 - 2.01 (m, 2H), 1.82 - 1.77 (m, 2H), 1.54 (s, 1H), 1.48 - 1.37 (m, 2H), 1.10 - 0.99 (m, 2H) Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(2-methoxy-2-o xoethyl)cyclohexyl)oxy)-2- methylphenyl)picolinate To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) picolinate (195.82 mg, 319.69 µmol, 1.0 equiv.) in dioxane (2 mL) was added Ad2nBuP Pd G 3 (23.28 mg, 31.97 µmol, 0.1 equiv.), KF (1.5 M, 639.38 µL, 3.0 equiv.), and methyl 2-((1r,4r)-4-(3-bromo-2- methylphenoxy) cyclohexyl) acetate (120 mg, 351.66 µmol, 1.1 equiv.). The mixture was stirred at 100 °C for 1 hour. The reaction was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 30 % ethyl acetate / petroleum ether) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(2-methoxy-2-o xoethyl)cyclohexyl)oxy)-2- methylphenyl)picolinate (200 mg, 267.7 µmol, 83.7 % yield) as a yellow oil. MS (ESI) m/z: 747.8 [M+H] + . Step E. Procedure for preparation of 2-((1r,4r)-4-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)cyclohexyl)acetic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(2-methoxy-2-oxoethyl)cyclohexyl) oxy)-2- methylphenyl)picolinate (190 mg, 254.38 µmol, 1.0 equiv.) in THF (2 mL) was added LiOH•H2O (32.02 mg, 763.14 µmol, 3.0 equiv.) and H2O (0.5 mL). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was acidified to pH = 5-6 with citric acid, filtered, and concentrated under reduced pressure to give a residue. 2-((1r,4r)-4-(3-(6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)cyclohexyl)acet ic acid (150 mg, 204.67 µmol, 80.46 % yield) was provided as a yellow solid. It was used in the next step without further purification. MS (ESI) m/z: 733.4 [M+H] + . Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(2-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-2-oxoethyl)cyclohexyl )oxy)-2- methylphenyl)picolinate To a solution of 2-((1r,4r)-4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)cyclohexyl)acetic acid (40 mg, 54.58 µmol, 1.0 equiv.) in DMF (1 mL) was added HATU (62.26 mg, 163.74 µmol, 3.0 equiv.), DIEA (10.58 mg, 81.87 µmol, 14.26 µL, 1.5 equiv.), and 3-(1-methyl-6-(piperazin-1-yl)-1H-indazol-3-yl)piperidine-2, 6-dione (19.65 mg, 60.04 µmol, 1.1 equiv.). The mixture was stirred at 50 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by prep- TLC (SiO2, petroleum ether / ethyl acetate = 0 / 1) to give tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r) -4-(2-(4-(3-(2,6-dioxopiperidin- 3-yl)-1-methyl-1H-indazol-6-yl)piperazin-1-yl)-2-oxoethyl)cy clohexyl)oxy)-2- methylphenyl)picolinate (55 mg, 52.8 µmol, 96.7 % yield) as a yellow oil. MS (ESI) m/z: 1043.5 [M+H] + . Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(2-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-2-oxoethyl)cyclohexyl )oxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(2-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-6- yl)piperazin-1-yl)-2-oxoethyl)cyclohexyl)oxy)-2-methylphenyl )picolinate (70 mg, 67.16 µmol, 1.0 equiv.) in DCM (1 mL) was added TFA (7.66 mg, 67.16 µmol, 4.97 µL, 1.0 equiv.). The mixture was stirred at 40 °C for 4 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1r,4r)-4-(2-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin -1-yl)-2- oxoethyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (25.0 mg, 24.8 µmol, 36.9 % yield, 97.7 % purity) as a yellow solid. MS (ESI) m/z: 986.4 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.92 - 12.81 (m, 1H), 12.76 - 12.31 (m, 1H), 10.86 (s, 1H), 8.04 (d, J = 8.0 Hz, 1H), 7.80 (d, J = 8.0 Hz, 1H), 7.69 - 7.59 (m, 1H), 7.53 (d, J = 8.8 Hz, 1H), 7.50 - 7.43 (m, 3H), 7.41 - 7.33 (m, 2H), 7.12 - 7.04 (m, 1H), 6.99 - 6.88 (m, 4H), 6.62 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.27 (dd, J = 5.2, 9.2 Hz, 1H), 4.24 - 4.15 (m, 1H), 3.92 ( s, 2H), 3.90 (s, 3H), 3.65 (s, 4H), 3.24 (d, J = 4.0 Hz, 2H), 3.19 (d, J = 1.6 Hz, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.61 (t, J = 6.0 Hz, 2H), 2.40 - 2.22 (m, 4H), 2.16 (dd, J = 5.6, 12.7 Hz, 1H), 2.12 - 2.05 (m, 2H), 1.88 (s, 3H), 1.82 (d, J = 13.2 Hz, 2H), 1.44 - 1.34 (m, 2H), 1.22 - 1.12 (m, 2H) Example 148. Preparation of Compound 177a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[(3R,5S)-4- [2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]- 2-oxo-ethyl]-3,5-dimethyl- piperazin-1-yl]-2,2-difluoro-propoxy]-2-methyl-phenyl]pyridi ne-2-carboxylic acid Step A. Procedure for preparation of 3-[tert-butyl(diphenyl)silyl]oxy-2,2-difluoro- propan-1-ol To a solution of 2, 2-difluoropropane-1,3-diol (12.0 g, 107.07 mmol, 1.0 equiv.) in THF (120 mL) was added NaH (4.71 g, 117.78 mmol, 60% purity, 1.1 equiv.) in portions under N2 at 0 °C. The reaction mixture was degassed and purged with N 2 and stirred at 25 °C for 1 hour under N 2 atmosphere. Tert-butyl-chloro-diphenyl-silane (32.37 g, 117.78 mmol, 30.25 mL, 1.1 equiv.) was added to the mixture, and then the mixture was stirred under N2 at 25 °C for 15 hours. The reaction mixture was quenched by addition NH4Cl (200 mL) at 0 °C, and extracted with ethyl acetate (300 mL × 3). The combined organic layers were washed with brine (300 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~15% ethyl acetate/petroleum ether) to give 3-[tert-butyl(diphenyl)silyl]oxy-2,2-difluoro-propan-1-ol (42.0 g, crude) was obtained as colorless oil 1 H NMR (400 MHz, CDCl 3 -d) δ = 7.68 (d, J = 6.4 Hz, 4H), 7.50 - 7.38 (m, 6H), 4.00 - 3.86 (m, 4H), 1.88 - 1.76 (m, 1H), 1.08 (s, 9H). Step B. Procedure for preparation of [3-(3-bromo-2-methyl-phenoxy)-2, 2-difluoro- propoxy]-tert-butyl-diphenyl-silane A mixture of 3-bromo-2-methyl-phenol (10.0 g, 53.47 mmol, 1.0 equiv.), 3-[tert- butyl(diphenyl)silyl]oxy-2,2-difluoro-propan-1-ol (22.49 g, 64.16 mmol, 1.2 equiv.), 2- (tributyl-λ5-phosphanylidene)acetonitrile (15.49 g, 64.16 mmol, 1.2 equiv.) in toluene (200 mL) was degassed and purged with N2 and stirred at 120 °C for 16 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by flash silica gel chromatography (Eluent of 0~5% ethyl acetate/petroleum ether) to give [3-(3-bromo-2-methyl-phenoxy)-2, 2-difluoro-propoxy]-tert-butyl-diphenyl-silane (19.0 g, 36.5 mmol, 68.4% yield) as colorless oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.57 (d, J = 7.2 Hz, 4H), 7.50 - 7.45 (m, 2H), 7.42 - 7.37 (m, 4H), 7.25 (d, J = 7.2 Hz, 1H), 7.15 - 7.08 (m, 2H), 4.44 (t, J = 12.0 Hz, 2H), 4.02 (t, J = 12.8 Hz, 2H), 2.11 (s, 3H), 0.97 (s, 9H). Step C. Procedure for preparation of 3-(3-bromo-2-methyl-phenoxy)-2, 2-difluoro- propan-1-ol A mixture of [3-(3-bromo-2-methyl-phenoxy)-2,2-difluoro-propoxy]-tert-but yl-diphenyl- silane (19.0 g, 36.57 mmol, 1.0 equiv.) and TBAF (1 M, 73.15 mL, 2.0 equiv.) in THF (150 mL), and then the mixture was stirred at 25 °C for 3 hours. The reaction mixture was quenched by addition H2O (200 mL) at 25 °C, and then diluted with ethyl acetate (200 mL) and extracted with ethyl acetate (200 mL × 1). The combined organic layers were washed with H 2 O (200mL × 2), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~12% ethyl acetate/petroleum ether) to give 3-(3-bromo-2-methyl-phenoxy)-2, 2-difluoro-propan-1-ol (7.2 g, 25.6 mmol, 70.0% yield) was obtained as light yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.23 (d, J = 8.0 Hz, 1H), 7.13 (t, J = 8.0 Hz, 1H), 7.08 - 7.04 (m, 1H), 5.67 (t, J = 6.4 Hz, 1H), 4.33 (t, J = 12.8 Hz, 2H), 3.79 (dt, J = 6.0, 13.6 Hz, 2H), 3.17 (s, 1H), 2.25 (s, 3H). Step D. Procedure for preparation of 3-(3-bromo-2-methyl-phenoxy)-2, 2-difluoro- propanal To a solution of (COCl)2 (4.52 g, 35.58 mmol, 3.11 mL, 2.0 equiv.) in DCM (100 mL) was added DMSO (5.56 g, 71.15 mmol, 5.56 mL, 4.0 equiv.) in DCM (20 mL) under -70 °C. The solution was stirred for 1 hour.3-(3-Bromo-2-methyl-phenoxy)-2,2-difluoro-propan-1-ol (5.0 g, 17.79 mmol, 1.0 equiv.) in DCM (20 mL) was added into the mixture and stirred for 1 hour. TEA (10.80 g, 106.73 mmol, 14.85 mL, 6.0 equiv.) in DCM (10 mL) was added into the mixture and stirred at -70 °C for 2 hours. The reaction mixture was quenched by addition H2O (100 mL) at 0 °C, and then diluted with DCM (200 mL) and extracted with DCM (100 mL × 2). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give 3-(3-bromo-2-methyl-phenoxy)-2, 2-difluoro-propanal (5.0 g, crude) as yellow oil, was used into the next step without further purification. Step E. Procedure for preparation of ethyl 2-[(2R,6S)-4-[3-(3-bromo-2-methyl-phenoxy)- 2,2-difluoro-propyl]-2,6-dimethyl-piperazin-1-yl]acetate To a solution of 3-(3-bromo-2-methyl-phenoxy)-2,2-difluoro-propanal (5.0 g, 17.92 mmol, crude purity, 1.0 equiv.), ethyl 2-[(2R,6S)-2,6-dimethylpiperazin-1-yl]acetate (5.09 g, 21.50 mmol, 1.2 equiv., HCl), 4A MS (2.0 g) in DCM (150 mL). The mixture was stirred at 25 °C for 1 hour, was added sodium triacetoxyborohydride (15.19 g, 71.66 mmol, 4.0 equiv.).The mixture was stirred at 25 °C for 19 hours. The reaction mixture was quenched by addition water (200 mL) at 25°C, and then diluted with DCM (100 mL) and extracted with DCM 200 mL (200 mL × 1). The combined organic layers were concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~20% ethyl acetate/petroleum ether) to give ethyl 2-[(2R,6S)-4-[3-(3-bromo-2-methyl-phenoxy)-2,2- difluoro-propyl]-2,6-dimethyl-piperazin-1-yl]acetate (0.9 g, 1.8 mmol, 10.3% yield, 95% purity) as light brown oil. 1 H NMR (400 MHz, CDCl 3 ) δ = 7.22 (d, J = 7.6 Hz, 1H), 7.03 (t, J = 8.0 Hz, 1H), 6.82 (d, J = 8.0 Hz, 1H), 4.25 (t, J = 11.6 Hz, 2H), 4.16 (q, J = 7.2 Hz, 2H), 3.54 (s, 2H), 2.97 (s, 2H), 2.85 (t, J = 13.6 Hz, 2H), 2.74 (d, J = 10.8 Hz, 2H), 2.33 (s, 3H), 2.16 (t, J = 10.8 Hz, 2H), 1.27 (t, J = 7.2 Hz, 3H), 1.04 (d, J = 6.4 Hz, 6H) Step F. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(3R,5S)-4-(2-ethox y-2-oxo-ethyl)-3,5-dimethyl- piperazin-1-yl]-2,2-difluoro-propoxy]-2-methyl-phenyl]pyridi ne-2-carboxylate Ethyl 2-[(2R,6S)-4-[3-(3-bromo-2-methyl-phenoxy)-2,2-difluoro-prop yl]-2,6-dimethyl- piperazin-1-yl]acetate (900 mg, 1.94 mmol, 1.0 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tet ramethyl-1,3,2-dioxaborolan-2- yl)pyridine-2-carboxylate (1.25 g, 2.04 mmol, 1.05 equiv.), K 3 PO 4 (1.24 g, 5.83 mmol, 3.0 equiv.), and Ad2nBuP Pd G3 (282.91 mg, 388.47 µmol, 0.2 equiv.) were taken up into a microwave tube in dioxane (8 mL) and H2O (0.5 mL).The sealed tube was heated at 100 °C for 2 hours under microwave. The crude reaction mixture was concentrated in vacuo to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~45% ethyl acetate/petroleum ether) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[(3R,5S)-4-(2-ethoxy-2-oxo-ethy l)-3,5-dimethyl-piperazin-1- yl]-2,2-difluoro-propoxy]-2-methyl-phenyl]pyridine-2-carboxy late (1.1 g, 1.2 mmol, 63.3% yield, 97.1% purity) was obtained as a yellow solid. MS (ESI) m/z: 869.4 [M+H] + . Step G. Procedure for preparation of 2-[(2R,6S)-4-[3-[3-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-2- methyl-phenoxy]-2,2-difluoro-propyl]-2,6-dimethyl-piperazin- 1-yl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[3-[(3R,5S)-4-(2-ethoxy-2-oxo-ethyl)-3,5-dimethyl -piperazin-1-yl]-2,2-difluoro- propoxy]-2-methyl-phenyl]pyridine-2-carboxylate (1.0 g, 1.15 mmol, 1.0 equiv.) in H 2 O (1 mL) and MeOH (5 mL), was added LiOH•H2O (241.44 mg, 5.75 mmol, 5.0 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to remove MeOH. The residue was diluted with H 2 O (5 mL) and adjust pH=4 by citric acid, extracted with DCM (100 mL × 2). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give 2-[(2R,6S)-4-[3-[3-[6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -2-tert-butoxycarbonyl-3- pyridyl]-2-methyl-phenoxy]-2,2-difluoro-propyl]-2,6-dimethyl -piperazin-1-yl]acetic acid (870 mg, 1.0 mmol, 89.9% yield) as a yellow solid. MS (ESI) m/z: 841.4 [M+H] + . Step H. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(3R,5S)-4-[2-[[3-( 2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]amino]-2-oxo-ethyl]-3,5-dimethyl-piperaz in-1-yl]-2,2-difluoro- propoxy]-2-methyl-phenyl]pyridine-2-carboxylate To a solution of 2-[(2R,6S)-4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3, 4-dihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]-2,2-difluoro-propyl]- 2,6-dimethyl-piperazin-1-yl]acetic acid (130 mg, 154.58 µmol, 1.0 equiv.) and 3-(6-amino-1- methyl-indazol-3-yl)piperidine-2,6-dione (51.90 mg, 200.96 µmol, 1.3 equiv.) in DMF (2 mL) was added HATU (88.17 mg, 231.87 µmol, 1.5 eq) and DIEA (99.89 mg, 772.91 µmol, 134.62 µL, 5.0 equiv.). The mixture was stirred at 25 °C for 16 hours. The mixture was added into H2O (5.0 ml) and filtered to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(3R,5S)-4-[2-[[3-(2,6- dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-3,5-dimethyl-piperazin-1-yl ]-2,2-difluoro-propoxy]-2- methyl-phenyl]pyridine-2-carboxylate (166 mg, 153.5 µmol, 99.3% yield) as a brown solid. MS (ESI) m/z: 1081.7 [M+H] + . Step I. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[(3R,5S)-4-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-3,5-dimethyl-piperazin-1-yl]-2,2-di fluoro-propoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[3-[(3R,5S)-4-[2-[[3-(2,6-dioxo-3-piperidyl)-1-me thyl-indazol-6-yl]amino]-2-oxo- ethyl]-3,5-dimethyl-piperazin-1-yl]-2,2-difluoro-propoxy]-2- methyl-phenyl]pyridine-2- carboxylate (166 mg, 153.53 µmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL). The mixture was stirred at 25 °C for 16 hours. The mixture was concentrated to remove DCM. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[3-[(3R,5S)-4-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-3,5-dimethyl-piperazin-1-yl]-2,2-difl uoro-propoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid (53.6 mg, 48.9 µmol, 31.8% yield, 93.5% purity) as a yellow solid. MS (ESI) m/z: 513.4 [M/2+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.94 - 12.12 (m, 1H), 10.88 (s, 1H), 9.77 (s, 1H), 8.14 (s, 1H), 8.02 (d, 2H), 7.81 (d, J = 8.0 Hz, 1H), 7.63 (t, J = 8.0 Hz, 2H), 7.54 - 7.47 (m, 3H), 7.37 - 7.34 (m, 2H), 7.21 (d, J = 1.2, 8.8 Hz, 1H), 7.16 - 7.11 (m, 1H), 7.00 (t, J = 8.4 Hz, 2H), 6.72 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.35 - 4.32 (m, 3H), 3.92 - 3.90 (m, 5H), 3.02 (t, J = 5.6 Hz, 2H), 3.00 - 2.91 (m, 3H), 2.83 - 2.71 (m, 4H), 2.70 - 2.61 (m, 2H), 2.38 - 2.32 (m, 1H), 2.20 - 2.14 (m, 3H), 1.93 (s, 3H), 0.95 (d, J = 6.0 Hz, 6H) 19 F NMR (400 MHz, DMSO-d6) δ =106.40 Example 149. Preparation of Compound 179a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[(3R,5S)-4- [2-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]piper azin-1-yl]-2-oxo-ethyl]-3,5- dimethyl-piperazin-1-yl]-2,2-difluoro-propoxy]-2-methyl-phen yl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(3R,5S)-4-[2-[4-[3 -(2,6-dioxo-3-piperidyl)-1- methyl-indazol-7-yl]piperazin-1-yl]-2-oxo-ethyl]-3,5-dimethy l-piperazin-1-yl]-2,2- difluoro-propoxy]-2-methyl-phenyl]pyridine-2-carboxylate To a solution of 2-[(2R,6S)-4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3, 4-dihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]-2,2-difluoro-propyl]- 2,6-dimethyl-piperazin-1-yl]acetic acid (120 mg, 142.69 µmol, 1.0 equiv.) and 3-(1-methyl-7- piperazin-1-yl-indazol-3-yl)piperidine-2,6-dione (51.39 mg, 156.96 µmol, 1.1 equiv.) in DMF (2 mL) was added HATU (81.38 mg, 214.04 µmol, 1.5 equiv.) and DIEA (92.21 mg, 713.46 µmol, 124.27 µL, 5.0 equiv.). The mixture was stirred at 25 °C for 16 hours. The mixture was added into H 2 O (5.0 ml) and filtered to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(3R,5S)-4-[2-[4-[3 -(2,6-dioxo-3-piperidyl)-1- methyl-indazol-7-yl]piperazin-1-yl]-2-oxo-ethyl]-3,5-dimethy l-piperazin-1-yl]-2,2-difluoro- propoxy]-2-methyl-phenyl]pyridine-2-carboxylate (145 mg, 126.0 µmol, 88.3% yield) was obtained as a brown solid. MS (ESI) m/z: 1150.7 [M+H] + . Step B. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[(3R,5S)-4-[2-[4-[3-(2,6-dioxo- 3-piperidyl)-1-methyl- indazol-7-yl]piperazin-1-yl]-2-oxo-ethyl]-3,5-dimethyl-piper azin-1-yl]-2,2-difluoro- propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[3-[(3R,5S)-4-[2-[4-[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-7-yl]piperazin-1- yl]-2-oxo-ethyl]-3,5-dimethyl-piperazin-1-yl]-2,2-difluoro-p ropoxy]-2-methyl- phenyl]pyridine-2-carboxylate (145 mg, 126.05 µmol, 1.0 equiv.) in DCM (1 mL) and TFA (1 mL). The mixture was stirred at 25 °C for 16 hours. The mixture was concentrated to remove DCM. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(3R,5S)-4-[2-[4-[3 -(2,6-dioxo-3-piperidyl)-1- methyl-indazol-7-yl]piperazin-1-yl]-2-oxo-ethyl]-3,5-dimethy l-piperazin-1-yl]-2,2-difluoro- propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid (49.2 mg, 43.4 µmol, 34.4% yield, 96.6% purity) as a white solid. MS (ESI) m/z: 548.0 [M/2+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.89 (d, J = 9.6 Hz, 1H), 10.88 (s, 1H), 8.13 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.49 - 7.31 (m, 6H), 7.17 - 7.10 (m, 1H), 7.05 - 6.95 (m, 4H), 6.71 (d, J = 8.0 Hz, 1H), 4.98 (s, 2H), 4.38 - 4.29 (m, 3H), 4.26 (s, 3H), 3.92 (t, J = 5.6 Hz, 2H), 3.10 (s, 4H), 3.03 (t, J = 5.6 Hz, 4H), 2.86 (t, J = 13.6 Hz, 4H), 2.75 - 2.57 (m, 7H), 2.33 (s, 2H), 2.16 (dd, J = 5.2, 12.8 Hz, 1H), 2.07 (s, 2H), 1.92 (s, 3H), 0.92 (d, J = 4.8 Hz, 6H). 19 F NMR (400 MHz, DMSO-d 6 ) δ = 106.40. Example 150. Preparation of Compound 182a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(2- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pip erazin-1-yl)-2- oxoethyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(2-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)-2-oxoethyl)cyclohexyl )oxy)-2- methylphenyl)picolinate To a solution of 2-((1r,4r)-4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)cyclohexyl)acetic acid (30 mg, 40.93 µmol, 1.0 equiv.) in pyridine (1 mL) was added EDCI (10.20 mg, 53.21 µmol, 1.3 equiv.) and 3-(1-methyl-7-(piperazin-1-yl)-1H- indazol-3-yl)piperidine-2,6-dione (13.40 mg, 40.93 µmol, 1.0 equiv.). The mixture was stirred at 60 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by prep-TLC (SiO 2 , petroleum ether / ethyl acetate = 0 / 1) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- (3-(((1r,4r)-4-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1 H-indazol-7-yl)piperazin-1-yl)-2- oxoethyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (30 mg, 28.8 µmol, 70.3 % yield) as a yellow oil. MS (ESI) m/z: 1042.5 [M+H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(2-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)-2-oxoethyl)cyclohexyl )oxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(2-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-7- yl)piperazin-1-yl)-2-oxoethyl)cyclohexyl)oxy)-2-methylphenyl )picolinate (50 mg, 47.97 µmol, 1.0 equiv.) in DCM (1 mL) was added TFA (5.47 mg, 47.97 µmol, 3.55 µL, 1.0 equiv.). The mixture was stirred at 40 °C for 4 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1r,4r)-4-(2-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin -1-yl)-2- oxoethyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (16.7 mg, 15.8 µmol, 33.0 % yield, 93.5 % purity) as a yellow solid. MS (ESI) m/z: 986.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.10 - 12.26 (m, 2H), 10.89 (s, 1H), 8.04 (d, J = 8.0 Hz, 1H), 7.84 - 7.75 (m, 1H), 7.63 (d, J = 7.6 Hz, 1H), 7.52 - 7.41 (m, 4H), 7.40 - 7.32 (m, 2H), 7.12 - 7.00 (m, 3H), 6.95 (dd, J = 8.4, 14.8 Hz, 2H), 6.62 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.35 (dd, J = 5.2, 9.6 Hz, 1H), 4.27 (s, 3H), 4.20 (t, J = 9.2 Hz, 1H), 3.92 (s, 2H), 3.29 - 3.14 (m, 4H), 3.04 (d, J = 5.2 Hz, 2H), 2.94 - 2.72 (m, 2H), 2.71 - 2.52 (m, 6H), 2.32 (d, J = 4.4 Hz, 2H), 2.17 (dd, J = 5.6, 13.2 Hz, 1H), 2.12 - 2.03 (m, 2H), 1.88 (s, 3H), 1.83 (d, J = 12.0 Hz, 2H), 1.45 - 1.33 (m, 2H), 1.22 - 1.11 (m, 2H) Example 151. Preparation of Compound 183a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4-[3-[(3S,5R)-4- [2-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]piper azin-1-yl]-2-oxo-ethyl]-3,5- dimethyl-piperazin-1-yl]propoxy]-2-methyl-phenyl]pyridine-2- carboxylic acid Step A. Procedure for preparation of (3R,5S)-tert-butyl 4-(2-methoxy-2-oxoethyl)-3,5- dimethylpiperazine-1-carboxylate A mixture of methyl 2-bromoacetate (9.35 g, 56.00 mmol, 6.19 mL, 1.5 equiv.), (3R, 5S)-tert- butyl 3,5-dimethylpiperazine-1-carboxylate (8 g, 37.33 mmol, 1 equiv.), and K2CO3 (15.48 g, 111.99 mmol, 3 equiv.) in CH3CN (100 mL) was degassed and purged with N2 and stirred at 60 °C for 3 hours under N 2 atmosphere. The reaction mixture was diluted with water (100 mL) and extracted with ethyl acetate (120 mL × 3). The combined organic layers were washed with brine (120 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~25% ethyl acetate/petroleum ether) to give (3R,5S)-tert-butyl 4-(2-methoxy-2- oxoethyl)-3,5-dimethylpiperazine-1-carboxylate (10 g, 33.2 mmol, 89.1% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 4.10 - 4.05 (m, 2H), 3.67 (s, 2H), 3.50 (s, 2H), 2.78 - 2.69 (m, 2H), 2.49 - 2.32 (m, 2H), 1.39 (s, 9H), 1.20 - 1.16 (m, 3H), 0.97 (d, J = 6.4 Hz, 6H) Step B. Procedure for preparation of methyl 2-((2R,6S)-2,6-dimethylpiperazin-1- yl)acetate A mixture of (3R,5S)-tert-butyl 4-(2-methoxy-2-oxoethyl)-3,5-dimethylpiperazine-1- carboxylate (10 g, 33.29 mmol, 1 equiv.) and HCl/1,4-dioxane (4 M, 83.22 mL, 10 equiv.) in DCM (80 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 25 °C for 16 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give methyl 2-((2R,6S)-2,6-dimethylpiperazin-1-yl)acetate (12.25 g, crude, HCl) as a yellow solid. 1 H NMR (400 MHz,CD3OD) δ = 4.44 (s, 2H), 4.36 (q, J = 7.2 Hz, 2H), 4.29 - 4.21 (m, 2H), 3.74 - 3.67 (m, 2H), 3.58 - 3.50 (m, 2H), 1.49 (d, J = 6.4 Hz, 6H), 1.35 (t, J = 7.2 Hz, 3H) Step C. Procedure for preparation of 1-bromo-4-(3-bromopropoxy)-2-methylbenzene A mixture of 4-bromo-3-methylphenol (3 g, 16.04 mmol, 1 equiv.), 1,3-dibromopropane (16.19 g, 80.20 mmol, 8.18 mL, 5 equiv.) and K 2 CO 3 (6.65 g, 48.12 mmol, 3 equiv.) in CH 3 CN (30 mL) was degassed and purged with N 2 and stirred at 70 °C for 12 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~5% ethyl acetate/petroleum ether) to give compound 1-bromo-4-(3-bromopropoxy)-2-methylbenzene (2.5 g, 8.1 mmol, 50.6% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.44 (d, J = 8.8 Hz, 1H), 6.98 (d, J = 2.8 Hz, 1H), 6.74 (dd, J = 8.8, 2.8 Hz, 1H), 4.08 - 4.02 (m, 2H), 3.65 (t, J = 6.4 Hz, 2H), 2.30 (s, 3H), 2.26 – 2.19 (m, 2H) Step D. Procedure for preparation of ethyl 2-((2R,6S)-4-(3-(4-bromo-3- methylphenoxy)propyl)-2,6-dimethylpiperazin-1-yl)acetate A mixture of 1-bromo-4-(3-bromopropoxy)-2-methylbenzene (2.34 g, 7.60 mmol, 1.2 equiv.), methyl 2-((2R,6S)-2,6-dimethylpiperazin-1-yl)acetate (1.5 g, 6.34 mmol, 1 equiv., HCl) and DIEA (4.09 g, 31.68 mmol, 5.52 mL, 5 equiv.) in DMF (40 mL) was degassed and purged with N 2 and stirred at 60 °C for 16 hours under N 2 atmosphere. The reaction mixture was diluted with water (100 mL) and extracted with DCM (100 mL × 3). The combined organic layers were washed with brine (100 mL × 6), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~7% DCM / MeOH) to give compound ethyl 2-((2R,6S)-4-(3- (4-bromo-3-methylphenoxy)propyl)-2,6-dimethylpiperazin-1-yl) acetate (1.3 g, 3.0 mmol, 48.0% yield) as an orange oil. 1 H NMR (400 MHz, CD 3 OD) δ = 7.37 (d, J = 8.8 Hz, 1H), 6.86 (d, J = 3.0 Hz, 1H), 6.66 (dd, J = 8.8, 3.2 Hz, 1H), 4.16 (q, J = 7.2 Hz, 2H), 3.98 (t, J = 6.0 Hz, 2H), 3.55 (s, 2H), 3.04 - 2.98 (m, 2H), 2.86 - 2.84 (m, 1H), 2.82 (s, 1H), 2.54 - 2.46 (m, 2H), 2.33 (s, 3H), 2.00 - 1.92 (m, 2H), 1.92 - 1.85 (m, 2H), 1.27 (t, J = 7.2 Hz, 3H), 1.08 (d, J = 6.4 Hz, 6H) Step E. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(3-((3R,5S)-4-(2-ethoxy-2- oxoethyl)-3,5- dimethylpiperazin-1-yl)propoxy)-2-methylphenyl)picolinate Ethyl 2-((2R,6S)-4-(3-(4-bromo-3-methylphenoxy)propyl)-2,6-dimethy lpiperazin-1-yl)acetate (400 mg, 935.95 µmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)picolinate (687.97 mg, 1.12 mmol, 1.2 equiv.), Ad2nBuP Pd G3(cataCXium® A Pd G3 ) (68.16 mg, 93.59 µmol, 0.1 equiv.) and KF (1.5 M, 1.87 mL, 3 equiv.) were taken up into a microwave tube in 1,4-dioxane (11 mL).The sealed tube was heated at 100 °C for 60 minutes under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by reverse-phase HPLC (with 0.1% HCOOH) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(4-(3-((3R,5S)-4-(2- ethoxy-2-oxoethyl)-3,5-dimethylpiperazin-1-yl)propoxy)-2-met hylphenyl)picolinate (410 mg, 477.4 µmol, 51.0% yield, 97.0% purity) as a yellow solid. MS (ESI) m/z: 833.4 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.12 - 12.55 (m, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.77 (d, J = 8.0 Hz, 1H), 7.60 (d, J = 7.6 Hz, 1H), 7.48 - 7.41 (m, 3H), 7.39 - 7.31 (m, 2H), 6.92 (d, J = 8.8 Hz, 1H), 6.85 (d, J = 8.4 Hz, 1H), 6.81 (s, 1H), 6.75 - 6.69 (m, 1H), 4.96 (s, 2H), 4.06 (q, J = 7.2 Hz, 2H), 3.98 (t, J = 6.0 Hz, 2H), 3.86 (t, J = 6.0 Hz, 2H), 3.45 (s, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.87 - 2.80 (m, 2H), 2.72 (d, J = 10.4 Hz, 2H), 2.34 (t, J = 6.8 Hz, 2H), 2.00 (s, 3H), 1.87 -1.78 (m, 2H), 1.66 (t, J = 10.4 Hz, 2H), 1.17 (t, J = 7.2 Hz, 3H), 1.03 (s, 9H), 0.95 (d, J = 6.4 Hz, 6H) Step F. Procedure for preparation of 2-((2R,6S)-4-(3-(4-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-3- methylphenoxy)propyl)-2,6-dimethylpiperazin-1-yl)acetic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4-(3-((3R,5S)-4-(2-ethoxy-2-oxoethyl)-3,5-dimethylpip erazin-1-yl)propoxy)-2- methylphenyl)picolinate (400 mg, 480.17 µmol, 1 equiv.) and LiOH·H 2 O (100.75 mg, 2.40 mmol, 5 equiv.) in THF (4 mL) and H 2 O (1 mL) was degassed and purged with N 2 and stirred at 40 °C for 7 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give 2-((2R,6S)-4-(3-(4-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4 - dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-3- methylphenoxy)propyl)-2,6-dimethylpiperazin-1-yl)acetic acid (400 mg, crude) as a yellow solid. MS (ESI) m/z: 805.7 [M+H] + Step G. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[3-[(3S,5R)-4-[2-[4-[3 -(2,6-dioxo-3-piperidyl)-1- methyl-indazol-7-yl]piperazin-1-yl]-2-oxo-ethyl]-3,5-dimethy l-piperazin-1-yl]propoxy]- 2-methyl-phenyl]pyridine-2-carboxylate To a solution of 2-((2R,6S)-4-(3-(4-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4 - dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-3- methylphenoxy)propyl)-2,6-dimethylpiperazin-1-yl)acetic acid (80 mg, 99.38 µmol, 1 equiv.), 3-(1-methyl-7-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (39.04 mg, 119.26 µmol, 1.2 equiv.) in DMF (1 mL) was added DIEA (51.38 mg, 397.52 µmol, 69.24 µL, 4 equiv.) and HATU (56.68 mg, 149.07 µmol, 1.5 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was quenched by water (2 mL) at 25 °C and precipitated in water to give compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[4-[3-[(3S,5R)-4-[2-[4-[3-(2,6-dioxo-3-piperidyl)-1-me thyl-indazol-7-yl]piperazin-1- yl]-2-oxo-ethyl]-3,5-dimethyl-piperazin-1-yl]propoxy]-2-meth yl-phenyl]pyridine-2- carboxylate (180 mg, crude) as a white solid. MS (ESI) m/z: 1114.9[M+H] + Step H. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[4-[3-[(3S,5R)-4-[2-[4-[3-(2,6-dioxo- 3-piperidyl)-1-methyl- indazol-7-yl]piperazin-1-yl]-2-oxo-ethyl]-3,5-dimethyl-piper azin-1-yl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[4-[3-[(3S,5R)-4-[2-[4-[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-7-yl]piperazin-1- yl]-2-oxo-ethyl]-3,5-dimethyl-piperazin-1-yl]propoxy]-2-meth yl-phenyl]pyridine-2- carboxylate (180 mg, 161.53 µmol, 1 equiv.) in DCM (2 mL) was added TFA (18.42 mg, 161.53 µmol, 11.96 µL, 1 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]- 3-[4-[3-[(3S,5R)-4-[2-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl -indazol-7-yl]piperazin-1-yl]-2- oxo-ethyl]-3,5-dimethyl-piperazin-1-yl]propoxy]-2-methyl-phe nyl]pyridine-2-carboxylic acid as a white solid. MS (ESI) m/z: 1058.8 [M+H] + . 1 HNMR (400 MHz, DMSO-d 6 ) δ = 13.22 - 12.27 (m, 2H), 10.90 (s, 1H), 8.13 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 7.6 Hz, 1H), 7.62 (d, J = 8.0 Hz, 1H), 7.49 - 7.42 (m, 4H), 7.40 - 7.32 (m, 2H), 7.06 - 7.02 (m, 2H), 6.94 (dd, J = 8.8, 10.8 Hz, 2H), 6.79 (d, J = 2.0 Hz, 1H), 6.72 (dd, J = 2.4, 8.4 Hz, 1H), 4.97 (s, 2H), 4.35 (dd, J = 5.2, 9.6 Hz, 1H), 4.27 (s, 3H), 4.03 - 3.99 (m, 2H), 3.91 (t, J = 6.0 Hz, 2H), 3.78 - 3.57 (m, 2H), 3.29 - 3.15 (m, 8H), 3.02 (t, J = 5.2 Hz, 4H), 2.71 - 2.60 (m, 4H), 2.37 - 2.30 (m, 2H), 2.19 – 2.13 (m, 2H), 2.09 - 1.84 (m, 7H), 1.02 (s, 6H). Example 152. Preparation of Compound 185a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[(3R,5S)-4- [2-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]piper azin-1-yl]-2-oxo-ethyl]-3,5- dimethyl-piperazin-1-yl]-2,2-difluoro-propoxy]-2-methyl-phen yl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(3R,5S)-4-[2-[4-[3 -(2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]piperazin-1-yl]-2-oxo-ethyl]-3,5-dimethy l-piperazin-1-yl]-2,2- difluoro-propoxy]-2-methyl-phenyl]pyridine-2-carboxylate To a solution of 2-[(2R, 6S)-4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihyd ro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]-2,2-difluoro-propyl]- 2,6-dimethyl-piperazin-1-yl]acetic acid (100 mg, 118.91 µmol, 1.0 equiv.) and 3-(1-methyl-6- piperazin-1-yl-indazol-3-yl)piperidine-2,6-dione (58.39 mg, 178.36 µmol, 1.5 equiv.) in DMF (3 mL) was added HATU (54.26 mg, 142.69 µmol, 1.2 equiv.) and DIEA (46.10 mg, 356.73 µmol, 62.14 µL, 3.0 equiv.).The mixture was stirred at 25 °C for 2 hours. The reaction mixture was added into H 2 O (5 mL). The mixture was triturated with water (5 mL) at 25 °C for 30 minutes and filtered to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[3-[(3R,5S)-4-[2-[4-[3-(2,6-dioxo-3-p iperidyl)-1-methyl-indazol-6- yl]piperazin-1-yl]-2-oxo-ethyl]-3,5-dimethyl-piperazin-1-yl] -2,2-difluoro-propoxy]-2- methyl-phenyl]pyridine-2-carboxylate (135 mg, 117.4 µmol, 98.7% yield) as a yellow solid. MS (ESI) m/z: 1150.6 [M+H] + . Step B. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[(3R,5S)-4-[2-[4-[3-(2,6-dioxo- 3-piperidyl)-1-methyl- indazol-6-yl]piperazin-1-yl]-2-oxo-ethyl]-3,5-dimethyl-piper azin-1-yl]-2,2-difluoro- propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[(3R,5S)-4-[2-[4-[3-(2,6-dioxo-3-piperidyl)-1-me thyl-indazol-6-yl]piperazin-1- yl]-2-oxo-ethyl]-3,5-dimethyl-piperazin-1-yl]-2,2-difluoro-p ropoxy]-2-methyl- phenyl]pyridine-2-carboxylate (135 mg, 117.36 µmol, 1.0 equiv.) in DCM (1 mL) and TFA (1 mL) was stirred at 25 °C for 16 hours. The mixture was concentrated to remove DCM. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(3R,5S)-4-[2-[4-[3-(2, 6-dioxo-3-piperidyl)-1-methyl- indazol-6-yl]piperazin-1-yl]-2-oxo-ethyl]-3,5-dimethyl-piper azin-1-yl]-2,2-difluoro- propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid (68.0 mg, 59.6 µmol, 50.8% yield, 95.8% purity) as a white solid. MS (ESI) m/z: 1094.4 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.90 - 12.43 (m, 2H), 10.85 (s, 1H), 8.13 (s, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.53 (d, J = 8.8 Hz, 1H), 7.50 - 7.43 (m, 3H), 7.41 - 7.302 (m, 2H), 7.18 - 7.13 (m, 1H), 7.04 - 6.85 (m, 4H), 6.71 (d, J = 7.6 Hz, 1H), 4.99 (s, 2H), 4.41 - 4.22 (m, 3H), 3.97 - 3.85 (m, 5H), 3.68 - 3.50 (m, 6H), 3.22 (d, J = 2.4 Hz, 4H), 3.03 ( t, J = 5.6 Hz, 2H), 2.93 - 2.80 (m, 2H), 2.68 - 2.50 (m, 6H), 2.37 - 2.10 (m, 3H), 2.09 - 1.98 (m, 1H), 1.92 (s, 3H), 1.17 - 0.78 (m, 6H) 19 F NMR (400 MHz, DMSO-d6) δ = 106.40. Example 153. Preparation of Compound 186a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[(3R)-1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-o xo-ethyl]-3-piperidyl]butoxy]- 2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl (3R)-3-[4-(3-bromo-2-methyl- phenoxy)butyl]piperidine-1-carboxylate A mixture of tert-butyl (3R)-3-(4-hydroxybutyl)piperidine-1-carboxylate (1.3 g, 5.05 mmol, 1 equiv.), 3-bromo-2-methyl-phenol (755.79 mg, 4.04 mmol, 0.8 equiv.), and 2-(tributyl-λ5- phosphanylidene)acetonitrile (1.46 g, 6.06 mmol, 1.2 equiv.) in toluene (30 mL) was stirred at 120 °C for 2 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 10% ethyl acetate/petroleum ether). Then the residue was further separated by SFC to give tert-butyl (3R)-3-[4-(3-bromo-2-methyl-phenoxy)butyl]piperidine-1- carboxylate (1.1 g, 2.5 mmol, 50.7% yield, 99.3% purity) as a yellow oil. MS (ESI) m/z: 328.0 [M-100+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 7.17 - 7.13 (m, 1H), 7.12 - 7.06 (m, 1H), 6.97 (d, J = 8.0 Hz, 1H), 3.98 (t, J = 6.4 Hz, 2H), 3.85 - 3.68 (m, 2H), 2.80 - 2.73 (m, 1H), 2.49 - 2.26 (m, 1H), 2.23 (s, 3H), 1.78 - 1.69 (m, 3H), 1.60 - 1.53 (m, 1H), 1.49 - 1.43 (m, 2H), 1.38 (s, 9H), 1.36 - 1.27 (m, 2H), 1.27 - 1.20 (m, 2H), 1.14 - 1.01 (m, 1H). Step B. Procedure for preparation of (3R)-3-[4-(3-bromo-2-methyl- phenoxy)butyl]piperidine A mixture of tert-butyl (3R)-3-[4-(3-bromo-2-methyl-phenoxy)butyl]piperidine-1-carbo xylate (400 mg, 938.12 µmol, 1 equiv.) in DCM (2.5 mL) and HCl/dioxane (12 mL) was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give (3R)- 3-[4-(3-bromo-2-methyl-phenoxy)butyl]piperidine (349 mg, crude) as a white solid. MS (ESI) m/z: 328.1 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.17 - 7.13 (m, 1H), 7.12 - 7.06 (m, 1H), 6.96 (d, J = 8.0 Hz, 1H), 3.98 (t, J = 6.4 Hz, 2H), 3.18 (d, J = 12.0 Hz, 2H), 2.78 - 2.67 (m, 1H), 2.48 - 2.45 (m, 1H), 2.23 (s, 3H), 1.83 - 1.52 (m, 7H), 1.48 - 1.39 (m, 2H), 1.32 - 1.22 (m, 2H), 1.16 - 1.04 (m, 1H). Step C. Procedure for preparation of ethyl 2-[(3R)-3-[4-(3-bromo-2-methyl- phenoxy)butyl]-1-piperidyl]acetate A mixture of (3R)-3-[4-(3-bromo-2-methyl-phenoxy)butyl]piperidine (349 mg, 1.07 mmol, 1 equiv.), ethyl 2-bromoacetate (142.91 mg, 855.73 µmol, 94.64 µL, 0.8 equiv.), and K2CO3 (443.50 mg, 3.21 mmol, 3 equiv.) in DMF (10 mL) was stirred at 60 °C for 2 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product was purified by reverse-phase HPLC (with 0.1% HCOOH) to give ethyl 2- [(3R)-3-[4-(3-bromo-2-methyl-phenoxy)butyl]-1-piperidyl]acet ate (374 mg, 896.9 µmol, 83.8% yield, 98.9% purity) as a yellow oil. MS (ESI) m/z: 414.0 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 8.14 (s, 1H), 7.16 - 7.12 (m, 1H), 7.11 - 7.05 (m, 1H), 6.95 (d, J = 7.6 Hz, 1H), 4.12 (q, J = 7.2 Hz, 2H), 3.97 (t, J = 6.4 Hz, 2H), 3.51 (s, 2H), 3.05 - 2.92 (m, 2H), 2.44 - 2.33 (m, 1H), 2.22 (s, 3H), 2.13 (t, J = 10.8 Hz, 1H), 1.73 - 1.69 (m, 2H), 1.66 - 1.58 (m, 2H), 1.43 (td, J = 7.6, 14.8 Hz, 2H), 1.33 - 1.09 (m, 6H), 0.84 - 0.95 (m, 1H). Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[(3R)-1-(2-ethoxy-2 -oxo-ethyl)-3- piperidyl]butoxy]-2-methyl-phenyl]pyridine-2-carboxylate Ethyl 2-[(3R)-3-[4-(3-bromo-2-methyl-phenoxy)butyl]-1-piperidyl]ac etate (421 mg, 1.02 mmol, 1.05 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (596.69 mg, 974.12 µmol, 1 equiv.), Ad 2 nBuP Pd G 3 (cataCXium® A Pd G 3 ) (70.94 mg, 97.41 µmol, 0.1 equiv.), and KF (1.5 M, 1.95 mL, 3 equiv.) were taken up into a microwave tube in 1,4-dioxane (10 mL). The sealed tube was heated at 100 °C for 60 minutes under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[(3R)- 1-(2-ethoxy-2-oxo-ethyl)-3- piperidyl]butoxy]-2-methyl-phenyl]pyridine-2-carboxylate (511 mg, 593.5 µmol, 60.9% yield, 95.0% purity) as a yellow oil. MS (ESI) m/z: 818.4 [M+H] + . 1 H NMR (400 MHz, CDCl3) δ = 7.80 (d, J = 7.6 Hz, 1H), 7.59 (dd, J = 7.6, 15.2 Hz, 2H), 7.41 - 7.35 (m, 1H), 7.34 - 7.29 (m, 3H), 7.27 (d, J = 7.6 Hz, 1H), 7.03 (t, J = 8.0 Hz, 1H), 6.82 (d, J = 8.8 Hz, 1H), 6.73 (d, J = 8.0 Hz, 1H), 6.64 (d, J = 7.6 Hz, 1H), 5.24 (s, 1H), 5.05 - 4.91 (m, 2H), 4.18 (q, J = 7.2 Hz, 2H), 4.03 (t, J = 5.6 Hz, 2H), 3.90 (t, J = 6.0 Hz, 3H), 3.81 (s, 2H), 3.01 (t, J = 5.6 Hz, 3H), 2.04 - 1.95 (m, 2H), 1.91 (s, 5H), 1.76 - 1.71 (m, 2H), 1.54 - 1.38 (m, 3H), 1.27 - 1.22 (m, 4H), 1.20 (d, J = 6.0 Hz, 3H), 1.08 (s, 9H). Step E. Procedure for preparation of 2-[(3R)-3-[4-[3-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-2- methyl-phenoxy]butyl]-1-piperidyl]acetic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[4-[(3R)-1-(2-ethoxy-2-oxo-ethyl)-3-piperidyl]butox y]-2-methyl-phenyl]pyridine-2- carboxylate (112 mg, 136.91 µmol, 1 equiv.) and LiOH•H 2 O (17.24 mg, 410.74 µmol, 3 equiv.) in THF (2 mL) and H 2 O (1 mL) was stirred at 25 °C for 16 hours. The reaction mixture was quenched by water (5 mL), and then extracted with ethyl acetate (10 mL × 3). The organic layers was combined and concentrated under reduced pressure to give 2-[(3R)-3-[4-[3-[6-[8- (1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin- 2-yl]-2-tert-butoxycarbonyl-3- pyridyl]-2-methyl-phenoxy]butyl]-1-piperidyl]acetic acid (105 mg, crude) as a yellow oil. MS (ESI) m/z: 790.4 [M+H] + . Step F. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[(3R)-1-[2-[[3-(2,6 -dioxo-3-piperidyl)-1-methyl- indazol-7-yl]amino]-2-oxo-ethyl]-3-piperidyl]butoxy]-2-methy l-phenyl]pyridine-2- carboxylate A mixture of 2-[(3R)-3-[4-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]butyl]-1- piperidyl]acetic acid (95 mg, 120.26 µmol, 1 equiv.), 3-(7-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (37.27 mg, 144.31 µmol, 1.2 equiv.), HATU (54.87 mg, 144.31 µmol, 1.2 equiv.), and DIEA (46.63 mg, 360.78 µmol, 62.84 µL, 3 equiv.) in DMF (4 mL) was stirred at 25 °C for 12 hours. The reaction mixture was quenched by water (5 mL) and then extracted with ethyl acetate (10 mL × 3). The organic layers was combined and concentrated under reduced pressure to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[4-[(3R)-1-[2-[[3-(2,6-dioxo-3-piperi dyl)-1-methyl-indazol-7- yl]amino]-2-oxo-ethyl]-3-piperidyl]butoxy]-2-methyl-phenyl]p yridine-2-carboxylate (100 mg, crude) as a purple oil. MS (ESI) m/z: 1030.7 [M+H] + . Step G. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[4-[(3R)-1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-7- yl]amino]-2-oxo-ethyl]-3-piperidyl]butoxy]-2-methyl-phenyl]p yridine-2-carboxylic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[4-[(3R)-1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl- indazol-7-yl]amino]-2-oxo- ethyl]-3-piperidyl]butoxy]-2-methyl-phenyl]pyridine-2-carbox ylate (100 mg, 97.06 µmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL) was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep- HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3- [4-[(3R)-1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7 -yl]amino]-2-oxo-ethyl]-3- piperidyl]butoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid (27.9 mg, 27.4 µmol, 28.2% yield, 95.8% purity) as a yellow solid. MS (ESI) m/z: 974.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ,) δ = 10.89 (s, 1H), 9.83 (s, 1H), 8.13 (s, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.61 (d, J = 7.2 Hz, 1H), 7.55 (d, J = 8.0 Hz, 1H), 7.50 - 7.33 (m, 5H), 7.29 (d, J = 7.2 Hz, 1H), 7.00 - 7.10 (m, 2H), 6.95 (d, J = 8.8 Hz, 1H), 6.86 (d, J = 8.0 Hz, 1H), 6.61 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.35 (dd, J = 5.2, 10.0 Hz, 1H), 4.10 (s, 3H), 3.80 - 4.00 (m, 4H), 3.16 (s, 2H), 3.02 (t, J = 5.6 Hz, 2H), 2.95 - 2.84 (m, 2H), 2.67 - 2.57 (m, 2H), 2.38 - 2.28 (m, 1H), 2.21 - 2.07 (m, 2H), 1.87 (s, 4H), 1.77 - 1.40 (m, 9H), 1.32 - 1.20 (m, 2H). Example 154. Preparation of Compound 188 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[[7-[2-[4-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]piperazin-1-yl ]ethyl]-7-azaspiro[3.5]nonan-2- yl]methoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 2-((3-bromo-2-methylphenoxy)methyl)-7- azaspiro[3.5]nonane-7-carboxylate A mixture of tert-butyl 2-(hydroxymethyl)-7-azaspiro[3.5]nonane-7-carboxylate (1 g, 3.92 mmol, 1.2 equiv.), 3-bromo-2-methylphenol (610.38 mg, 3.26 mmol, 1 equiv.), and 2-(tributyl- λ 5 -phosphanylidene)acetonitrile (1.18 g, 4.90 mmol, 1.5 equiv.) in toluene (10 mL) was stirred at 120 °C for 2 h under N2 atmosphere. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 8% ethyl acetate / petroleum ether) to give tert-butyl 2-((3- bromo-2-methylphenoxy)methyl)-7-azaspiro[3.5]nonane-7-carbox ylate (1.3 g, 3.1 mmol, 96.7% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.16 (d, J = 8.0 Hz,1H), 7.09 (t, J = 8.0 Hz, 1H), 6.96 (d, J = 8.0 Hz, 1H), 3.95 ( d, J = 6.2 Hz, 2H), 3.29 - 3.25 (m, 2H), 3.22 - 3.15 (m, 2H), 2.77 - 2.64 (m, 1H), 2.24 (s, 3H), 1.94 - 1.86 (m, 2H), 1.69 - 1.62 (m, 2H), 1.56 - 1.50 (m, 2H), 1.44 - 1.40 (m, 2H), 1.38 (s, 9H) Step B. Procedure for preparation of 2-((3-bromo-2-methylphenoxy)methyl)-7- azaspiro[3.5]nonane A mixture of tert-butyl 2-((3-bromo-2-methylphenoxy)methyl)-7-azaspiro[3.5]nonane-7- carboxylate (1.34 g, 3.16 mmol, 1 equiv.) in HCl / dioxane (15 mL) was stirred at 25 °C for 1 h, The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was used in the next step without other purification. The compound 2- ((3-bromo-2-methylphenoxy)methyl)-7-azaspiro[3.5]nonane (1.32 g, crude, HCl) was obtained as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ = 9.0 - 8.75(m, 2H), 7.15 (d, J = 7.2 Hz, 1H), 7.09 (t, J = 8.0 Hz, 1H), 6.96 (d, J = 8.0 Hz, 1H), 3.95 (d, J = 6.2 Hz, 2H), 3.02 - 2.93(m, 2H), 2.93 - 2.84 (m, 2H), 2.74 - 2.63 (m, 1H), 2.23 (s, 3H), 2.00 - 1.92 (m, 2H), 1.82 - 1.77 (m, 2H), 1.74 - 1.66 (m, 4H) Step C. Procedure for preparation of 2-((3-bromo-2-methylphenoxy)methyl)-7-(2,2- diethoxyethyl)-7-azaspiro[3.5]nonane A mixture of 2-((3-bromo-2-methylphenoxy)methyl)-7-azaspiro[3.5]nonane (1.22 g, 3.38 mmol, 1 equiv., HCl), 2-bromo-1,1-diethoxy-ethane (666.52 mg, 3.38 mmol, 508.80 µL, 1 equiv.), K2CO3 (2.34 g, 16.91 mmol, 5 equiv.), and KI (280.72 mg, 1.69 mmol, 0.5 equiv.) in CH3CN (10 mL) was stirred at 80 °C for 16 hours. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The mixture was purified by flash silica gel chromatography (Eluent of 0 ~ 100% ethyl acetate / petroleum ether) to give 2-((3- bromo-2-methylphenoxy)methyl)-7-(2,2-diethoxyethyl)-7-azaspi ro[3.5]nonane (1.3 g, 2.9 mmol, 87.2% yield) as a yellow oil. MS (ESI) m/z: 442.4[M+H] + . Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-((7-(2-ethoxy-2-methoxyeth yl)-7-azaspiro[3.5]nonan- 2-yl)methoxy)-2-methylphenyl)picolinate A mixture of 2-((3-bromo-2-methylphenoxy)methyl)-7-(2,2-diethoxyethyl)-7- azaspiro[3.5]nonane (400 mg, 908.24 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tet ramethyl-1,3,2-dioxaborolan-2- yl)pyridine-2-carboxylate (723.24 mg, 1.18 mmol, 1.3 equiv.), [2-(2- aminophenyl)phenyl]palladium(1 + );bis(1-adamantyl)-butyl-phosphane;methanesulfonate (132.29 mg, 181.65 µmol, 0.2 equiv.), and K 2 CO 3 (1.5 M, 1.98 mL, 3 equiv.) in dioxane (4 mL) was degassed and purged with N2 and stirred at 100 °C for 1 h under N2 atmosphere. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The mixture was purified by flash silica gel chromatography (Eluent of 50 ~ 60% ethyl acetate / petroleum ether) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-((7-(2-ethoxy-2-methoxyeth yl)-7-azaspiro[3.5]nonan-2- yl)methoxy)-2-methylphenyl)picolinate (595 mg, 703.2 µmol, 77.4% yield) as a yellow oil. MS (ESI) m/z: 846.7[M+H] + . Step E. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[2-methyl-3-[[7-(2-oxoethyl)-7-azaspi ro[3.5]nonan-2- yl]methoxy]phenyl]pyridine-2-carboxylic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-((7-(2-ethoxy-2-methoxyethyl)-7-azaspiro[3.5]nonan- 2-yl)methoxy)-2- methylphenyl)picolinate (245 mg, 289.57 µmol, 1 equiv.), in HCOOH (3 mL) was stirred at 90 °C for 1.5 hours. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was used in the next step without other purification. The compound 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-((7-(2-oxoethyl)-7-azaspiro[3.5]nonan-2-yl)methoxy) phenyl)picolinic acid (200 mg, crude) was obtained as a yellow oil. MS (ESI) m/z: 716.6[M+H] + . Step F. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[[7-[2-[4-[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-7- yl]piperazin-1-yl]ethyl]-7-azaspiro[3.5]nonan-2-yl]methoxy]- 2-methyl-phenyl]pyridine- 2-carboxylic acid A mixture of 3-(1-methyl-7-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (27.44 mg, 83.82 µmol, 1.2 equiv.), NaBH(OAc)3 (44.41 mg, 209.54 µmol, 3.0 equiv.), and NMM (7.06 mg, 69.85 µmol, 7.68 µL, 1.0 equiv.) in DCM (1 mL) and isopropanol (1 mL) was stirred at 0 °C for 5 min. To the mixture was then added 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[2-methyl-3-[[7-(2-oxoethyl)-7-azaspi ro[3.5]nonan-2- yl]methoxy]phenyl]pyridine-2-carboxylic acid (50 mg, 69.85 µmol, 1.0 equiv.). The mixture was stirred at 0 °C for 30 min. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep–HPLC to give 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[[7-[2-[4 -[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-7-yl]piperazin-1-yl]ethyl]-7-azaspiro[3.5]non an-2-yl]methoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid (22.3 mg, 21.6 µmol, 15.5% yield, 99.5% purity) as a white solid. MS (ESI) m/z: 1027.4[M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.87 (s, 1H), 8.05 - 7.97 (m, 1H), 7.78 (d, J = 8.4 Hz, 1H), 7.65 - 7.59 (m, 1H), 7.48 - 7.31 (m, 6H), 7.10 - 6.98 (m, 3H), 6.94 - 6.83 (m, 2H), 6.64 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.33 (dd, J = 4.8, 9.2 Hz, 1H), 4.23 (s, 3H), 3.94 - 3.87 (m, 4H), 3.58 - 3.50 (m, 4H), 3.04 - 2.97 (m, 4H), 2.93 - 2.77 (m, 4H), 2.70 - 2.59 (m, 4H), 2.41 - 2.26 (m, 6H), 2.19 - 2.14 (m, 1H), 1.93 - 1.84 (m, 5H), 1.68 - 1.56 (m, 4H), 1.52 - 1.45 (m, 2H) Example 155. Preparation of Compound 194 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[2-[[1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-oxo-e thyl]-4-piperidyl]oxy]ethoxy]- 2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 4-(2-(3-bromo-2- methylphenoxy)ethoxy)piperidine-1-carboxylate A mixture of 3-bromo-2-methylphenol (655.68 mg, 3.51 mmol, 1 equiv.), tert-butyl 4-(2- hydroxyethoxy)piperidine-1-carboxylate (860 mg, 3.51 mmol, 1 equiv.) and 2-(tributyl-λ5- phosphanylidene)acetonitrile (1.02 g, 4.21 mmol, 1.2 equiv.) in toluene (20 mL) was degassed, purged with N 2 , and stirred at 120 °C for 12 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~23% ethyl acetate/petroleum ether) to give tert-butyl 4-(2-(3-bromo-2-methylphenoxy)ethoxy)piperidine-1-carboxylat e (800 mg, 1.7 mmol, 49.5% yield, 90% purity) as a yellow oil. 1 H NMR (400 MHz, CDCl3) δ = 7.17 (d, J = 8.0 Hz, 1H), 7.00 (t, J = 8.0 Hz, 1H), 6.79 (d, J = 8.4 Hz, 1H), 4.12 - 4.09 (m, 2H), 3.85 - 3.83 (m, 2H), 3.79 - 3.71 (m, 2H), 3.60 - 3.51 (m, 1H), 3.16 - 3.09 (m, 2H), 2.33 (s, 3H), 1.86 - 1.83 (m, 2H), 1.60 - 1.54 (m, 2H), 1.47 (s, 9H). Step B. Procedure for preparation of 4-(2-(3-bromo-2-methylphenoxy)ethoxy)piperidine To a solution of tert-butyl 4-(2-(3-bromo-2-methylphenoxy)ethoxy)piperidine-1-carboxylat e (800 mg, 1.93 mmol, 1 equiv.) was added HCl/dioxane (4 M, 965.41 µL, 2 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give 4-(2-(3-bromo-2-methylphenoxy)ethoxy)piperidine (800 mg, crude) as a white solid. Step C. Procedure for preparation of ethyl 2-(4-(2-(3-bromo-2- methylphenoxy)ethoxy)piperidin-1-yl)acetate To a solution of 4-(2-(3-bromo-2-methylphenoxy)ethoxy)piperidine (400 mg, 1.14 mmol, 1 eq, HCl) in CH3CN (4 mL) was added K2CO3 (472.95 mg, 3.42 mmol, 3 equiv.) and ethyl 2- bromoacetate (190.49 mg, 1.14 mmol, 126.15 µL, 1 equiv.). The mixture was stirred at 60 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~70% ethyl acetate/petroleum ether) to give ethyl 2-(4-(2-(3-bromo-2-methylphenoxy)ethoxy)piperidin-1- yl)acetate (420 mg, 944.28 µmol, 82.7% yield, 90% purity) as a yellow oil. MS (ESI) m/z: 402.0 [M+H] + . 1 H NMR (400 MHz, CDCl3) δ = 7.17 (d, J = 8.0 Hz, 1H), 7.01 (t, J = 8.2 Hz, 1H), 6.80 (d, J = 8.0 Hz, 1H), 4.22 (q, J = 6.8 Hz, 2H), 4.12 (t, J = 4.4 Hz, 2H), 3.87 - 3.80 (m, 2H), 3.62 (s, 1H), 3.48 (t, J = 6.0 Hz, 2H), 3.05 (s, 2H), 2.93 - 2.59 (m, 2H), 2.32 (s, 3H), 2.14 - 2.02 (m, 2H), 1.87 (s, 2H), 1.29 (t, J = 6.8 Hz, 3H). Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((1-(2-ethoxy-2-oxoethy l)piperidin-4- yl)oxy)ethoxy)-2-methylphenyl)picolinate A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)picolinat e (679.40 mg, 998.24 µmol, 90% purity, 1.2 equiv.) and ethyl 2-(4-(2-(3-bromo-2-methylphenoxy)ethoxy)piperidin-1-yl)aceta te (370 mg, 831.86 µmol, 90% purity, 1 equiv.) in dioxane (6 mL) was added KF (1.5 M, 1.66 mL, 3 equiv.) and Ad 2 nBuP Pd G 3 (cataCXium® A Pd G 3 ) (60.58 mg, 83.19 µmol, 0.1 equiv.). After addition, the mixture was degassed and purged with N 2 and stirred at 100 °C for 2 hours under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~65 % dichloromethane/methanol) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((1-(2-ethoxy-2-oxoethy l)piperidin-4-yl)oxy)ethoxy)-2- methylphenyl)picolinate (600 mg, 669.9 µmol, 80.5% yield, 90% purity) as a yellow solid. MS (ESI) m/z: 806.5 [(M-56)/2+375.6] + . 1 H NMR (400 MHz, CDCl3) δ = 10.47 - 9.88 (m, 1H), 7.86 (d, J = 8.0 Hz, 1H), 7.62 - 7.55 (m, 2H), 7.38 - 7.28 (m, 5H), 7.10 (t, J = 8.0 Hz, 1H), 6.90 (d, J = 8.8 Hz, 1H), 6.84 (d, J = 8.0 Hz, 1H), 6.72 (d, J = 7.2 Hz, 1H), 5.15 - 4.90 (m, 2H), 4.18 (q, J = 7.2 Hz, 2H), 4.12 - 4.09 (m, 4H), 3.84 (t, J = 5.2 Hz, 2H), 3.51 - 3.46 (m, 1H), 3.20 (s, 2H), 3.07 (t, J = 5.6 Hz, 2H), 2.81 (s, 2H), 2.40 - 2.29 (m, 2H), 2.00 (s, 3H), 1.96 - 1.90 (m, 2H), 1.77 - 1.70 (m, 2H), 1.27 (t, J = 7.2 Hz, 3H), 1.15 (s, 9H). Step E. Procedure for preparation of 2-[4-[2-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyr idyl]-2-methyl- phenoxy]ethoxy]-1-piperidyl]acetic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-((1-(2-ethoxy-2-oxoethyl)piperidin-4-yl)ox y)ethoxy)-2- methylphenyl)picolinate (600 mg, 699.77 µmol, 94% purity, 1 equiv.) in THF (6 mL) was added LiOH•H 2 O (88.09 mg, 2.10 mmol, 3 equiv.) and H 2 O (1.5 mL) at 25 °C. The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove THF. The aqueous phase was adjusted to pH=4~5 with 1M HCl. The reaction mixture was filtered. After filtering, the filter cake was diluted in ethyl acetate. The combined organic layers were dried over Na2SO4, filtered, and concentrated under reduced pressure to give 2-[4-[2-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]ethoxy]-1- piperidyl]acetic acid (800 mg, crude) as a yellow solid. MS (ESI) m/z: 778.4 [M+H] + Step F. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[[1-[2-[[3-(2,6-dio xo-3-piperidyl)-1-methyl- indazol-7-yl]amino]-2-oxo-ethyl]-4-piperidyl]oxy]ethoxy]-2-m ethyl-phenyl]pyridine-2- carboxylate To a solution of 2-[4-[2-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]ethoxy]-1- piperidyl]acetic acid (100 mg, 128.55 µmol, 1 equiv.) and 3-(7-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (33.20 mg, 128.55 µmol, 1 equiv.) in pyridine (2 mL) was added EDCI (36.96 mg, 192.82 µmol, 1.5 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[[1-[2 -[[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-7-yl]amino]-2-oxo-ethyl]-4-piperidyl]oxy]etho xy]-2-methyl-phenyl]pyridine- 2-carboxylate (100 mg, 98.2 µmol, 76.4% yield) as a yellow solid. MS (ESI) m/z: 1018.7[M+H] + Step G. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[[1-[2-[[3-(2,6-dioxo-3 -piperidyl)-1-methyl-indazol- 7-yl]amino]-2-oxo-ethyl]-4-piperidyl]oxy]ethoxy]-2-methyl-ph enyl]pyridine-2- carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[2-[[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-in dazol-7-yl]amino]-2-oxo-ethyl]- 4-piperidyl]oxy]ethoxy]-2-methyl-phenyl]pyridine-2-carboxyla te (100 mg, 98.21 µmol, 1 equiv.) in DCM (0.5 mL) was added TFA (641.67 mg, 5.63 mmol, 416.67 µL, 57.30 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[2-[[1-[2-[[3-(2,6-dioxo- 3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-oxo-ethyl]-4-pip eridyl]oxy]ethoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid (41.4 mg, 43.0 µmol, 43.8% yield, 97.6% purity) as a white solid. MS (ESI) m/z: 962.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.03 - 12.38 (m, 2H), 10.89 (s, 1H), 9.87 (s, 1H), 8.14 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.56 (d, J = 8.0 Hz, 1H), 7.50 - 7.42 (m, 3H), 7.38 - 7.32 (m, 2H), 7.29 (d, J = 7.2 Hz, 1H), 7.12 - 7.04 (m, 2H), 6.97 (d, J = 8.8 Hz, 1H), 6.91 (d, J = 8.0 Hz, 1H), 6.65 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.37 (dd, J = 5.2, 10.4 Hz, 1H), 4.12 - 4.05 (m, 5H), 3.92 (t, J = 6.0 Hz, 2H), 3.81 - 3.75 (m, 2H), 3.48 - 3.43 (m, 2H), 3.17 (s, 2H), 3.02 (t, J = 5.2 Hz, 2H), 2.86 - 2.79 (m, 2H), 2.68 - 2.60 (m, 2H), 2.38 - 2.32 (m, 2H), 2.19 - 2.13 (m, 1H), 1.91 (s, 5H), 1.67 - 1.56 (m, 2H). Example 156. Preparation of Compound 195 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[3-[4-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]piperazin-1-yl ]propyl]phenoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of ethyl 3-(3-(3-bromo-2- methylphenoxy)phenyl)propanoate A mixture of ethyl 3-(3-hydroxyphenyl)propanoate (1 g, 5.15 mmol, 1 equiv.), 1,3-dibromo-2- methylbenzene (2.57 g, 10.30 mmol, 2 equiv.), 2,2,6,6-tetramethylheptane-3,5-dione (237.19 mg, 1.29 mmol, 265.02 µL, 0.25 equiv.), and Cs 2 CO 3 (2.52 g, 7.72 mmol, 1.5 equiv.) in NMP (10 mL) was added CuI (490.28 mg, 2.57 mmol, 0.5 equiv.). The reaction mixture was purged with N2 and stirred at 120 °C for 15 hours under N2 atmosphere. The reaction mixture was diluted with H 2 O (10 mL) and extracted with ethyl acetate (10 mL × 3). The combined organic layers were washed with sat. NaHCO3 (10 mL × 2), filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~12% ethyl acetate/petroleum ether) to give ethyl 3-(3-(3-bromo-2- methylphenoxy)phenyl)propanoate (650 mg, 1.7 mmol, 34.7% yield) as a yellow oil 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.42 (d, J = 8.0 Hz, 1H), 7.29 - 7.21 (m, 1H), 7.15 (t, J = 8.0 Hz, 1H), 6.97 (d, J = 7.6 Hz, 1H), 6.88 (d, J = 8.0 Hz, 1H), 6.82 - 6.77 (m, 1H), 6.75 - 6.69 (m, 1H), 4.00 (q, J = 7.2 Hz, 2H), 2.85 - 2.78 (m, 2H), 2.58 (t, J = 7.6 Hz, 2H), 2.24 (s, 3H), 1.12 (t, J = 7.2 Hz, 3H) Step B. Procedure for preparation of 3-(3-(3-bromo-2-methylphenoxy)phenyl)propan-1- ol A mixture of ethyl 3-(3-(3-bromo-2-methylphenoxy)phenyl)propanoate (650 mg, 1.79 mmol, 1 equiv.) in THF (7 mL) was slowly added LiAlH4 (54.33 mg, 1.43 mmol, 0.8 equiv.) at 0 °C, and then the mixture was stirred at 25 ℃ for 4 hours. The mixture was quenched by Na 2 SO 4 (2g). The solution was poured into ice-water (15 mL) slowly, and the pH was adjusted to 4-5 with 1 M HCl. The resulting mixture was extracted with EtOAc (15 mL × 3), washed with brine (10 mL × 2), dried by sodium sulfate, filtered, and concentrated under reduced pressure to give 3-(3-(3-bromo-2-methylphenoxy)phenyl)propan-1-ol (556 mg, 1.5 mmol, 84.2% yield, 87.1% purity) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.42 (d, J = 8.0 Hz, 1H), 7.29 - 7.22 (m, 1H), 7.16 (t, J = 8.0 Hz, 1H), 6.96 (d, J = 7.6 Hz, 1H), 6.92 - 6.86 (m, 1H), 6.78 (s, 1H), 6.73 - 6.67 (m, 1H), 4.45 (t, J = 5.2 Hz, 1H), 3.42 - 3.36 (m, 2H), 2.61 - 2.55 (m, 2H), 2.26 (s, 3H), 1.72 - 1.63 (m, 2H) Step C. Procedure for preparation of 3-(3-(3-bromo-2-methylphenoxy)phenyl)propanal A mixture of (COCl)2 (439.41 mg, 3.46 mmol, 303.04 µL, 2 equiv.) in DCM (8 mL) at -78°C was charged with DMSO (540.98 mg, 6.92 mmol, 540.98 µL, 4 equiv.) and stirred for 0.5 hours. The solution was charged with 3-(3-(3-bromo-2-methylphenoxy)phenyl)propan-1-ol (556 mg, 1.73 mmol, 1 equiv.) in DCM (4 mL) and stirred at-70 °C for 1 hour. Then the mixture was charged with TEA (1.05 g, 10.39 mmol, 1.45 mL, 6 equiv.), warmed to 25 °C, and stirred 0.5 hours under N2 atmosphere. The reaction mixture was diluted with H2O (10 mL) and extracted with DCM (10 mL × 3). The combined organic layers were washed with sat. NaCl (10 mL × 2), filtered, and concentrated under reduced pressure to give 3-(3-(3-bromo-2- methylphenoxy)phenyl)propanal (293 mg, 728.8 µmol, 42.1% yield, 79.4% purity) as a yellow solid 1 H NMR (400 MHz, DMSO-d 6 ) δ = 9.69 (s, 1H), 7.43 (d, J = 8.0 Hz, 1H), 7.30 - 7.22 (m, 1H), 7.16 (t, J = 8.0 Hz, 1H), 7.01 - 6.94 (m, 1H), 6.91 - 6.83 (m, 2H), 6.74 - 6.67 (m, 1H), 2.87 - 2.80 (m, 2H), 2.79 - 2.72 (m, 2H), 2.26 (s, 3H) Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(2-methyl-3-(3-(3-oxopropyl)p henoxy)phenyl)picolinate 3-(3-(3-Bromo-2-methylphenoxy)phenyl)propanal (210 mg, 525.67 µmol, 79.9% purity, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)picolinate (354.20 mg, 578.24 µmol, 1.1 eq), K2CO3 (217.95 mg, 1.58 mmol, 1.13 mL, 3 equiv.), and Ad2nBuP Pd G3 (76.57 mg, 105.13 µmol, 0.2 equiv.) were taken up into a microwave tube in dioxane (2.4 mL) and H 2 O (0.8 mL). The sealed tube was heated at 100 °C for 1 hour under microwave. The reaction mixture was partitioned between H2O (10 mL) and ethyl acetate (8 mL × 3). The organic phase was separated, washed with aqueous NaCl (5 mL × 3), dried over (Na 2 SO 4 ), filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~26% ethyl acetate/petroleum ether) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(2-methyl-3-(3-(3- oxopropyl)phenoxy)phenyl)picolinate (120 mg, 132.7 µmol, 25.2% yield, 80.2% purity) as a yellow solid MS (ESI) m/z: 725.3 [M+H] + . Step E. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[3-[4-[3-(2,6-dioxo -3-piperidyl)-1-methyl- indazol-6-yl]piperazin-1-yl]propyl]phenoxy]-2-methyl-phenyl] pyridine-2-carboxylate To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2-methyl-3-(3-(3-oxopropyl)phenoxy)phenyl)picol inate (70 mg, 96.57 µmol, 1 equiv.) and 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (31.62 mg, 96.57 µmol, 1 equiv.) in DCM (3 mL) was added AcOH (5.80 mg, 96.57 µmol, 5.52 µL, 1 equiv.), and the mixture was stirred at 25 °C for 1 hour. Then NaBH(OAc)3 (61.40 mg, 289.71 µmol, 3 equiv.) was added to the mixture. The resulting mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[3-[3-[4-[3-(2,6-dioxo- 3-piperidyl)-1-methyl-indazol-6-yl]piperazin-1-yl]propyl]phe noxy]-2-methyl- phenyl]pyridine-2-carboxylate (80 mg, crude) as a green oil. MS (ESI) m/z: 519.1 [M/2+H] + . Step F. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[3-[4-[3-(2,6-dioxo-3-piperidyl )-1-methyl-indazol-6- yl]piperazin-1-yl]propyl]phenoxy]-2-methyl-phenyl]pyridine-2 -carboxylic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[3-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazo l-6-yl]piperazin-1- yl]propyl]phenoxy]-2-methyl-phenyl]pyridine-2-carboxylate (80 mg, 77.20 µmol, 1 equiv.) in DCM (1 mL) and TFA (1 mL) was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to remove solvent to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[3-[3-[4-[3-(2,6-dioxo-3-piperidyl)-1 -methyl-indazol-6-yl]piperazin-1- yl]propyl]phenoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid (20.4 mg, 20.5 µmol, 26.5% yield, 98.1% purity) as a white solid. MS (ESI) m/z: 980.9 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.16 - 12.60 (m, 1H), 10.85 (s, 1H), 8.13 (s, 1H), 8.01 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.61 ( d, J = 7.2 Hz, 1H), 7.54 - 7.43 (m, 3H), 7.41 - 7.30 (m, 3H), 7.27 - 7.17 (m, 2H), 7.02 - 6.81 (m, 7H), 6.74 ( d, J = 8.4 Hz, 1H), 6.68 ( s, 1H), 4.95 (s, 2H), 4.26 ( d, J = 5.2, 8.8 Hz, 1H), 3.88 (s, 6H), 2.91 ( s, 4H), 2.72 ( d, J = 1.6 Hz, 2H), 2.64 - 2.55 (m, 6H), 2.38 - 2.23 (m, 2H), 2.19 - 2.12 (m, 1H), 1.96 - 1.75 (m, 6H). Example 157. Preparation of Compound 201 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[3-[4-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]piperazin-1-yl ]-2,2-difluoro-propyl]phenoxy]- 2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 3-(4-(3-bromo-2-methylphenoxy)phenyl)-2,2- difluoropropan-1-ol To a solution of ethyl 3-(4-(3-bromo-2-methylphenoxy)phenyl)-2,2-difluoropropanoate (1 g, 2.50 mmol, 1 equiv.) in THF (10 mL) was added LiAlH4 (95.07 mg, 2.50 mmol, 1 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was quenched by addition H2O (1.8 mL), 15% NaOH (1.8 mL), and H 2 O 5.4 mL. The resulting mixture was extracted with ethyl acetate (10 mL × 3). The combined organic layers were dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 10% ethyl acetate / petroleum ether) to give 3-(4-(3-bromo- 2-methylphenoxy)phenyl)-2,2-difluoropropan-1-ol (530 mg, 1.1 mmol, 47.3% yield, 80% purity) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.44 (d, J = 8.0 Hz, 1H), 7.27 (d, J = 8.4 Hz, 2H), 7.17 (t, J = 8.1 Hz, 1H), 6.93 - 6.87 (m, 3H), 5.61 - 5.57 (m, 1H), 3.56 - 3.50 (m, 2H), 3.24 - 3.17 (m, 2H), 2.26 (s, 3H) F NMR (400 MHz, DMSO-d6) δ = -105.73. Step B. Procedure for preparation of 3-(4-(3-bromo-2-methylphenoxy)phenyl)-2,2- difluoropropyl trifluoromethanesulfonate To a solution of 3-(4-(3-bromo-2-methylphenoxy)phenyl)-2,2-difluoropropan-1-o l (100 mg, 279.96 µmol, 1 equiv.), Na 2 SO 4 (71.58 mg, 503.93 µmol, 51.13 µL, 1.8 equiv.), and pyridine (33.22 mg, 419.95 µmol, 33.90 µL, 1.5 equiv.) in DCM (5 mL) was added trifluoromethylsulfonyl trifluoromethanesulfonate (118.48 mg, 419.95 µmol, 69.29 µL, 1.5 equiv.). The mixture was stirred at 0 °C for 1 hour. The reaction mixture was diluted with H 2 O (2 mL) and extracted with DCM (10 mL). The combined organic layers were washed with 1N citric acid (2 mL) and 1N NaHCO3 (2 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was used for next step without further purification. The compound 3-(4-(3-bromo-2-methylphenoxy)phenyl)-2,2-difluoropropyl trifluoromethanesulfonate (130 mg, crude) was obtained as a colorless oil. Step C. Procedure for preparation of 3-[7-[4-[3-[4-(3-bromo-2-methyl-phenoxy)phenyl]- 2,2-difluoro-propyl]piperazin-1-yl]-1-methyl-indazol-3-yl]pi peridine-2,6-dione A mixture of [3-[4-(3-bromo-2-methyl-phenoxy)phenyl]-2,2-difluoro-propyl] trifluoromethanesulfonate (120 mg, 245.27 µmol, 1 equiv.), 3-(1-methyl-7-piperazin-1-yl- indazol-3-yl)piperidine-2,6-dione (64.24 mg, 196.22 µmol, 0.8 equiv.), K2CO3 (84.75 mg, 613.18 µmol, 2.5 equiv.), and KI (20.36 mg, 122.64 µmol, 0.5 equiv.) in CH3CN (1.5 mL) was stirred at 50 °C for 10 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 20% ethyl acetate / petroleum ether) to give 3-[7-[4-[3-[4-(3-bromo-2-methyl- phenoxy)phenyl]-2,2-difluoro-propyl]piperazin-1-yl]-1-methyl -indazol-3-yl]piperidine-2,6- dione (40 mg, 60.0 µmol, 24.4% yield) as a colorless oil. MS (ESI) m/z: 668.3 [M+H] + . Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo -3-piperidyl)-1-methyl- indazol-6-yl]piperazin-1-yl]-2,2-difluoro-propyl]phenoxy]-2- methyl-phenyl]pyridine-2- carboxylate A mixture of 3-[7-[4-[3-[4-(3-bromo-2-methyl-phenoxy)phenyl]-2,2-difluoro - propyl]piperazin-1-yl]-1-methyl-indazol-3-yl]piperidine-2,6- dione (40.00 mg, 60.01 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-2-carb oxylate (51.46 mg, 84.01 µmol, 1.4 equiv.), [2-(2-aminophenyl)phenyl]palladium(1 + );bis(1-adamantyl)-butyl- phosphane;methanesulfonate (8.74 mg, 12.00 µmol, 0.2 equiv.), and KF (1.5 M, 60.01 µL, 1.5 equiv.) in dioxane (0.4 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 100 °C for 1 hour under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 100% ethyl acetate / petroleum ether) to give tert-butyl 6-[8-(1,3-benzothiazol- 2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[ 4-[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]piperazin-1-yl]-2,2-difluoro-propyl]phen oxy]-2-methyl-phenyl]pyridine- 2-carboxylate (60 mg, 55.9 µmol, 93.2% yield) as a yellow solid. MS (ESI) m/z: 1072.6 [M+H] + . Step E. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo-3-piperidyl )-1-methyl-indazol-7- yl]piperazin-1-yl]-2,2-difluoro-propyl]phenoxy]-2-methyl-phe nyl]pyridine-2-carboxylic acid tert-Butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4- [3-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]piper azin-1-yl]-2,2-difluoro- propyl]phenoxy]-2-methyl-phenyl]pyridine-2-carboxylate (60 mg, 55.96 µmol, 1 equiv.) in TFA (0.5 mL) and DCM (0.5 mL) was stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[3- [4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]piperazi n-1-yl]-2,2-difluoro- propyl]phenoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid (29.9 mg, 28.2 µmol, 50.4% yield, 95.9% purity) as an off-white solid. MS (ESI) m/z: 1016.9 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.97 - 12.50 (m, 2H), 10.89 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.54 (d, J = 8.8 Hz, 1H), 7.49 - 7.44 (m, 2H), 7.41 - 7.33 (m, 3H), 7.28 (d, J = 8.4 Hz, 2H), 7.20 (t, J = 7.6 Hz, 1H), 7.08 - 6.99 (m, 3H), 6.96 - 6.79 (m, 4H), 4.99 (s, 2H), 4.34 (dd, J = 5.2, 9.6 Hz, 1H), 4.23 (s, 3H), 3.93 (t, J = 5.6 Hz, 2H), 3.29 - 3.24 (m, 2H), 3.15 - 2.85 (m, 8H), 2.80 - 2.60 (m, 6H), 2.35 - 2.12 (m, 2H), 1.89 (s, 3H). F NMR (400 MHz, DMSO-d 6 ) δ = -96.31 Example 158. Preparation of Compound 204a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pip erazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of 1-(((1r,4r)-4-((E)-4-(benzyloxy)but-1-en-1- yl)cyclohexyl)oxy)-3-bromo-2-methylbenzene A mixture of 3-benzyloxypropyl(triphenyl)phosphonium;bromide (5.56 g, 11.31 mmol, 1.2 equiv.) in THF (30 mL) was added LiHMDS (1 M, 12.25 mL, 1.3 equiv.) at -70°C under N 2 atmosphere for 1 hour. Then 4-(3-bromo-2-methyl-phenoxy)cyclohexanecarbaldehyde (2.8 g, 9.42 mmol, 1 equiv.) was added to the mixture at -70°C under N2 atmosphere. The mixture was stirred at 25 °C for 11 hours under N 2 atmosphere. The reaction mixture was quenched by addition NH4Cl (100 mL) at 0 °C, diluted with H2O (80 mL), and extracted with ethyl acetate (80 mL × 3). The combined organic layers were washed with aqueous NaCl (100 mL × 3), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=30/1 to 10/1) to give 1-(((1r,4r)-4-((E)-4-(benzyloxy)but-1-en-1-yl)cyclohexyl)oxy )-3-bromo-2- methylbenzene (3 g, 6.9 mmol, 74.1% yield) as a yellow oil. 1 H NMR (400 MHz, CDCl 3 ) δ = 7.21 - 7.12 (m, 5H), 6.98 (d, J = 8.0 Hz, 1H), 6.81 (t, J = 8.0 Hz, 1H), 6.65 (d, J = 8.0 Hz, 1H), 5.25 - 5.06 (m, 2H), 4.42 - 4.33 (m, 2H), 4.00 - 3.87 (m, 1H), 3.33 (t, J = 6.8 Hz, 2H), 2.29 - 2.21 (m, 2H), 2.14 (s, 3H), 1.97 (d, J = 10.4 Hz, 2H), 1.66 - 1.50 (m, 2H), 1.41 - 1.26 (m, 3H), 1.10 - 0.97 (m, 2H) Step B. Procedure for preparation of 4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butan-1-ol To a solution of 1-(((1r,4r)-4-((E)-4-(benzyloxy)but-1-en-1-yl)cyclohexyl)oxy )-3-bromo-2- methylbenzene (2.6 g, 6.06 mmol, 1 equiv.) in EtOAc (20 mL) was added PtO2 (1.37 g, 6.06 mmol, 1.0 equiv.) under N2 atmosphere. The suspension was degassed and purged with H2. The mixture was stirred under H 2 (15 Psi) at 40 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butan-1-ol (1 g, 2.9 mmol, 48.3% yield) as a yellow oil. 1 H NMR (400 MHz, CDCl3) δ = 7.06 (d, J = 7.6 Hz, 1H), 6.89 (t, J = 8.0 Hz, 1H), 6.76 - 6.69 (m, 1H), 4.06 - 3.97 (m, 1H), 3.58 (t, J = 6.8 Hz, 2H), 2.22 (s, 3H), 2.08 - 2.01 (m, 2H), 1.80 - 1.74 (m, 2H), 1.51 - 1.47 (m, 2H), 1.39 - 1.28 (m, 4H), 1.23 - 1.14 (m, 3H), 1.00 - 0.88 (m, 2H) Step C. Procedure for preparation of 4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butanal To a solution of oxalyl dichloride (371.92 mg, 2.93 mmol, 256.50 uL, 2 equiv.) in DCM (5 mL) was added dropwise to a solution of DMSO (457.86 mg, 5.86 mmol, 457.86 uL, 4 equiv.) in DCM (5 mL) at -70 °C under N2 atmosphere. The mixture was stirred at -70 °C for 30 minutes. After which time, 4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butan-1-ol (500 mg, 1.47 mmol, 1 equiv.) in DCM (5 mL) was added dropwise at -70 °C. The solution was stirred for 30 minutes at -70 °C. Then TEA (889.50 mg, 8.79 mmol, 1.22 mL, 6 equiv.) was added into the solution. The solution was stirred at -70 °C for 2 hour under N 2 atmosphere. The reaction was diluted with water (50 mL) and extracted with DCM (80 mL × 2). The combined organic layers were washed with brine (60 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=100/1 to 15/1) to give 4-((1r,4s)-4-(3- bromo-2-methylphenoxy)cyclohexyl)butanal (400 mg, 1.1 mmol, 80.4% yield) as a yellow oil. Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(2-methyl-3-(((1s,4r)-4-(4- oxobutyl)cyclohexyl)oxy)phenyl)picolinate A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)picolinat e (400 mg, 653.01 µmol, 1 equiv.), 4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butanal (243.70 mg, 718.31 µmol, 1.1 equiv.), [2-(2-aminophenyl)phenyl]palladium (1+); bis(1-adamantyl)-butyl-phosphane; methanesulfonate (47.56 mg, 65.30 µmol, 0.1 equiv.), and K 2 CO 3 (270.75 mg, 1.96 mmol, 3 equiv.) in dioxane (4 mL) and H2O (1 mL) was degassed and purged with N2 and stirred at 100 °C for 1 hour under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=20/1 to 2/1) to give tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(2-methyl-3- (((1s,4r)-4-(4- oxobutyl)cyclohexyl)oxy)phenyl)picolinate (120 mg, 161.0 µmol, 24.6% yield) as a yellow solid MS (ESI) m/z: 745.5 [M+H] + . Step E. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinate To a solution of 3-(1-methyl-7-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (52.74 mg, 161.09 µmol, 1.2 equiv.) in DCM (1 mL) was added NaBH(OAc)3 (85.35 mg, 402.72 µmol, 3 equiv.), AcOH (16.12 mg, 268.48 µmol, 15.35 µL, 2 equiv.), and tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(2-methyl-3-(((1s,4r)-4- (4-oxobutyl)cyclohexyl)oxy)phenyl)picolinate (100 mg, 134.24 µmol, 1 equiv.) at 0 °C. The mixture was stirred at 25 °C for 1 hour. The reaction was diluted with water (20 mL) and extracted with DCM (20 mL × 2). The combined organic layers were washed with brine (30 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give tert- butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)- 4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-y l)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (120 mg, 113.6 µmol, 84.6% yield) as a yellow solid MS (ESI) m/z: 1056.9 [M+H] + . Step F. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-7- yl)piperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picol inate (120 mg, 113.60 µmol, 1 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 118.89 equiv.). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r) -4-(4-(4-(3-(2,6-dioxopiperidin- 3-yl)-1-methyl-1H-indazol-7-yl)piperazin-1-yl)butyl)cyclohex yl)oxy)-2- methylphenyl)picolinic acid (52.3 mg, 48.2 µmol, 42.4% yield, 96.4% purity, FA) as a yellow solid. MS (ESI) m/z: 1000.9 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 13.01 - 12.71 (m, 1H), 10.88 (s, 1H), 8.13 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.49 - 7.41 (m, 4H), 7.40 - 7.31 (m, 2H), 7.10 - 7.01 (m, 3H), 6.98 - 6.89 (m, 2H), 6.61 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.37 - 4.30 (m, 1H), 4.24 (s, 3H), 4.19 (s, 1H), 3.91 (t, J = 5.6 Hz, 2H), 3.26 - 3.18 (m, 4H), 3.02 (t, J = 5.6 Hz, 2H), 2.68 - 2.61 (m, 2H), 2.57 - 2.51 (m, 4H), 2.36 - 2.29 (m, 1H), 2.20 - 2.09 (m, 2H), 2.07 (s, 2H), 1.87 (s, 3H), 1.83 - 1.74 (m, 2H), 1.62 - 1.50 (m, 2H), 1.45 - 1.19 (m, 8H), 1.12 - 1.00 (m, 2H) Example 159. Preparation of Compound 206 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[7-[4-[3-(2,6- dioxo-3-piperidyl)-1-methyl-indazol-7-yl]piperazin-1-yl]hept oxy]-2-methyl-phenyl]pyridine- 2-carboxylic acid Step A. Procedure for preparation of tert-butyl 4-[3-(2,6-dibenzyloxy-3-pyridyl)-1- methyl-indazol-7-yl]piperazine-1-carboxylate To a solution of 7-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (5 g, 9.99 mmol, 1 equiv.) in dioxane (100 mL) was added Pd2(dba)3 (915.01 mg, 999.23 µmol, 0.1 equiv.), RuPhos (932.56 mg, 2.00 mmol, 0.2 eq) and Cs2CO3 (9.77 g, 29.98 mmol, 3 equiv.) then then tert-butyl piperazine-1-carboxylate (5.58 g, 29.98 mmol, 3 equiv.) was added in the mixture. The suspension was degassed and purged with N2 and stirred under N2 at 100 °C for 3 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. Then diluted with H 2 O 100 mL and extracted with ethyl acetate 300 mL (100 mL × 3). The combined organic layers were washed with H 2 O 300 mL (100 mL × 3), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=100/1 to 4/1). The compound tert-butyl 4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-7-yl]piper azine-1-carboxylate (5.6 g, 8.6 mmol, 86.9% yield, 94% purity) was obtained as a yellow oil. MS (ESI) m/z: 606.5 [M+H] + . Step B. Procedure for preparation of tert-butyl 4-[3-(2,6-dioxo-3-piperidyl)-1-methyl- indazol-7-yl] piperazine-1-carboxylate To a solution of tert-butyl 4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-7-yl]piper azine- 1-carboxylate (5.6 g, 9.25 mmol, 1 equiv.) in EtOH (50 mL) and THF (50 mL) was added AcOH (555.17 mg, 9.25 mmol, 528.73 µL, 1 equiv.), Pd(OH)2 (5 g, 7.12 mmol, 20% purity, 0.77 equiv.) and Pd/C (5 g, 4.63 mmol, 10% purity, 0.5 equiv.) under N2 atmosphere. The suspension was degassed and purged with H 2 three times. The mixture was stirred under H 2 (50 Psi) at 50 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to remove solvent. The compound tert-butyl 4-[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-7-yl]piperazine-1-carboxylate (1.3 g, crude) was obtained as a white solid. MS (ESI) m/z: 428.3 [M+H] + . Step C. Procedure for preparation of 3-(1-methyl-7-piperazin-1-yl-indazol-3- yl)piperidine-2, 6-dione To a solution of tert-butyl 4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]piperazin e-1- carboxylate (1 g, 2.34 mmol, 1 equiv.) was added HCl/dioxane (4 M, 584.80 µL, 1 equiv.) .The mixture was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to remove solvent. The compound 3-(1-methyl-7-piperazin-1-yl-indazol-3- yl)piperidine-2, 6-dione (500 mg, crude, HCl) was obtained as a gray solid. Step D. Procedure for preparation of 7-(3-bromo-2-methyl-phenoxy)heptanal To a solution of DMSO (830.02 mg, 10.62 mmol, 830.02 uL, 4 equiv.) in DCM (10 mL) was added (COCl)2 (674.22 mg, 5.31 mmol, 464.98 µL, 2 equiv.) at -70 °C for 1 hour, after addition, 7-(3-bromo-2-methyl-phenoxy)heptan-1-ol (800 mg, 2.66 mmol, 1 equiv.)was added at -70 °C . The reaction mixture was stirred at -70°C for 1 hour, Et 3 N (1.61 g, 15.94 mmol, 2.22 mL, 6 equiv.) was added at -70 °C, the reaction mixture was stirred at -70°C for 1 hour. The mixture solution diluted with H 2 O 10 mL and extracted with DCM 30 mL (10 mL × 3). The combined organic layers were washed with H 2 O 30 mL (10 mL × 3), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 100/1 to 10/1). The compound 7-(3- bromo-2-methyl-phenoxy)heptanal (640 mg, 1.9 mmol, 72.4% yield, 90% purity) was obtained as a yellow oil. Step E. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[2-methyl-3-(7-oxoheptoxy )phenyl]pyridine-2- carboxylate To a solution of 7-(3-bromo-2-methyl-phenoxy)heptanal (203.52 mg, 680.22 µmol, 1 equiv.) in dioxane (5 mL) was added K 2 CO 3 (141.02 mg, 1.02 mmol, 1.5 equiv.), di-tert- butyl(cyclopentyl) phosphane;dichloropalladium;iron (88.67 mg, 136.04 µmol, 0.2 equiv.) and tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) pyridine-2-carboxylate (500 mg, 816.27 µmol, 1.2 equiv.). The mixture was stirred at 100 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by column chromatography (SiO 2 , petroleum ether: ethyl acetate = 2:1). The compound tert-butyl 6-[8- (1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin- 2-yl]-3-[2-methyl-3-(7- oxoheptoxy)phenyl]pyridine-2-carboxylate (250 mg, 329.1 µmol, 48.3% yield, 92.8% purity) was obtained as a yellow solid. MS (ESI) m/z: 705.4 [M+H] + Step F. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[7-[4-[3-(2,6-dioxo-3- piperidyl) -1-methyl-indazol- 7-yl]piperazin-1-yl]heptoxy]-2-methyl-phenyl]pyridine-2-carb oxylate To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[2-methyl-3-(7-oxoheptoxy)phenyl]pyridine-2-carboxyl ate (100 mg, 141.87 µmol, 1 equiv.) in DCM (2 mL) was added 3-(1-methyl-7-piperazin-1-yl-indazol-3-yl)piperidine-2,6- dione (55.73 mg, 170.24 µmol, 1.2 equiv.) and NMM (43.05 mg, 425.61 µmol, 46.79 µL, 3 equiv.). The suspension was degassed and purged with N2 and stirred under N2 at 20 °C for 3 hours. Then the NaBH 3 CN (26.75 mg, 425.61 µmol, 3 equiv.) was added in the mixture. The mixture was stirred under N 2 at 20 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to remove solvent. The compound tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[7-[4-[3- (2,6-dioxo-3-piperidyl) -1- methyl-indazol-7-yl]piperazin-1-yl]heptoxy]-2-methyl-phenyl] pyridine-2-carboxylate (100 mg, crude) was obtained as a yellow oil. MS (ESI) m/z: 1016.8 [M+H] + Step G. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[7-[4-[3-(2,6-dioxo-3-piperidyl)-1 -methyl-indazol-7- yl]piperazin-1-yl]heptoxy]-2-methyl-phenyl]pyridine-2-carbox ylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[7-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol -7-yl]piperazin-1-yl]heptoxy]-2- methyl-phenyl] pyridine-2-carboxylate (80 mg, 78.72 µmol, 1 equiv.) in DCM (1.5 mL) was added TFA (8.98 mg, 78.72 µmol, 5.83 µL, 1 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was diluted with DMF (1.5 mL). The residue was purified by prep-HPLC. The compound 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[7- [4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]piperazi n-1-yl]heptoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid (22.8 mg, 21.0 µmol, 26.7% yield, 92.7% purity, FA) was obtained as a pink solid. MS (ESI) m/z: 960.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.89 (s, 1H), 8.14 (s, 1H), 8.02 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.50 - 7.41 (m, 4H), 7.41 - 7.34 (m, 3H), 7.13 - 7.06 (m, 1H), 7.05 - 6.98 (m, 2H), 6.95 (d, J = 8.8 Hz, 1H), 6.89 (d, J = 8.4 Hz, 1H), 6.63 (d, J = 8.0 Hz, 1H), 4.98 (s, 2H), 4.36 - 4.30 (m, 1H), 4.23 (s, 3H), 3.98 (d, J = 5.6 Hz, 2H), 3.93 - 3.89 (m, 2H), 3.08 - 2.98 (m, 7H), 2.67 (d, J = 2.0 Hz, 2H), 2.60 ( s, 1H), 2.33 (d, J = 2.0 Hz, 3H), 2.16 (d, J = 7.2 Hz, 2H), 1.91 (s, 3H), 1.74 (d, J = 6.8 Hz, 3H), 1.52 - 1.45 (m, 4H), 1.40 - 1.33 (m, 4H) Example 160. Preparation of Compound 207b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((3R)-1-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino )-2-oxoethyl)piperidin-3- yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of (S)-tert-butyl 3-formylpiperidine-1-carboxylate A mixture of (S)-tert-butyl 3-(hydroxymethyl)piperidine-1-carboxylate (5.00 g, 23.22 mmol, 1 equiv.) and DMP (10.84 g, 25.55 mmol, 7.91 mL, 1.1 equiv.) in DCM (50 mL) was degassed and purged with N 2 and stirred at 0 °C for 3 hours under N 2 atmosphere. The reaction mixture was diluted with saturated sodium bicarbonate solution (70 mL) and extracted with DCM (70 mL × 3). The combined organic layers were washed with brine (70 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give (S)-tert- butyl 3-formylpiperidine-1-carboxylate (4 g, crude) as a white solid. Step B. Procedure for preparation of (R,E)-tert-butyl 3-(3-ethoxy-3-oxoprop-1-en-1- yl)piperidine-1-carboxylate A mixture of (S)-tert-butyl 3-formylpiperidine-1-carboxylate (4.00 g, 18.76 mmol, 1 equiv.) in THF (100 mL) was added NaH (0.99 g, 24.75 mmol, 60% purity, 1.32 equiv.) at 0 °C, and the mixture was stirred at 0 °C for 60 minutes. Ethyl 2-(diethoxyphosphoryl)acetate (8.41 g, 37.51 mmol, 7.44 mL, 2 equiv.) was added to the mixture, and then the mixture was stirred at 25 °C for 48 hours under N2 atmosphere. The reaction mixture was quenched by addition water (150 mL) at 0 °C and extracted with EtOAc (200 mL × 3). The combined organic layers were washed with brine (200 mL × 3), dried over anhydrous sodium sulfate filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~18% ethyl acetate/petroleum ether) to give (R,E)-tert-butyl 3-(3-ethoxy-3- oxoprop-1-en-1-yl)piperidine-1-carboxylate (3.1 g, 11.0 mmol, 59.1% yield, 99.1% purity) as a colorless oil. MS (ESI) m/z: 184.1 [M-100+H] + 1 H NMR (400 MHz, CD3Cl) δ = 6.84 (dd, J = 16.0, 6.8 Hz, 1H), 5.86 (d, J = 16.0 Hz, 1H), 4.19 (q, J = 7.2 Hz, 2H), 3.93 - 3.88 (m, 1H), 2.88 - 2.57 (m, 2H), 2.38 - 2.26 (m, 1H), 1.92 - 1.83 (m, 1H), 1.77 - 1.62 (m, 2H), 1.54 - 1.48 (m, 1H), 1.46 (s, 9H), 1.42 - 1.33 (m, 1H), 1.29 (t, J = 7.2 Hz, 3H). Step C. Procedure for preparation of (R)-tert-butyl 3-(3-ethoxy-3-oxopropyl)piperidine- 1-carboxylate To a solution of (R,E)-tert-butyl 3-(3-ethoxy-3-oxoprop-1-en-1-yl)piperidine-1-carboxylate (3.16 g, 11.15 mmol, 1 equiv.) in EtOH (30 mL) was added PtO 2 (253.13 mg, 1.11 mmol, 0.1 equiv.) under N 2 . The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred at 25 °C for 16 hours under H2 (15 psi) atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give (R)-tert-butyl 3-(3- ethoxy-3-oxopropyl)piperidine-1-carboxylate (2.1 g, 7.6 mmol, 68.5% yield) as a colorless oil. 1 H NMR (400 MHz, CD3Cl) δ = 4.09 (q, J = 7.2 Hz, 2H), 3.88 - 3.80 (m, 1H), 2.84 - 2.69 (m, 1H), 2.61 - 2.37 (m, 1H), 2.30 (t, J = 8.0 Hz, 2H), 1.83 - 1.75 (m, 1H), 1.66 - 1.45 (m, 4H), 1.42 (s, 9H), 1.41 - 1.32 (m, 2H), 1.22 (t, J = 7.2 Hz, 3H), 1.14 - 1.02 (m, 1H) Step D. Procedure for preparation of (R)-tert-butyl 3-(3-hydroxypropyl)piperidine-1- carboxylate A mixture of (R)-tert-butyl 3-(3-ethoxy-3-oxopropyl)piperidine-1-carboxylate (1 g, 3.50 mmol, 1 equiv.) in THF (20 mL) was added LiAlH4 (106.40 mg, 2.80 mmol, 0.8 equiv.) at 0 °C. The mixture was stirred at 0 °C for 10 minutes, and then the mixture was stirred at 0 °C for 18 hours under N 2 atmosphere. The reaction mixture was quenched by addition saturated sodium carbonate solution (0.3 mL) at 0 °C, and then diluted with EtOAc (20 mL). The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 30% ethyl acetate/petroleum ether) and prep-HPLC to give (R)-tert-butyl 3-(3-hydroxypropyl)piperidine- 1-carboxylate (370 mg, 1.4 mmol, 42.2% yield, 97.4% purity) as a colorless oil. 1 H NMR (400 MHz, CD3Cl) δ = 4.01 - 3.83 (m, 2H), 3.65 (t, J = 6.4 Hz, 2H), 2.85 - 2.74 (m, 1H), 2.55 - 2.43 (m, 1H), 1.90 - 1.79 (m, 1H), 1.66 - 1.58 (m, 3H), 1.46 (s, 9H), 1.45 - 1.36 (m, 2H), 1.36 - 1.20 (m, 2H), 1.15 - 1.03 (m, 1H). SFC Column: Chiralpak IC-350×4.6mm I.D., 3µm; Mobile phase: Phase A for CO2, and Phase B for MeOH (0.05%DEA); Gradient elution: B in A from 5% to 40%; Flow rate: 3mL/min; Detector: PDA; Column Temp: 35C; Back Pressure: 100 Bar. Step E. Procedure for preparation of (R)-tert-butyl 3-(3-(3-bromo-2- methylphenoxy)propyl)piperidine-1-carboxylate A mixture of (R)-tert-butyl 3-(3-hydroxypropyl)piperidine-1-carboxylate (320 mg, 1.32 mmol, 1 equiv.), 3-bromo-2-methyl-phenol (270.55 mg, 1.45 mmol, 1.1 equiv.), and 2-(tributyl-λ 5 - phosphanylidene)acetonitrile (380.86 mg, 1.58 mmol, 1.2 equiv.) in toluene (10 mL) was degassed and purged with N 2 and stirred at 120 °C for 16 hours under N 2 atmosphere. The reaction mixture was diluted with water (30 mL) and extracted with EtOAc (30 mL × 3). The combined organic layers were washed with brine (30 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by reverse-phase HPLC (with 0.1% HCOOH) to give (R)-tert-butyl 3-(3-(3-bromo-2- methylphenoxy)propyl)piperidine-1-carboxylate (300 mg, 727.5 µmol, 55.3% yield, 100% purity) as a yellow oil. MS (ESI) m/z: 313.8 [M-100+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 7.16 - 7.12 (m, 1H), 7.11 - 7.05 (m, 1H), 6.95 (d, J = 7.6 Hz, 1H), 3.96 (t, J = 6.4 Hz, 2H), 3.84 - 3.62 (m, 2H), 2.84 - 2.75 (m, 1H), 2.23 (s, 3H), 1.80 - 1.70 (m, 3H), 1.62 - 1.52 (m, 1H), 1.37 (s, 9H), 1.35 - 1.04 (m, 6H). Step F. Procedure for preparation of (R)-3-(3-(3-bromo-2- methylphenoxy)propyl)piperidine A mixture of (R)-tert-butyl 3-(3-(3-bromo-2-methylphenoxy)propyl)piperidine-1-carboxylat e (270 mg, 654.77 µmol, 1 equiv.) in HCl/dioxane (5 mL) and DCM (5 mL) was degassed and purged with N 2 and stirred at 25 °C for 1 hour under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give (R)-3-(3-(3-bromo-2- methylphenoxy)propyl)piperidine (260 mg, crude, HCl) as a white solid. MS (ESI) m/z: 311.9 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.17 - 7.13 (m, 1H), 7.12 - 7.06 (m, 1H), 6.96 (d, J = 8.0 Hz, 1H), 3.97 (t, J = 6.0 Hz, 2H), 3.39 (s, 3H), 3.24 - 3.12 (m, 2H), 2.78 - 2.66 (m, 1H), 2.23 (s, 3H), 1.80 - 1.72 (m, 4H), 1.46 - 1.31 (m, 2H), 1.26 - 1.02 (m, 2H) Step G. Procedure for preparation of (R)-ethyl 2-(3-(3-(3-bromo-2- methylphenoxy)propyl)piperidin-1-yl)acetate A mixture of (R)-3-(3-(3-bromo-2-methylphenoxy)propyl)piperidine (250 mg, 716.94 µmol, 1 equiv., HCl), ethyl 2-bromoacetate (143.68 mg, 860.33 µmol, 95.15 µL, 1.2 equiv.), and K2CO3 (495.43 mg, 3.58 mmol, 5 equiv.) in CH3CN (4 mL) and DMF (5 mL) was degassed and purged with N 2 and stirred at 70 °C for 1 hour under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by reverse- phase HPLC (with 0.1% HCOOH) to give (R)-ethyl 2-(3-(3-(3-bromo-2- methylphenoxy)propyl)piperidin-1-yl)acetate (130 mg, 283.1 µmol, 39.4% yield, 96.9% purity, FA) as a yellow oil. MS (ESI) m/z: 398.0 [M+H] + 1 H NMR (400 MHz, DMSO-d6,) δ = 8.13 (s, 2H), 7.17 - 7.13 (m, 1H), 7.11 - 7.06 (m, 1H), 6.95 (d, J = 8.0 Hz, 1H), 4.13 - 4.06 (m, 2H), 3.96 (t, J = 6.0 Hz, 2H), 3.34 (s, 2H), 2.93 - 2.81 (m, 2H), 2.23 (s, 3H), 2.04 - 1.97 (m, 1H), 1.77 - 1.69 (m, 3H), 1.67 - 1.42 (m, 4H), 1.37 - 1.29 (m, 2H), 1.19 (t, J = 7.2 Hz, 3H), 0.93 - 0.82 (m, 1H) Step H. Procedure for preparation of (R)-tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2- ethoxy-2- oxoethyl)piperidin-3-yl)propoxy)-2-methylphenyl)picolinate (R)-ethyl 2-(3-(3-(3-bromo-2-methylphenoxy)propyl)piperidin-1-yl)aceta te (120 mg, 301.26 µmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pic olinate (221.44 mg, 361.51 µmol, 1.2 equiv.), KF (1.5 M, 602.51 µL, 3 equiv.), and Ad 2 nBuP Pd G 3 (cataCXium® A Pd G 3 ) (21.94 mg, 30.13 µmol, 0.1 equiv.) were taken up into a microwave tube in dioxane (3 mL). The sealed tube was heated at 100 °C for 1 hour under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep- TLC (SiO 2 , petroleum ether : ethyl acetate = 1:1) to give (R)-tert-butyl 6-(8-(benzo[d]thiazol- 2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-( 2-ethoxy-2-oxoethyl)piperidin- 3-yl)propoxy)-2-methylphenyl)picolinate (200 mg, 233.3 µmol, 77.4% yield, 93.8% purity) as a yellow oil. MS (ESI) m/z: 804.8 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 12.99 - 12.72 (m, 1H), 8.05 - 7.98 (m, 1H), 7.77 (d, J = 8.0 Hz, 1H), 7.60 (d, J = 7.6 Hz, 1H), 7.48 - 7.41 (m, 3H), 7.39 - 7.31 (m, 2H), 7.12 - 7.06 (m, 1H), 6.96 – 6.88 (m, 2H), 6.57 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.10 - 3.99 (m, 4H), 3.93 (s, 6H), 3.15 (s, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.80 - 2.67 (m, 2H), 1.99 (s, 2H), 1.86 (s, 3H), 1.77 - 1.65 (m, 4H), 1.18 - 1.15 (m, 4H), 1.00 (s, 9H) Step I. Procedure for preparation of (R)-2-(3-(3-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)propyl)piperidin-1-yl)acetic acid A mixture of (R)-tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-ethoxy-2-oxoethyl)piperidin-3-yl)pro poxy)-2- methylphenyl)picolinate (190 mg, 236.32 µmol, 1 equiv.) and LiOH·H2O (49.58 mg, 1.18 mmol, 5 equiv.) in THF (4 mL) and H 2 O (1 mL) was degassed and purged with N 2 and stirred at 25 °C for 36 hours under N 2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (25 mL × 3). The combined organic layers were washed with brine (15 mL × 3), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (R)-2-(3-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)propyl)piperidin-1-yl)acetic acid (100 mg, 128.8 µmol, 54.5% yield) as a yellow oil. MS (ESI) m/z: 776.3 [M+H] + 1 H NMR (400 MHz, DMSO-d6,) δ = 13.24 - 12.50 (m, 1H), 8.73 - 8.37 (m, 1H), 8.09 - 7.99 (m, 1H), 7.82 - 7.72 (m, 1H), 7.64 - 7.57 (m, 1H), 7.53 - 7.29 (m, 4H), 7.20 - 7.05 (m, 1H), 7.04 - 6.83 (m, 2H), 6.50 (d, J = 5.2 Hz, 1H), 5.27 - 4.82 (m, 4H), 4.11 - 3.94 (m, 6H), 3.91 - 3.83 (s, 3H), 3.44 -3.36 (m, 2H), 3.08 - 3.00 (m, 2H), 1.99 (s, 2H), 1.91 - 1.87 (m, 4H), 1.81 - 1.76 (m, 4H), 1.07 (s, 9H). Step J. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3R)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-3-yl)propoxy)-2- methylphenyl)picolinate A mixture of (R)-2-(3-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)piperidin-1- yl)acetic acid (90 mg, 115.99 µmol, 1 equiv.), 3-(6-amino-1-methyl-1H-indazol-3- yl)piperidine-2,6-dione (32.95 mg, 127.59 µmol, 1.1 equiv.), and DIEA (44.97 mg, 347.97 µmol, 60.61 µL, 3 equiv.) in DMF (1.2 mL) was degassed and purged with N2 and stirred at 25 °C for 5 minutes. After 5 minutes, HATU (52.92 mg, 139.19 µmol, 1.2 equiv.) was added, and the mixture was stirred at 25 °C for 1 hour under N 2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (25 mL × 3). The combined organic layers were washed with brine (15 mL × 6), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3R)-1-(2-((3-(2,6 -dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-3-yl)propoxy)-2- methylphenyl)picolinate (110 mg, crude) as a pink oil. MS (ESI) m/z: 508.9 [M/2+H] + Step K. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3R)-1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-3-yl)propoxy)-2- methylphenyl)picolinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-((3R)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-6-yl)amino)-2- oxoethyl)piperidin-3-yl)propoxy)-2-methylphenyl)picolinate (100 mg, 98.40 µmol, 1 equiv.) in DCM (0.5 mL) and TFA (0.5 mL) was degassed and purged with N2 and stirred at 25 °C for 16 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-((3R)-1-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2- oxoethyl)piperidin-3- yl)propoxy)-2-methylphenyl)picolinic acid (19.0 mg, 19.8 µmol, 20.1% yield, 100% purity) as a yellow solid. MS (ESI) m/z: 960.6 [M+H] + 1 H NMR (400 MHz, DMSO-d6,) δ = 13.11 - 12.36 (m, 1H), 10.92 - 10.84 (m, 1H), 9.85 (s, 1H), 8.15 (s, 1H), 8.06 - 7.99 (m, 2H), 7.81 - 7.75 (m, 1H), 7.65 - 7.58 (m, 2H), 7.50 - 7.31 (m, 5H), 7.23 - 7.17 (m, 1H), 7.10 - 7.03 (m, 1H), 6.97 - 6.91 (m, 1H), 6.88 - 6.82 (m, 1H), 6.64 - 6.59 (m, 1H), 4.97 (s, 2H), 4.34 - 4.27 (m, 1H), 4.00 - 3.91 (m, 3H), 3.90 (s, 3H), 3.21 - 3.08 (m, 4H), 3.05 - 2.99 (m, 2H), 2.89 - 2.78 (m, 2H), 2.67 - 2.58 (m, 2H), 2.37 - 2.28 (m, 2H), 2.20 - 2.11 (m, 2H), 1.88 (s, 3H), 1.82 - 1.67 (m, 4H), 1.66 - 1.58 (m, 2H), 1.41 - 1.32 (m, 2H). Example 161. Preparation of Compound 209a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[(3R,5S)-4- [2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]- 2-oxo-ethyl]-3,5-dimethyl- piperazin-1-yl]-2,2-difluoro-propoxy]-2-methyl-phenyl]pyridi ne-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(3R,5S)-4-[2-[[3-( 2,6-dioxo-3-piperidyl)-1- methyl-indazol-7-yl]amino]-2-oxo-ethyl]-3,5-dimethyl-piperaz in-1-yl]-2,2-difluoro- propoxy]-2-methyl-phenyl]pyridine-2-carboxylate A solution of 2-[(2R,6S)-4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3, 4-dihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]-2,2-difluoro-propyl]- 2,6-dimethyl-piperazin-1-yl]acetic acid (100 mg, 118.91 µmol, 1.0 equiv.), 3-(7-amino-1- methyl-indazol-3-yl)piperidine-2,6-dione (46.07 mg, 178.36 µmol, 1.5 equiv.), and EDCI (34.19 mg, 178.36 µmol, 1.5 equiv.) in pyridine (1.0 mL) was stirred at 25 °C for 5 hours. The mixture was quenched by water (10 mL) and filtered. The filter cake was concentrated under vacuum to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[3-[(3R,5S)-4-[2-[[3-(2,6-dioxo-3-piperidyl)-1-me thyl-indazol-7-yl]amino]-2-oxo- ethyl]-3,5-dimethyl-piperazin-1-yl]-2,2-difluoro-propoxy]-2- methyl-phenyl]pyridine-2- carboxylate (135 mg, crude) as a white solid. MS (ESI) m/z: 1082.8 [M+H] + . Step B. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[(3R,5S)-4-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol- 7-yl]amino]-2-oxo-ethyl]-3,5-dimethyl-piperazin-1-yl]-2,2-di fluoro-propoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[(3R,5S)-4-[2-[[3-(2,6-dioxo-3-piperidyl)-1-meth yl-indazol-7-yl]amino]-2-oxo- ethyl]-3,5-dimethyl-piperazin-1-yl]-2,2-difluoro-propoxy]-2- methyl-phenyl]pyridine-2- carboxylate (130 mg, 120.23 µmol, 1.0 equiv.) in TFA (1 mL) and DCM (1 mL) was stirred at 25 °C for 16 hours. The mixture was concentrated to remove DCM. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[(3R,5S)-4-[2-[[3-(2,6-dioxo-3-piperidyl)-1-meth yl-indazol-7-yl]amino]-2-oxo- ethyl]-3,5-dimethyl-piperazin-1-yl]-2,2-difluoro-propoxy]-2- methyl-phenyl]pyridine-2- carboxylic acid (61.9 mg, 58.1 µmol, 48.3% yield, 96.1% purity) as a white solid. MS (ESI) m/z: 513.0 [M/2+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.87 (s, 1H), 10.90 (s, 1H), 9.79 (s, 1H), 8.14 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.57 (d, J = 8.0 Hz, 1H), 7.49 - 7.42 (m, 3H), 7.39 - 7.32 (m, 2H), 7.30 (d, J = 7.2 Hz, 1H), 7.16 - 7.11 (m, 1H), 7.07 (t, J = 8.0 Hz, 1H), 6.98 (dd, J = 6.0, 8.4 Hz, 2H), 6.71 (d, J = 7.6 Hz, 1H), 5.06 - 4.93 (m, 2H), 4.40 - 4.31 (m, 3H), 4.10 (s, 3H), 3.92 (t, J = 6.0 Hz, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.94 (s, 1H), 2.92 - 2.85 (m, 2H), 2.78 (s, 1H), 2.75 (d, J = 10.8 Hz, 2H), 2.70 - 2.66 (m, 2H), 2.62 (t, J = 5.2 Hz, 1H), 2.39 - 2.32 (m, 1H), 2.24 - 2.15 (m, 3H), 1.95 (s, 1H), 1.93 (s, 3H), 1.04 (d, J = 6.0 Hz, 6H). 19 F NMR (400 MHz, DMSO-d6) δ = 106.40. Example 162. Preparation of Compound 211a

6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(2- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1- yl)ethyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of methyl 4-(3-bromo-2-methyl-phenoxy)cyclohexane carboxylate To a solution of methyl 4-hydroxycyclohexanecarboxylate (16 g, 101.14 mmol, 1.0 equiv.) and 3-bromo-2-methyl-phenol (20.81 g, 111.26 mmol, 1.1 equiv.) in Tol. (500 mL) was added 2- (tributyl-λ5-phosphanylidene)acetonitrile (29.29 g, 121.37 mmol, 1.2 equiv.).The mixture was stirred at 120 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give the compound methyl 4-(3-bromo-2-methyl-phenoxy)cyclohexane carboxylate (125 g, crude) was obtained as a black oil, which was used in the next step without further purification. Step B. Procedure for preparation of [4-(3-bromo-2-methyl- phenoxy)cyclohexyl]methanol A mixture of methyl 4-(3-bromo-2-methyl-phenoxy)cyclohexane carboxylate (28 g, 34.23 mmol, 40% purity, 1.0 equiv.) in THF (300 mL) was slowly added LAH (1.36 g, 35.94 mmol, 1.05 equiv.) at 0 °C then the mixture was stirred at 0 ℃ for 1 hour. The mixture was quenched by 2.5 g Na2SO4.10 H2O. The solution was poured into ice-water (250 mL) slowly and the pH was adjusted to pH 4-5 with 1 M HCl, extracted with EtOAc 600 mL (200 mL×3), washed with brine 300 mL (150 mL×2), dried over by sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (Eluent of 0~18% ethyl acetate/petroleum ether). The compound [4-(3-bromo-2-methyl- phenoxy)cyclohexyl]methanol (10 g, 32.3 mmol, 47.2% yield, 96.7% purity) was obtained as a brown oil. Step C. Procedure for preparation of 4-(3-bromo-2-methyl-phenoxy)cyclohexane carbaldehyde To a solution of DMSO (5.22 g, 66.84 mmol, 5.22 mL, 4 equiv.) in DCM (200 mL) was added the mixture of (COCl)2 (4.24 g, 33.42 mmol, 2.93 mL, 2 equiv.) under -78°C and stirred for 1 hour. And [4-(3-bromo-2-methyl-phenoxy)cyclohexyl]methanol (5 g, 16.71 mmol, 1.0 equiv.) was added into the mixture and stirred for 1 hour. TEA (10.15 g, 100.27 mmol, 13.96 mL, 6.0 equiv.) was added into the mixture and stirred at -70 °C for 1 hour. The reaction mixture was diluted with H 2 O (200) mL and extracted with DCM 60 mL (30 mL × 2).Dried over [Na 2 SO 4 ] and concentrated under reduced pressure to give 4-(3-bromo-2-methyl-phenoxy)cyclohexane carbaldehyde (17 g, crude) as a yellow oil, which was used into nest step without further purification. Step D. Procedure for preparation of 1-bromo-3-[4-[(E)-2-methoxyvinyl]cyclohexoxy]-2- methyl-benzene A mixture of methoxymethyl (triphenyl)phosphonium; bromide (1.69 g, 4.37 mmol, 1.30 equiv.) in THF (10 mL) was degassed and purged with N 2 . The mixture was slowly added LiHMDS (1 M, 4.37 mL, 1.30 equiv.) at 0°C and stirred for 2 hours. Then, 4-(3-bromo-2- methyl-phenoxy)cyclohexane carbaldehyde (1 g, 3.36 mmol, 1.00 equiv.) was added to the mixture and stirred at 25 °C for 13 hours under N 2 atmosphere. The reaction mixture was quenched by addition saturated NH4Cl (50 mL), and then extracted with ethyl acetate (40 mL × 3). The combined organic layers were washed with brine 90 mL (30 mL × 3), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=1/0 to 10/1) to give 1-bromo-3-[4-[(E)-2-methoxyvinyl]cyclohexoxy]-2-methyl-benze ne (0.6 g, 1.8 mmol, 54.8% yield) as a white solid. Step E. Procedure for preparation of 2-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]acetaldehyde A mixture of 1-bromo-3-[4-[(E)-2-methoxyvinyl]cyclohexoxy]-2-methyl-benze ne (0.6 g, 1.84 mmol, 1 equiv.), HCl (2 M, 2.77 mL, 3 equiv.) in THF (2 mL) was stirred at 80 °C for 12 hours under N2 atmosphere. The reaction mixture was diluted with water 10 mL and extracted with ethyl acetate (20 mL × 3). The combined organic layers were washed with brine 30 mL (10 mL × 3), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=1/0 to 10/1) to give 2-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]acetaldehyde (0.3 g, 889.7 µmol, 48.3% yield, 92.3% purity) was obtained as a colorless oil. Step F. Procedure for preparation of 2-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]ethanol A mixture of 2-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]acetaldehyde (200.00 mg, 642.65 µmol, 1.00 equiv.), LiAlH4 (24.39 mg, 642.65 µmol, 1.00 equiv.) in THF (5 mL) was degassed and purged with N 2 and stirred at 0 °C for 1 hour under N 2 atmosphere. The reaction mixture was quenched by addition water 5 mL, and then extracted with ethyl acetate (10 mL × 2). The combined organic layers were washed with brine 20 mL (10 mL × 2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was further separated by SFC. The compound 2-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]ethanol (60 mg, 191.5 µmol, 29.8% yield) was obtained as a white solid. 1 H NMR (400 MHz, CH 3 OD) δ = 7.05 (d, J = 8.0 Hz, 1H), 6.95 (t, J = 8.0 Hz, 1H), 6.85 (d, J = 8.0 Hz, 1H), 4.20 - 4.06 (m, 1H), 3.62 - 3.52 (m, 2H), 2.21 (s, 3H), 2.08 (d, J = 12.4 Hz, 2H), 1.81 (d, J = 13.2 Hz, 2H), 1.46 - 1.33 (m, 5H), 1.12 - 0.99 (m, 2H). Step G. Procedure for preparation of 2-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]acetaldehyde To a solution of oxalyl dichloride (50.91 mg, 401.07 µmol, 35.11 µL, 2 equiv.) in DCM (3 mL) was added the mixture of DMSO (62.67 mg, 802.13 µmol, 62.67 µL, 4 equiv.) under -70°C and stirred for 1 h. And 2-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]ethanol (62.81 mg, 200.53 µmol, 1 equiv.) was added into the mixture and stirred for 1 h. TEA (121.75 mg, 1.20 mmol, 167.47 µL, 6 equiv.) was added into the mixture and stirred at -70 °C for 1 h. The reaction mixture was filtered and concentrated under reduced pressure to remove solvent. The residue was diluted with ethyl acetate (200 mL) and extracted with H 2 O (30 mL × 2). Dried over Na2SO4, and concentrated under reduced pressure to give a residue. The compound 2-[4- (3-bromo-2-methyl-phenoxy)cyclohexyl]acetaldehyde (50 mg, 160.66 µmol, 80.1% yield) was obtained as a colorless oil. Step H. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[2-methyl-3-[4-(2- oxoethyl)cyclohexoxy]phenyl]pyridine-2-carboxylate A mixture of 2-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]acetaldehyde (50 mg, 160.66 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (108.25 mg, 176.73 µmol, 1.1 equiv.), Ad2nBuP Pd G3 (11.70 mg, 16.07 µmol, 0.1 equiv.), KF (1.5 M, 321.32 µL, 3 equiv.) in dioxane (2 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 100 °C for 1 h under N 2 atmosphere in microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, petroleum ether : ethyl acetate = 2:1). The compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[2-methyl-3-[4- (2-oxoethyl)cyclohexoxy]phenyl]pyridine-2-carboxylate (130 mg, 117.6 µmol, 73.2% yield, 64.9% purity) was obtained as a yellow solid. MS (ESI) m/z: 717.5 [M+H] + . Step I. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[2-[4-[3-(2,6-dioxo-3-p iperidyl)-1-methyl-indazol- 6-yl]piperazin-1-yl]ethyl]cyclohexoxy]-2-methyl-phenyl]pyrid ine-2-carboxylate A mixture of 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (27.40 mg, 83.70 µmol, 1 equiv.), NaBH(OAc)3 (26.61 mg, 125.54 µmol, 1.5 equiv.), AcOH (1.01 mg, 16.74 µmol, 9.57e-1 µL, 0.2 equiv.) in DCM (2 mL) was slowly added tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[2-methyl-3-[4-(2- oxoethyl)cyclohexoxy]phenyl]pyridine-2-carboxylate (60 mg, 83.70 µmol, 1 equiv.) at 0°C, the mixture was stirred at 25°C for 1 h under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The compound tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[4-[2-[4-[3-(2,6-dioxo- 3-piperidyl)-1-methyl-indazol-6-yl]piperazin-1-yl]ethyl]cycl ohexoxy]-2-methyl- phenyl]pyridine-2-carboxylate (65 mg, crude) was obtained as a yellow solid. Step J. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(2-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)ethyl)cyclohexyl)oxy)- 2-methylphenyl)picolinic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[4-[2-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazo l-6-yl]piperazin-1- yl]ethyl]cyclohexoxy]-2-methyl-phenyl]pyridine-2-carboxylate (62 mg, 60.30 µmol, 1 equiv.) in TFA (0.5 mL) and DCM (0.5 mL) was stirred at 40 °C for 1 h under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(2-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-6- yl)piperazin-1-yl)ethyl)cyclohexyl)oxy)-2-methylphenyl)picol inic acid (24.1 mg, 23.8 µmol, 39.5% yield, 95.9% purity) as a yellow solid. MS (ESI) m/z: 972.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.87 (d, J = 4.8 Hz, 1H), 12.76 - 12.28 (m, 1H), 10.86 (s, 1H), 8.04 (d, J = 7.2 Hz, 1H), 7.80 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 7.2 Hz, 1H), 7.57 (d, J = 9.2 Hz, 1H), 7.45 (d, J = 8.4 Hz, 3H), 7.37 (d, J = 10.0 Hz, 2H), 7.09 (s, 1H), 6.99 (s, 4H), 6.63 (d, J = 7.2 Hz, 1H), 4.99 (s, 2H), 4.29 (dd, J = 5.2, 9.2 Hz, 1H), 4.24 (s, 1H), 3.93 (s, 3H), 3.66 - 3.60 (m, 2H), 3.24 - 3.16 (m, 4H), 3.04 (d, J = 6.0 Hz, 4H), 2.62 (s, 2H), 2.34 (s, 2H), 2.21 - 2.06 (m, 4H), 1.88 (s, 3H), 1.85 - 1.79 (m, 2H), 1.66 - 1.59 (m, 2H), 1.38 (d, J = 1.6 Hz, 4H), 1.24 (s, 1H), 1.17 (s, 2H) Example 163. Preparation of Compound 214a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pip erazin-1-yl)-4- oxobutyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of 4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butanoic acid A mixture of 4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butanal (500 mg, 1.47 mmol, 1 equiv.), NaH2PO4 (529.11 mg, 4.41 mmol, 3 equiv.), sodium chlorite (398.85 mg, 4.41 mmol, 3 equiv.), and 2-methylbut-2-ene (618.55 mg, 8.82 mmol, 934.36 µL, 6 equiv.) in t-BuOH (6 mL), THF (2 mL), and H 2 O (1 mL) was stirred at 25 °C for 12 hours under N 2 atmosphere. The reaction was diluted with water (50 mL) and extracted with DCM (80 mL × 2). The combined organic layers were washed with brine (60 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 20/1 to 8/1) to give 4- ((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butanoic acid (400 mg, 1.1 mmol, 76.5% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ = 11.98 (s, 1H), 7.17 - 7.11 (m, 1H), 7.10 - 6.99 (m, 2H), 4.30 - 4.16 (m, 1H), 2.21 (s, 3H), 2.20 - 2.17 (m, 1H), 2.04 (d, J = 11.6 Hz, 2H), 1.76 (d, J = 11.6 Hz, 2H), 1.56 - 1.47 (m, 2H), 1.42 - 1.33 (m, 2H), 1.31 - 1.15 (m, 4H), 1.09 - 0.98 (m, 2H) Step B. Procedure for preparation of 4-((1r,4s)-4-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)cyclohexyl)butanoic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)picolinat e (600 mg, 979.52 µmol, 1 equiv.), 4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butanoic acid (382.79 mg, 1.08 mmol, 1.1 equiv.), [2-(2-aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-buty l- phosphane;methanesulfonate (71.34 mg, 97.95 µmol, 0.1 equiv.), and KF (170.73 mg, 2.94 mmol, 68.84 µL, 3 equiv.) in dioxane (6 mL) and H2O (2 mL) was degassed and purged with N 2 and stirred at 100 °C for 12 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, dichloromethane : methanol = 20/1 to 10/1) to give 4-((1r,4s)-4-(3-(6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2( 1H)-yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)cyclohexyl)buta noic acid (400 mg, 525.6 µmol, 53.6% yield) as a yellow solid MS (ESI) m/z: 761.8 [M+H] + . Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)-4-oxobutyl)cyclohexyl )oxy)-2- methylphenyl)picolinate To a solution of 4-((1r,4s)-4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)cyclohexyl)butanoic acid (100 mg, 131.42 µmol, 1 equiv.) in pyridine (1 mL) was added EDCI (32.75 mg, 170.84 µmol, 1.3 equiv.) and 3-(1-methyl-7-piperazin-1-yl- indazol-3-yl)piperidine-2,6-dione (51.63 mg, 157.70 µmol, 1.2 equiv.). The mixture was stirred at 60 °C for 12 hours. The reaction was diluted with water (20 mL) and extracted with DCM (20 mL × 2). The combined organic layers were washed with brine (30 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)piperazin-1-yl)-4-oxobutyl)cyclohexyl)oxy)-2 -methylphenyl)picolinate (60 mg, 56.0 µmol, 42.6% yield) as a yellow solid. MS (ESI) m/z: 1170.8 [M+H] + . Step D. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)-4-oxobutyl)cyclohexyl )oxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-7- yl)piperazin-1-yl)-4-oxobutyl)cyclohexyl)oxy)-2-methylphenyl )picolinate (60 mg, 56.06 µmol, 1 equiv.) in DCM (0.5 mL) was added TFA (770.00 mg, 6.75 mmol, 0.5 mL, 120.47 equiv.). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1s,4r)-4-(4-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin -1-yl)-4- oxobutyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (15.3 mg, 14.8 µmol, 26.5% yield, 97.8% purity) as a yellow solid. MS (ESI) m/z: 1014.9 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.87 (s, 1H), 12.72 - 12.45 (m, 1H), 10.89 (s, 1H), 8.04 (d, J = 8.0 Hz, 1H), 7.80 (d, J = 8.4 Hz, 1H), 7.63 (d, J = 7.2 Hz, 1H), 7.48 - 7.42 (m, 4H), 7.40 - 7.33 (m, 2H), 7.08 - 7.02 (m, 3H), 6.98 - 6.92 (m, 2H), 6.62 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.53 - 4.43 (m, 1H), 4.37 - 4.33 (m, 1H), 4.28 (s, 3H), 4.24 - 4.17 (m, 1H), 3.92 (t, J = 5.6 Hz, 2H), 3.24 - 3.17 (m, 2H), 3.03 (t, J = 5.2 Hz, 2H), 2.68 - 2.61 (m, 4H), 2.40 - 2.30 (m, 4H), 2.24 - 2.12 (m, 2H), 2.11 - 2.05 (m, 2H), 2.00 - 1.99 (m, 1H), 1.88 (s, 3H), 1.80 (d, J = 12.8 Hz, 2H), 1.59 - 1.53 (m, 2H), 1.41 - 1.34 (m, 2H), 1.30 - 1.20 (m, 4H), 1.12 - 1.02 (m, 2H) Example 164. Preparation of Compound 215a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(2- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pip erazin-1- yl)ethyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[2-[4-[3-(2,6-dioxo -3-piperidyl)-1-methyl- indazol-7-yl]piperazin-1-yl]ethyl]cyclohexoxy]-2-methyl-phen yl]pyridine-2-carboxylate To a mixture of 3-(1-methyl-7-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (27.40 mg, 83.70 µmol, 1 equiv.), NaBH(OAc)3 (26.61 mg, 125.54 µmol, 1.5 equiv.), and AcOH (5.03 mg, 83.70 µmol, 4.79 µL, 0.1 equiv.) in DCM (2 mL) was added tert-butyl 6-[8-(1,3-benzothiazol- 2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[2-methyl- 3-[4-(2- oxoethyl)cyclohexoxy]phenyl]pyridine-2-carboxylate (60 mg, 83.70 µmol, 1 equiv.) at 0 °C. The mixture was degassed and purged with N2 and stirred at 25 °C for 1 hour under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4- [2-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]piper azin-1-yl]ethyl]cyclohexoxy]-2- methyl-phenyl]pyridine-2-carboxylate (62 mg, crude) as a yellow solid. MS (ESI) m/z: 514.9 [M+2H/2] + Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(2-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)ethyl)cyclohexyl)oxy)- 2-methylphenyl)picolinic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[4-[2-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazo l-7-yl]piperazin-1- yl]ethyl]cyclohexoxy]-2-methyl-phenyl]pyridine-2-carboxylate (60 mg, 58.35 µmol, 1 equiv.) in TFA (0.5 mL) and DCM (1.5 mL) was degassed and purged with N 2 and stirred at 25 °C for 25 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC. The compound 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1r,4r)-4-(2-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin -1-yl)ethyl)cyclohexyl)oxy)-2- methylphenyl)picolinic acid (23.2 mg, 22.7 µmol, 38.9% yield, 95.0% purity) was obtained as a yellow solid. MS (ESI) m/z: 972.6 [M+H] + Example 165. Preparation of Compound 222 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-((6-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin-1-yl )hexyl)oxy)-2- methylphenyl)picolinic acid Step A. Procedure for preparation of methyl 6-(3-bromo-2-methylphenoxy)hexanoate To a solution of 6-(3-bromo-2-methylphenoxy)hexan-1-ol (2 g, 10.69 mmol, 1 equiv.) in CH 3 CN (20 mL) was added K 2 CO 3 (4.43 g, 32.08 mmol, 3 equiv.) and methyl 6- bromohexanoate (2.46 g, 11.76 mmol, 1.1 equiv.). The mixture was stirred at 60 °C for 2 hours. After cooling to room temperature, ethyl acetate (50 mL) and water (50 mL) were added, and layers were separated. The aqueous phase was extracted with organic layers and washed with ethyl acetate (30 mL × 2). The combined organic extracts were dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum. The residue was purified by flash silica gel chromatography (Eluent of 0~2% ethyl acetate/petroleum ether) to give methyl 6-(3-bromo-2- methylphenoxy)hexanoate (3 g, 9.5 mmol, 89.0% yield) as a colorless oil. 1 H NMR (400 MHz, CDC13) δ = 7.14 (d, J = 8.0 Hz, 1H), 6.99 (t, J = 8.0 Hz, 1H), 6.75 (d, J = 8.4 Hz, 1H), 3.95 (t, J = 6.4 Hz, 2H), 3.68 (s, 3H), 2.36 (t, J = 7.6 Hz, 2H), 2.31 (s, 3H), 1.85 - 1.80 (m, 2H), 1.75 - 1.69 (m, 2H), 1.55 - 1.50 (m, 2H) Step B. Procedure for preparation of 6-(3-bromo-2-methylphenoxy)hexan-1-ol To a solution of methyl 6-(3-bromo-2-methylphenoxy)hexanoate (1.5 g, 4.76 mmol, 1 equiv.) in THF (15 mL) was added LiAlH4 (180.60 mg, 4.76 mmol, 1 equiv.) at 0 °C. The mixture was stirred at 0 °C for 2 hours. The reaction mixture was quenched by addition of water (0.2 mL) and 15% NaOH (0.2 mL) and water (0.6 mL). The combined mixture was treated with Na 2 SO 4 , filtered, and concentrated under reduced pressure to give 6-(3-bromo-2-methylphenoxy)hexan- 1-ol (1.2 g, 4.1 mmol, 87.8% yield) as a yellow oil. The 6-(3-bromo-2-methylphenoxy)hexan- 1-ol residue was further purified by prep-HPLC to give 6-(3-bromo-2-methylphenoxy)hexan- 1-ol (501.28 mg, 1.7 mmol, 66.2% yield, 99.3% purity) as a yellow oil. MS (ESI) m/z: 288.8 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 7.16 - 7.13 (m, 1H), 7.11 - 7.06 (m, 1H), 6.96 (d, J = 8.0 Hz, 1H), 4.33 (t, J = 5.2 Hz, 1H), 3.97 (t, J = 6.4 Hz, 2H), 3.43 - 3.36 (m, 2H), 2.23 (s, 3H), 1.79 - 1.67 (m, 2H), 1.46 - 1.40 (m, 4H), 1.39 - 1.30 (m, 2H) Step D. Procedure for preparation of 6-(3-bromo-2-methylphenoxy)hexanal To a solution of oxalyl dichloride (1.06 g, 8.36 mmol, 731.51 uL, 2 equiv.) in DCM (2 mL) was dropwise added a solution of DMSO (1.31 g, 16.71 mmol, 1.31 mL, 4 equiv.) in DCM (2 mL) at -70 °C under N2 atmosphere. The mixture was stirred at -70 °C for 1 hour. Then 6-(3- bromo-2-methylphenoxy)hexan-1-ol (1.2 g, 4.18 mmol, 1 equiv.) in DCM (10 mL) was added dropwise at -70 °C. The solution was stirred for 1 hour at -70 °C. Then TEA (2.54 g, 25.07 mmol, 3.49 mL, 6 equiv.) was added to the solution. The solution was stirred at -70 °C for 0.5 hour under N2 atmosphere. Water (50 mL) was added, and layers were separated. The aqueous phase was extracted with DCM 60 mL (30 mL × 2), dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum to give 6-(3-bromo-2-methylphenoxy)hexanal (1.0 g, 3.5 mmol, 83.9% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 9.72 - 9.61 (m, 1H), 7.17 - 7.12 (m, 1H), 7.11 - 7.06 (m, 1H), 6.96 (d, J = 8.0 Hz, 1H), 3.97 (t, J = 6.4 Hz, 2H), 2.47 - 2.43 (m, 2H), 2.23 (s, 3H), 1.79 - 1.68 (m, 2H), 1.62 - 1.55 (m, 2H), 1.47 - 1.38 (m, 2H) Step E. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(2-methyl-3-((6-oxohexyl)oxy) phenyl)picolinate To a solution of 6-(3-bromo-2-methylphenoxy)hexanal (446.94 mg, 1.57 mmol, 1.2 equiv.) in 1,4-dioxane (8 mL) was added KF (1.5 M, 2.61 mL, 3 equiv.), [2-(2- aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-butyl-phos phane;methanesulfonate (95.11 mg, 130.60 µmol, 0.1 equiv.), and tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)picolinate (800 mg, 1.31 mmol, 1 equiv.). The mixture was stirred at 100 °C for 1 hour under N 2 . Water (50 mL) was added, and the mixture was extracted with DCM 60 (30 mL × 2), dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum. The residue was purified by flash silica gel chromatography (Eluent of 0~30% ethyl acetate/petroleum ether) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2-methyl-3-((6- oxohexyl)oxy)phenyl)picolinate (220 mg, 287.8 µmol, 22.0% yield, 90.3% purity) as a yellow solid. MS (ESI) m/z: 691.4 [M+H] + Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-((6-(4-(3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)hexyl)oxy)-2-methylphenyl)picoli nate To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2-methyl-3-((6-oxohexyl)oxy)phenyl)picolinate (90 mg, 130.27 µmol, 1 equiv.) in DCM (1 mL) was added NMM (13.18 mg, 130.27 µmol, 14.32 µL, 1 equiv.) and 3-(1- methyl-6-(piperazin-1-yl)-1H-indazol-3-yl)piperidine-2,6-dio ne (46.91 mg, 143.30 µmol, 1.1 equiv.). The mixture was stirred at 25 °C for 15 hours. Then NaBH3CN (24.56 mg, 390.82 µmol, 3 equiv.) was added into the mixture and stirred at 25 °C for 1 hour. Water (50 mL) was added, and layers were separated. The organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum. The residue was purified by prep-TLC (SiO2, DCM:MeOH = 10:1) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-((6-(4-(3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H-indazol- 6-yl)piperazin-1-yl)hexyl)oxy)-2-methylphenyl)picolinate (125 mg, 124.7 µmol, 95.7% yield) as a yellow oil. MS (ESI) m/z: 1002.5 [M+H] + Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-((6-(4-(3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)hexyl)oxy)-2-methylphenyl)picoli nic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-((6-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1- yl)hexyl)oxy)-2-methylphenyl)picolinate (125 mg, 124.72 µmol, 1 equiv.) in DCM (2 mL) was added TFA (1.54 g, 13.47 mmol, 1 mL, 114.3 equiv.). The mixture was stirred at 40 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-((6-(4-(3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H-indazol- 6-yl)piperazin-1-yl)hexyl)oxy)-2-methylphenyl)picolinic acid (37.0 mg, 38.0 µmol, 30.5% yield, 97.1% purity) as a yellow solid. MS (ESI) m/z: 947.1 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 13.02 - 12.73 (m, 1H), 10.85 (s, 1H), 9.58 - 9.35 (m, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.56 (d, J = 8.8 Hz, 1H), 7.49 - 7.42 (m, 3H), 7.40 - 7.32 (m, 2H), 7.10 (t, J = 8.0 Hz, 1H), 6.99 - 6.94 (m, 3H), 6.89 (d, J = 8.4 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.30 - 4.25 (m, 1H), 3.98 (t, J = 6.0 Hz, 2H), 3.91 (s, 3H), 3.60 (d, J = 10.4 Hz, 6H), 3.18 (d, J = 10.0 Hz, 4H), 3.07 - 2.99 (m, 4H), 2.65 - 2.58 (m, 2H), 2.35 - 2.28 (m, 1H), 2.19 - 2.15 (m, 1H), 1.90 (s, 3H), 1.80 - 1.69 (m, 4H), 1.55 - 1.46 (m, 2H), 1.44 - 1.34 (m, 2H) Example 166. Preparation of Compound 224b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(((1R,3r,5S)- 8-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl) amino)-2-oxoethyl)-8- azabicyclo[3.2.1]octan-3-yl)oxy)ethoxy)-2-methylphenyl)picol inic acid Step A. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[[(1R,5S)-8-[2-[[3- (2,6-dioxo-3-piperidyl)-1- methyl-indazol-7-yl]amino]-2-oxo-ethyl]-8-azabicyclo[3.2.1]o ctan-3-yl]oxy]ethoxy]-2- methyl-phenyl]pyridine-2-carboxylate To a solution of 2-[(1R,5S)-3-[2-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3, 4-dihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]ethoxy]-8- azabicyclo[3.2.1]octan-8-yl]acetic acid (170 mg, 211.45 µmol, 1.0 equiv.) and 3-(7-amino-1- methyl-indazol-3-yl)piperidine-2,6-dione (60.07 mg, 232.60 µmol, 1.1 equiv.) in DMF (2 mL) was added HATU (104.52 mg, 274.89 µmol, 1.3 equiv.) and DIPEA (81.98 mg, 634.36 µmol, 110.49 µL, 3.0 equiv.).The mixture was stirred at 25 °C for 16 hours. The reaction mixture was added to H 2 O (5.0 ml). The mixture was triturated with H 2 O (5.0 mL) at 25 °C for 30 min and filtered to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[2-[[(1R,5S)-8-[2-[[3-(2,6-dioxo-3-piperidyl)-1-m ethyl-indazol-7-yl]amino]-2-oxo- ethyl]-8-azabicyclo[3.2.1]octan-3-yl]oxy]ethoxy]-2-methyl-ph enyl]pyridine-2-carboxylate (200 mg, 191.5 µmol, 90.5% yield) as a brown solid. MS (ESI) m/z: 1044.6 [M+H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(((1R,3r,5S)-8-(2-((3-( 2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)amino)-2-oxoethyl)-8-azabicyclo[3.2.1 ]octan-3-yl)oxy)ethoxy)-2- methylphenyl)picolinic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[2-[[(1R,5S)-8-[2-[[3-(2,6-dioxo-3-piperidyl)-1-met hyl-indazol-7-yl]amino]-2-oxo- ethyl]-8-azabicyclo[3.2.1]octan-3-yl]oxy]ethoxy]-2-methyl-ph enyl]pyridine-2-carboxylate (200 mg, 191.53 µmol, 1.0 equiv.) in DCM (1 mL) and TFA (1 mL) was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to remove DCM. The resulting residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(((1R,3r,5S)-8-(2-( (3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)amino)-2-oxoethyl)-8-azabicyclo[3.2.1 ]octan-3-yl)oxy)ethoxy)-2- methylphenyl)picolinic acid (31.4 mg, 29.1 µmol, 15.1% yield, 91.3% purity) as a yellow solid. MS (ESI) m/z: 988.4 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 13.01 - 12.74 (m, 1H), 12.69 - 12.21 (m, 1H), 10.90 (s, 1H), 10.06 (d, J = 8.4 Hz, 1H), 8.13 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.4 Hz, 1H), 7.60 (dd, J = 7.6, 14.0 Hz, 2H), 7.49 - 7.43 (m, 3H), 7.40 - 7.29 (m, 3H), 7.13 - 7.06 (m, 2H), 6.97 (d, J = 8.8 Hz, 1H), 6.92 (d, J = 8.4 Hz, 1H), 6.65 (d, J = 8.0 Hz, 1H), 4.98 (s, 2H), 4.38 (dd, J = 4.8, 10.4 Hz, 1H), 4.13 - 4.07 (m, 5H), 3.92 (t, J = 6.0 Hz, 2H), 3.71 (s, 2H), 3.64 - 3.60 (m, 1H), 3.44 - 3.40 (m, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.65 - 2.55 (m, 5H), 2.37 (s, 1H), 2.20 - 2.04 (m, 6H), 1.92 (s, 3H), 1.89 - 1.82 (m, 2H). Example 167. Preparation of Compound 225 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-((7-(2-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin -1-yl)ethyl)-7- azaspiro[3.5]nonan-2-yl)methoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-((7-(2-(4-(3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)ethyl)-7-azaspiro[3.5]nonan-2-yl )methoxy)-2- methylphenyl)picolinic acid A mixture of 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (27.44 mg, 83.82 µmol, 1.2 equiv.), NaBH(OAc) 3 (44.41 mg, 209.54 µmol, 3.0 equiv.), NMM (7.06 mg, 69.85 µmol, 7.68 µL, 1.0 equiv.) in DCM (0.5 mL) and isopropanol (0.5 mL) was stirred at 0 °C for 5 min.6-[8-(1,3-Benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-iso quinolin-2-yl]-3-[2-methyl- 3-[[7-(2-oxoethyl)-7-azaspiro[3.5]nonan-2-yl]methoxy]phenyl] pyridine-2-carboxylic acid (50 mg, 69.85 µmol, 1.0 equiv.) was then added to the mixture, which was stirred at 0 °C for 30 min. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep–HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-((7-(2-(4-(3-(2,6-dioxopiperidin-3-yl)-1-meth yl-1H-indazol-6-yl)piperazin-1- yl)ethyl)-7-azaspiro[3.5]nonan-2-yl)methoxy)-2-methylphenyl) picolinic acid (21.1 mg, 19.9 µmol, 9.5% yield, 96.8% purity) as a white solid MS (ESI) m/z: 1027.4 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 10.84 (s, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.50 - 7.33 (m, 6H), 7.07 (t, J = 8.0 Hz, 1H), 6.93 - 6.81 (m, 4H), 6.65 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.25 (dd, J = 5.2, 9.2 Hz, 1H), 3.92 - 3.87 (m, 7H), 3.21 - 3.17 (m, 6H), 3.02 - 3.0 (m, 4H), 2.65 - 2.54 (m, 8H), 2.38 - 2.24 (m, 4H), 2.19 - 2.12 (m, 1H), 1.92 - 1.84 (m, 5H), 1.67 - 1.57 (m, 4H), 1.50 - 1.45 (m, 2H) Example 168. Preparation of Compound 227a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1-yl)-4- oxobutyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-4-oxobutyl)cyclohexyl )oxy)-2- methylphenyl)picolinate To a solution of 4-((1r,4s)-4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)cyclohexyl)butanoic acid (100 mg, 131.42 µmol, 1 equiv.) in pyridine (1 mL) was added EDCI (32.75 mg, 170.84 µmol, 1.3 equiv.) and 3-(1-methyl-6-piperazin-1-yl- indazol-3-yl)piperidine-2,6-dione (51.63 mg, 157.70 µmol, 1.2 equiv.). The mixture was stirred at 60 °C for 12 hours. The reaction was diluted with water (20 mL) and extracted with DCM (20 mL × 2). The combined organic layers were washed with brine (30 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2), to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1-yl)-4-oxobutyl)cyclohexyl)oxy)-2 -methylphenyl)picolinate (100 mg, 93.4 µmol, 71.1% yield) as a yellow solid MS (ESI) m/z: 1170.8 [M+H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-4-oxobutyl)cyclohexyl )oxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-6- yl)piperazin-1-yl)-4-oxobutyl)cyclohexyl)oxy)-2-methylphenyl )picolinate (100 mg, 93.43 µmol, 1 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 144.56 equiv.). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1s,4r)-4-(4-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin -1-yl)-4- oxobutyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (28.6 mg, 27.2 µmol, 29.1% yield, 96.4% purity) as a yellow solid. MS (ESI) m/z: 1015.0 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.87 (s, 1H), 12.69 - 12.43 (m, 1H), 10.86 (s, 1H), 8.04 (d, J = 8.0 Hz, 1H), 7.80 (d, J = 7.6 Hz, 1H), 7.63 (d, J = 6.4 Hz, 1H), 7.53 (d, J = 8.8 Hz, 1H), 7.50 - 7.43 (m, 3H), 7.40 - 7.33 (m, 2H), 7.11 - 7.05 (m, 1H), 6.98 - 6.88 (m, 4H), 6.61 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.30 - 4.25 (m, 1H), 4.23 - 4.17 (m, 1H), 3.92 (s, 2H), 3.90 (s, 3H), 3.63 (s, 4H), 3.24 (s, 2H), 3.18 (d, J = 1.2 Hz, 2H), 3.05 - 3.01 (m, 2H), 2.63 - 2.59 (m, 2H), 2.38 - 2.33 (m, 2H), 2.19 - 2.13 (m, 1H), 2.11 - 2.05 (m, 2H), 1.87 (s, 3H), 1.82 - 1.75 (m, 2H), 1.59 - 1.52 (m, 2H), 1.40 - 1.21 (m, 6H), 1.11 - 1.01 (m, 2H) Example 169. Preparation of Compound 230 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin -1-yl)ethyl)piperidin-4- yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 4-[3-(3-bromo-2-methyl- phenoxy)propyl]piperidine-1-carboxylate To a solution of 3-bromo-2-methyl-phenol (1 g, 5.35 mmol, 1 equiv.) and tert-butyl 4-(3- bromopropyl)piperidine-1-carboxylate (1.64 g, 5.35 mmol, 1 equiv.) in CH3CN (4 mL) was added K2CO3 (2.22 g, 16.04 mmol, 3 equiv.). The mixture was stirred at 60 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=1/0 to 10/1) to give tert-butyl 4-[3-(3-bromo-2-methyl-phenoxy)propyl]piperidine-1-carboxyla te (2.2 g, 5.3 mmol, 99.7% yield) as a colorless oil. MS (ESI) m/z: 312.4 [M+H] + 1 H NMR (400 MHz, CDCl 3 ) δ = 7.14 (d, J = 8.0 Hz, 1H), 6.99 (t, J = 8.0 Hz, 1H), 6.76 (d, J = 8.4 Hz, 1H), 4.14 - 4.06 (m, 2H), 3.94 (t, J = 6.4 Hz, 2H), 2.69 (t, J = 2.4, 12.8 Hz, 2H), 2.32 (s, 3H), 1.88 - 1.79 (m, 2H), 1.70 (d, J = 12.8 Hz, 2H), 1.48 - 1.43 (m, 12H), 1.18 - 1.08 (m, 2H) Step B. Procedure for preparation of 4-[3-(3-bromo-2-methyl-phenoxy)propyl]piperidine To a solution of tert-butyl 4-[3-(3-bromo-2-methyl-phenoxy)propyl]piperidine-1-carboxyla te (2.2 g, 5.34 mmol, 1 equiv.) in HCl/EtOAc (10 mL). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue to give 4-[3-(3-bromo-2-methyl-phenoxy)propyl]piperidine (1.6 g, 5.1 mmol, 96.0% yield) as a white solid. MS (ESI) m/z: 314.4 [M+H] + 1 HNMR (400 MHz, CDCl3) δ = 7.14 (d, J = 8.0 Hz, 1H), 6.99 (t, J = 8.0 Hz, 1H), 6.76 (d, J = 8.4 Hz, 1H), 4.14 - 4.06 (m, 2H), 3.94 (t, J = 6.4 Hz, 2H), 2.69 (t, J = 2.4, 12.8 Hz, 2H), 2.32 (s, 3H), 1.88 - 1.79 (m, 2H), 1.70 (d, J = 12.8 Hz, 2H), 1.48 - 1.43 (m, 12H), 1.18 - 1.08 (m, 2H) Step C. Procedure for preparation of 4-[3-(3-bromo-2-methyl-phenoxy)propyl]-1-(2,2- diethoxyethyl)piperidine To a solution of 4-[3-(3-bromo-2-methyl-phenoxy)propyl]piperidine (1.5 g, 4.80 mmol, 1 equiv.) 2-bromo-1,1-diethoxy-ethane (946.71 mg, 4.80 mmol, 722.68 µL, 1 equiv.) in CH 3 CN (5 mL) was added K2CO3 (1.99 g, 14.41 mmol, 3 equiv.) and KI (797.46 mg, 4.80 mmol, 1 equiv.). The mixture was stirred at 80 °C for 8 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, ethyl acetate/MeOH=1/0 to 20/1) to give 4-[3-(3-bromo-2-methyl-phenoxy)propyl]-1- (2,2-diethoxyethyl)piperidine (1.7 g, 3.9 mmol, 82.6% yield) as a yellow oil. MS (ESI) m/z: 430.5 [M+H] + Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2,2-diethoxyethyl)p iperidin-4-yl)propoxy)-2- methylphenyl)picolinate A mixture of 4-[3-(3-bromo-2-methyl-phenoxy)propyl]-1-(2,2-diethoxyethyl) piperidine (800 mg, 1.87 mmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (1.14 g, 1.87 mmol, 1 equiv.), [2-(2-aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-buty l- phosphane;methanesulfonate (136.00 mg, 186.74 µmol, 0.1 equiv.), and KF (1.5 M, 3.73 mL, 3 equiv.) in dioxane (5 mL) and H2O (1mL) was degassed and purged with N2 three times, and then the mixture was stirred at 100 °C for 1 hour under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, ethyl acetate/MeOH=1/0 to 10/1) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-(1-(2,2- diethoxyethyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolin ate (1.5 g, 1.7 mmol, 91.9% yield, 95.5% purity) as a yellow oil. MS (ESI) m/z: 834.8 [M+H] + 1 H NMR (400 MHz, CDCl 3 ) δ = 7.85 (d, J = 8.0 Hz, 1H), 7.59 (dd, J = 7.2, 15.6 Hz, 2H), 7.40 - 7.30 (m, 5H), 7.14 - 7.06 (m, 1H), 6.90 (d, J = 8.8 Hz, 1H), 6.80 (d, J = 8.4 Hz, 1H), 6.69 (d, J = 7.6 Hz, 1H), 5.12 - 4.95 (m, 2H), 4.81 - 4.60 (m, 1H), 4.12 - 4.08 (m, 2H), 3.97 (t, J = 6.4 Hz, 2H), 3.72 - 3.68 (m, 2H), 3.61 - 3.54 (m, 2H), 3.07 (t, J = 5.2 Hz, 4H), 2.64 - 2.52 (m, 2H), 2.05 (s, 3H), 1.84 - 1.80 (m, 2H), 1.69 - 1.55 (m, 9H), 1.24 - 1.20 (m, 6H), 1.15 (s, 9H). Step E. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(2-methyl-3-(3-(1-(2-oxoethyl )piperidin-4- yl)propoxy)phenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2,2-diethoxyethyl)piperidin-4-yl)propo xy)-2-methylphenyl)picolinate (200 mg, 239.79 µmol, 1.0 equiv.) was added HCOOH (11.52 mg, 239.79 µmol, 2 mL, 1.0 equiv.). The mixture was stirred at 90 °C for 1.5 hours. The reaction mixture was filtered and concentrated under reduced pressure to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(2-methyl-3-(3-(1-(2-oxoethyl )piperidin-4- yl)propoxy)phenyl)picolinic acid (100 mg, crude) as a black oil, which was used in the next step without further purification. MS (ESI) m/z: 704.3 [M+H] + . Step F. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-(4-(3-(2,6-dioxop iperidin-3-yl)-1-methyl-1H- indazol-7-yl)piperazin-1-yl)ethyl)piperidin-4-yl)propoxy)-2- methylphenyl)picolinic acid To a solution of 3-(1-methyl-7-(piperazin-1-yl)-1H-indazol-3-yl)piperidine-2, 6-dione (46.51 mg, 142.08 µmol, 1.0 equiv.) and NaBH(OAc)3 (90.34 mg, 426.23 µmol, 3.0 equiv.) in DCM (1 mL) and IPA (1 mL) was added 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(2-methyl-3-(3-(1-(2-oxoethyl )piperidin-4- yl)propoxy)phenyl)picolinic acid (100 mg, 142.08 µmol, 1.0 equiv.) at 0 °C. The mixture was stirred at 0 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-(1-(2-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin-1-yl )ethyl)piperidin-4-yl)propoxy)- 2-methylphenyl)picolinic acid (19.3 mg, 16.8 µmol, 11.8% yield, 88.2% purity) as a yellow solid. MS (ESI) m/z: 1015.4 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.87 (s, 1H), 8.00 (d, J = 7.6 Hz, 1H), 7.76 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.46 - 7.29 (m, 6H), 7.10 - 6.99 (m, 3H), 6.85 (d, J = 8.0 Hz, 2H), 6.65 (d, J = 7.6 Hz, 1H), 4.96 (s, 2H), 4.34 - 4.31 (m, 1H), 4.20 (s, 3H), 3.99 - 3.93 (m, 2H), 3.88 - 3.87 (m, 2H), 2.99 - 2.91 (m, 6H), 2.69 - 2.59 (m, 4H), 2.53 (s, 4H), 2.34 - 2.28 (m, 2H), 2.21 - 2.10 (m, 2H), 2.10 - 1.95 (m, 3H), 1.90 (s, 3H), 1.78 - 1.66 (m, 3H), 1.65 - 1.54 (m, 2H), 1.37 - 1.21 (m, 5H). Example 170. Preparation of Compound 232 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[2-[1-[2-[4-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]piperazin-1-yl ]ethyl]-4-piperidyl]ethoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[2-[1-[2-[4-[3-(2,6-dioxo-3-piperi dyl)-1-methyl-indazol-6- yl]piperazin-1-yl]ethyl]-4-piperidyl]ethoxy]-2-methyl-phenyl ]pyridine-2-carboxylic acid To a solution of 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (72.61 mg, 221.80 µmol, 0.9 equiv.) and NaBH(OAc)3 (156.69 mg, 739.32 µmol, 3.0 equiv.) in DCM (1.5 mL) and IPA (1.5 mL) was added 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[2-methyl-3-[2-[1-(2-oxoethyl)-4-piperid yl]ethoxy]phenyl]pyridine-2- carboxylic acid (170 mg, 246.44 µmol, 1.0 equiv.) at 0 °C slowly. The mixture was stirred at 0 °C for 1 hour. Then the mixture was warmed to 25 °C and stirred at 25 °C for 12 hours. The mixture was concentrated under reduced pressure to remove DCM and IPA and diluted with DMF (3 mL). The resulting mixture was purified by prep-HPLC to give 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[2-[1-[2-[4-[3-(2,6- dioxo-3-piperidyl)-1-methyl-indazol-6-yl]piperazin-1-yl]ethy l]-4-piperidyl]ethoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid (19.3 mg, 17.4 µmol, 7.0% yield, 94.2% purity) as a yellow solid. MS (ESI) m/z: 1001.5 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.84 (s, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.52 - 7.29 (m, 6H), 7.11 - 7.04 (m, 1H), 6.95 - 6.85 (m, 3H), 6.82 (s, 1H), 6.64 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.28 - 4.22 (m, 1H), 3.98 (d, J = 5.2 Hz, 2H), 3.93 - 3.86 (m, 5H), 3.22 - 2.17 (m, 4H), 3.01 (t, J = 5.6 Hz, 2H), 2.96 (d, J = 10.0 Hz, 2H), 2.65 - 2.59 (m, 2H), 2.59 - 2.52 (m, 7H), 2.35 - 2.24 (m, 2H), 2.18 - 2.12 (m, 1H), 2.12 - 2.04 (m, 2H), 1.90 (s, 3H), 1.72 - 1.63 (m, 4H), 1.55 - 1.47 (m, 1H), 1.27 - 1.17 (m, 2H) Example 171. Preparation of Compound 233 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[2-[1-[2-[4-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]piperazin-1-yl ]ethyl]-4-piperidyl]ethoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[2-[1-[2-[4-[3-(2,6-dioxo-3-piperi dyl)-1-methyl-indazol-7- yl]piperazin-1-yl]ethyl]-4-piperidyl]ethoxy]-2-methyl-phenyl ]pyridine-2-carboxylic acid To a solution of 3-(1-methyl-7-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (46.98 mg, 143.52 µmol, 1.1 equiv.) and NaBH(OAc) 3 (82.95 mg, 391.41 µmol, 3 equiv.) in DCM (1.5 mL) and IPA (1.5 mL) was added 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[2-methyl-3-[2-[1-(2-oxoethyl)-4-piperid yl]ethoxy]phenyl]pyridine-2- carboxylic acid (90 mg, 130.47 µmol, 1 equiv.) at 0 °C slowly. The mixture was stirred at 0 °C for 1 hour. Then the mixture was warmed to 25 °C and stirred at 25 °C for 12 hours. The mixture was concentrated under reduced pressure to remove DCM and IPA and diluted with DMF (2 mL). The resulting mixture was purified by prep-HPLC to give 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[2-[1-[2-[4-[3-(2,6- dioxo-3-piperidyl)-1-methyl-indazol-7-yl]piperazin-1-yl]ethy l]-4-piperidyl]ethoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid (12.5 mg, 11.8 µmol, 9.0% yield, 98.3% purity) as a yellow solid. MS (ESI) m/z: 1001.7 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.89 (s, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.48 - 7.40 (m, 3H), 7.40 - 7.32 (m, 3H), 7.11 - 7.06 (m, 1H), 7.04 - 6.99 (m, 2H), 6.96 - 6.86 (m, 2H), 6.64 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.36 - 4.31 (m, 1H), 4.23 (s, 3H), 3.99 (d, J = 5.2 Hz, 2H), 3.91 (t, J = 6.0 Hz, 2H), 3.15 - 3.07 (m, 5H), 3.03 - 3.00 (m, 2H), 2.76 - 2.57 (m, 10H), 2.38 - 2.28 (m, 4H), 2.20 - 2.13 (m, 1H), 1.90 (s, 3H), 1.78 - 1.66 (m, 4H), 1.63 - 1.55 (m, 1H), 1.36 - 1.25 (m, 2H) Example 172. Preparation of Compound 234 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[3-[4-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]piperazin-1-yl ]-2,2-difluoro-propyl]phenoxy]- 2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 3-(6-(4-(3-(4-(3-bromo-2-methylphenoxy)phenyl)- 2,2-difluoropropyl)piperazin-1-yl)-1-methyl-1H-indazol-3-yl) piperidine-2,6-dione A mixture of 3-(4-(3-bromo-2-methylphenoxy)phenyl)-2,2-difluoropropyl trifluoromethanesulfonate (120 mg, 245.27 µmol, 1 equiv.), 3-(1-methyl-6-piperazin-1-yl- indazol-3-yl)piperidine-2,6-dione (64.24 mg, 196.22 µmol, 0.8 equiv.) and K 2 CO 3 (84.75 mg, 613.18 µmol, 2.5 equiv.) in CH3CN (1.5 mL) was degassed and purged with N2 and stirred at 50 °C for 10 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 50% ethyl acetate : petroleum ether) to give 3-(6-(4-(3-(4-(3-bromo-2- methylphenoxy)phenyl)-2,2-difluoropropyl)piperazin-1-yl)-1-m ethyl-1H-indazol-3- yl)piperidine-2,6-dione (80 mg, 120.0 µmol, 48.9% yield) as a colorless oil. MS (ESI) m/z: 668.2 [M+H] + . Step B. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo -3-piperidyl)-1-methyl- indazol-6-yl]piperazin-1-yl]-2,2-difluoro-propyl]phenoxy]-2- methyl-phenyl]pyridine-2- carboxylate A mixture of 3-(6-(4-(3-(4-(3-bromo-2-methylphenoxy)phenyl)-2,2-difluorop ropyl)piperazin- 1-yl)-1-methyl-1H-indazol-3-yl)piperidine-2,6-dione (80 mg, 120.02 µmol, 1 equiv.), tert- butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-2-carboxylate (102.92 mg, 168.03 µmol, 1.4 equiv.), [2-(2-aminophenyl)phenyl]palladium(1 + );bis(1-adamantyl)-butyl- phosphane;methanesulfonate (17.48 mg, 24.00 µmol, 0.2 equiv.), and KF (1.5 M, 120.02 µL, 1.5 equiv.) in dioxane (0.1 mL) was degassed and purged with N2 and stirred at 100 °C for 1 hour under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0 ~ 50% ethyl acetate / petroleum ether) to give tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[4- [3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]piperazin-1-yl]-2,2-difluoro-propyl]phen oxy]-2-methyl-phenyl]pyridine- 2-carboxylate (110 mg, 102.5 µmol, 85.4% yield) as a colorless oil. MS (ESI) m/z: 1072.8 [M+H] + . Step C. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo-3-piperidyl )-1-methyl-indazol-6- yl]piperazin-1-yl]-2,2-difluoro-propyl]phenoxy]-2-methyl-phe nyl]pyridine-2-carboxylic acid tert-Butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4- [3-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]piper azin-1-yl]-2,2-difluoro- propyl]phenoxy]-2-methyl-phenyl]pyridine-2-carboxylate (110 mg, 102.59 µmol, 1 equiv.) in TFA (0.5 mL) and DCM (0.5 mL) was stirred at 25 °C for 10 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[3- [4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]piperazi n-1-yl]-2,2-difluoro- propyl]phenoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid (39.5 mg, 37.8 µmol, 36.9% yield, 97.4% purity) as an off-white solid MS (ESI) m/z: 1016.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.98 - 12.47 (m, 2H), 10.85 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 7.6 Hz, 1H), 7.55 - 7.43 (m, 4H), 7.36 (td, J = 7.6, 11.6 Hz, 2H), 7.27 (d, J = 8.4 Hz, 2H), 7.22 - 7.16 (m, 1H), 7.01 (d, J = 8.8 Hz, 1H), 6.95 - 6.82 (m, 6H), 5.00 (s, 2H), 4.26 (dd, J = 5.2, 9.2 Hz, 1H), 3.96 - 3.90 (m, 2H), 3.89 (s, 3H), 3.30 - 3.19 (m, 6H), 3.10 - 3.00 (m, 2H), 2.77 - 2.54 (m, 8H), 2.32 - 2.13 (m, 2H), 1.89 (s, 3H) F NMR (400 MHz, DMSO-d6) δ = -96.27 Example 173. Preparation of Compound 238 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-((1-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2- oxoethyl)piperidin-4- yl)oxy)ethoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((1-(2-((3-(2,6-dioxopi peridin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)oxy)ethoxy)-2- methylphenyl)picolinate A mixture of 2-(4-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)eth oxy)piperidin-1-yl)acetic acid (110 mg, 141.40 µmol, 1 equiv.) and 3-(6-amino-1-methyl-1H-indazol-3-yl)piperidine-2,6- dione (43.82 mg, 169.68 µmol, 1.2 equiv.) in pyridine (1.1 mL) was added EDCI (40.66 mg, 212.10 µmol, 1.5 equiv.), and then the mixture was stirred at 25 °C for 20 hours. The reaction mixture was partitioned between H 2 O (3 mL) and ethyl acetate (8 mL). The organic phase was separated, washed with sat. NaCl aq. (2 mL × 3), dried over (Na2SO4), filtered, and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)oxy)ethoxy)-2- methylphenyl)picolinate (163 mg, crude) as a red solid. MS (ESI) m/z: 1018.7 [M+H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((1-(2-((3-(2,6-dioxopi peridin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)oxy)ethoxy)-2- methylphenyl)picolinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(2-((1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H -indazol-6-yl)amino)-2- oxoethyl)piperidin-4-yl)oxy)ethoxy)-2-methylphenyl)picolinat e (150 mg, 147.32 µmol, 1 equiv.) in TFA (0.5 mL) and DCM (1.5 mL) was stirred at 25 °C for 18 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(2-((1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H -indazol-6-yl)amino)-2- oxoethyl)piperidin-4-yl)oxy)ethoxy)-2-methylphenyl)picolinic acid (54.21 mg, 55.5 µmol, 37.7% yield, 98.5% purity) as a pink solid. MS (ESI) m/z: 963.8 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 13.12 - 12.53 (m, 2H), 10.88 (s, 1H), 10.13 - 9.65 (m, 1H), 8.13 (s, 1H), 8.02 (d, J = 4.0 Hz, 2H), 7.81 - 7.76 (m, 1H), 7.62 (t, J = 7.6 Hz, 2H), 7.49 - 7.43 (m, 3H), 7.40 - 7.32 (m, 2H), 7.20 (d, J = 8.4 Hz, 1H), 7.10 (t, J = 8.0 Hz 1H), 6.97 (d, J = 8.4 Hz, 1H), 6.91 (d, J = 8.0 Hz, 1H), 6.65 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.32 (dd, J = 4.8, 9.6 Hz, 1H), 4.09 (s, J = 3.2 Hz, 2H), 3.92 (s, 5H), 3.79 (t, J = 4.0 Hz, 2H), 3.63 - 3.43 (m, 3H), 3.03 (t, J = 5.6 Hz, 2H), 2.85 - 2.72 (m, 2H), 2.67 - 2.60 (m, 3H), 2.36 - 2.28 (m, 2H), 2.19 - 2.13 (m, 1H), 1.91 (s, 5H), 1.74 - 1.54 (m, 2H) Example 174. Preparation of Compound 243 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(3-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino)-3-oxopr opyl)phenoxy)-2- methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(3-((3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H- indazol-7-yl)amino)-3-oxopropyl)phenoxy)-2-methylphenyl)pico linate To a solution of 3-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroiso quinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)phe nyl)propanoic acid (90 mg, 121.48 µmol, 1 equiv.) in DMF (1 mL) was added HATU (78.52 mg, 206.52 µmol, 1.7 equiv.), DIEA (47.10 mg, 364.44 µmol, 63.48 µL, 3 equiv.), and 3-(7-amino-1-methyl-1H-indazol-3- yl)piperidine-2,6-dione (34.51 mg, 133.63 µmol, 1.1 equiv.). The mixture was stirred at 40 °C for 1 hour. The reaction was added to H2O (5 mL) and filtered to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(2-(3-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino)-3-oxopr opyl)phenoxy)-2- methylphenyl)picolinate (103 mg, crude) as a yellow solid. MS (ESI) m/z: 981.9 [M+H] + Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(3-((3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H- indazol-7-yl)amino)-3-oxopropyl)phenoxy)-2-methylphenyl)pico linic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(3-((3-(2,6-dioxopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-3- oxopropyl)phenoxy)-2-methylphenyl)picolinate (103 mg, 104.98 µmol, 1 equiv.) in DCM (0.5 mL) was added TFA (770.00 mg, 6.75 mmol, 0.5 mL, 64.33 equiv.). The mixture was stirred at 25 °C for 12 hours. The mixture was concentrated under reduced pressure to remove DCM and diluted with DMF (2 mL). The resulting mixture was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(2-(3-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino)-3-oxopr opyl)phenoxy)-2- methylphenyl)picolinic acid (26.4 mg, 27.6 µmol, 26.3% yield, 96.7% purity) as a yellow solid. MS (ESI) m/z: 925.8 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 12.87 (s, 1H), 12.79 - 12.56 (m, 1H), 10.89 (s, 1H), 9.89 (s, 1H), 8.04 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 7.6 Hz, 1H), 7.62 (d, J = 6.8 Hz, 1H), 7.58 (d, J = 7.6 Hz, 1H), 7.54 (d, J = 8.8 Hz, 1H), 7.49 - 7.43 (m, 2H), 7.40 - 7.33 (m, 3H), 7.21 - 7.15 (m, 2H), 7.09 - 7.00 (m, 4H), 6.89 (d, J = 7.6 Hz, 1H), 6.83 (d, J = 8.0 Hz, 1H), 6.61 (d, J = 8.0 Hz, 1H), 5.00 (s, 2H), 4.39 - 4.32 (m, 1H), 3.94 (t, J = 5.2 Hz, 2H), 3.86 (s, 3H), 3.06 - 3.00 (m, 4H), 3.06 - 3.00 (m, 2H), 2.61 (d, J = 5.2 Hz, 2H), 2.57 (s, 1H), 2.19 - 2.12 (m, 1H), 2.00 - 1.94 (m, 3H) Example 175. Preparation of Compound 244 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(4-(3-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin -1-yl)-3-oxopropyl)phenoxy)- 2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(3-methoxy-3-oxopropyl) phenoxy)-2- methylphenyl)picolinate A mixture of methyl 3-(4-(3-bromo-2-methylphenoxy)phenyl)propanoate (300 mg, 859.06 µmol, 1 equiv.) and tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pic olinate (631.46 mg, 1.03 mmol, 1.2 equiv.), K2CO3 (1.5 M, 859.06 µL, 1.5 equiv.) in dioxane (3 mL) was added Ad2nBuP Pd G 3 (cataCXium® A Pd G 3 ) (125.13 mg, 171.81 µmol, 0.2 equiv.). The mixture was purged with N 2 and stirred at 80 °C for 3 hours under N 2 atmosphere. The reaction mixture was partitioned between H2O (4 mL) and ethyl acetate (4 mL). The organic phase was separated, washed with aqueous NaCl (1 mL × 3), dried over with Na2SO4, filtered, and concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (Eluent of 0~29% ethyl acetate/petroleum ether) to give tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(3-met hoxy-3-oxopropyl)phenoxy)- 2-methylphenyl)picolinate (377 mg, 464.4 µmol, 54.0% yield, 93% purity) as a yellow solid. MS (ESI) m/z: 755.4 [M+H] + . Step B. Procedure for preparation of 3-(4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)phenyl)propanoic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(4-(3-methoxy-3-oxopropyl)phenoxy)-2-methylphenyl)p icolinate (377 mg, 499.41 µmol, 1 equiv.) and LiOH•H2O (1 M, 1.50 mL, 3 equiv.) in THF (4 mL) and H2O (1 ml) was stirred at 25 °C for 2 hours. The mixture was concentrated and diluted with H2O (4 mL), and then the pH of the mixture was adjusted to 3 with 1 M HCl. Then the mixture was filtered, and the filtrate was concentrated to give 3-(4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)phenyl)propanoic acid (333 mg, crude) as a yellow solid. MS (ESI) m/z: 741.5 [M+H] + . Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(3-(4-(3-(2,6-dioxopipe ridin-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)-3-oxopropyl)phenoxy)-2-methylph enyl)picolinate To a mixture of 3-(4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroiso quinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)phe nyl)propanoic acid (90 mg, 121.48 µmol, 1 equiv.), HATU (69.29 mg, 182.22 µmol, 1.5 equiv.), and DIEA (62.80 mg, 485.92 µmol, 84.64 µL, 4 equiv.) in DMF (1.5 mL) was added 3-(1-methyl-6-(piperazin-1-yl)- 1H-indazol-3-yl)piperidin-2-one (47.72 mg, 145.78 µmol, 1.2 equiv.). The mixture was then stirred at 25 °C for 2 hours. To the reaction mixture was added water (5 mL), and the resulting mixture was filtered. The filter cake was washed with CH2Cl2 (5 mL) and CH3OH (5 mL) and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(3-(4-(3-(2,6-dioxo piperidin-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)-3-oxopropyl)phenoxy)-2-methylph enyl)picolinate (100 mg, crude) as a white solid. MS (ESI) m/z: 1050.7 [M+H] + . Step D. Procedure for preparation 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(3-(4-(3-(2,6-dioxopipe ridin-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)-3-oxopropyl)phenoxy)-2-methylph enyl)picolinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-6-yl)piperazin-1-yl)-3- oxopropyl)phenoxy)-2-methylphenyl)picolinate (100 mg, 95.22 µmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL) was stirred at 40 °C for 67 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin-1-yl )-3-oxopropyl)phenoxy)-2- methylphenyl)picolinic acid (39.5 mg, 38.7 µmol, 40.6% yield, 97.2% purity) as a white solid. MS (ESI) m/z: 994.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.86 (s, 1H), 12.74 - 12.57 (m, 1H), 10.84 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.4 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.52 (d, J = 8.8 Hz, 2H), 7.48 - 7.42 (m, 2H), 7.36 (td, J = 7.6, 10.8 Hz, 2H), 7.22 (d, J = 8.8 Hz, 2H), 7.11 (t, J = 7.6 Hz, 1H), 7.00 (d, J = 8.8 Hz, 1H), 6.92 (d, J = 9.2 Hz, 1H), 6.88 - 6.84 (m, 2H), 6.82 - 6.75 (m, 3H), 5.00 (s, 2H), 4.25 (dd, J = 5.2, 9.2 Hz, 1H), 3.93 (t, J = 5.6 Hz, 2H), 3.88 (s, 3H), 3.61 (d, J = 17.6 Hz, 4H), 3.14 (d, J = 4.0 Hz, 4H), 3.03 (t, J = 5.6 Hz, 2H), 2.85 - 2.78 (m, 2H), 2.69 - 2.65 (m, 2H), 2.60 (d, J = 5.6 Hz, 2H), 2.32 - 2.23 (m, 1H), 2.15 (s, 1H), 1.86 (s, 3H) Example 176. Preparation of Compound 247 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[1-[[1-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]-4-piperidyl]m ethyl]-4-piperidyl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[2-methyl-3-[3-(4-piperidyl)propoxy]p henyl]pyridine-2- carboxylic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[1-(2,2-diethoxyethyl)-4-piperidyl]propoxy]-2-me thyl-phenyl]pyridine-2- carboxylate (1 g, 1.20 mmol, 1.0 equiv.) in HCOOH (10 mL) was stirred at 100 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give 6-[8-(1,3-benzothiazol- 2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[2-methyl- 3-[3-(4- piperidyl)propoxy]phenyl]pyridine-2-carboxylic acid (1 g, crude) as a yellow oil, which was used in the next step without further purification. MS (ESI) m/z: 662.5 [M+H] + . Step B. Procedure for preparation of 3-[6-[4-(dimethoxymethyl)-1-piperidyl]-1-methyl- indazol-3-yl]piperidine-2,6-dione A mixture of 3-(2,6-dibenzyloxy-3-pyridyl)-7-[4-(dimethoxymethyl)-1-piper idyl]-1-methyl- indazole (800 mg, 1.38 mmol, 1.0 equiv.), Pd/C (500 mg, 138.24 µmol, 10% purity, 0.1 equiv.), Pd(OH) 2 (500 mg, 3.56 mmol, 2.58 equiv.) and in THF (5 mL) and EtOH (5 mL) was degassed, purged with H2 (50 Psi), and stirred at 25 °C for 12 hours under H2 (50 Psi) atmosphere. The reaction mixture was filtered, and the filter cake was immersed with MeOH (30ml). The filtrate was concentrated under reduced pressure to give 3-[6-[4-(dimethoxymethyl)-1-piperidyl]-1- methyl-indazol-3-yl]piperidine-2,6-dione (380 mg, 677.1 µmol, 48.9% yield, 71.3% purity) as a brown solid. MS (ESI) m/z: 401.1 [M+H] + . Step C. Procedure for preparation of 1-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6- yl]piperidine-4-carbaldehyde A mixture of 3-[6-[4-(dimethoxymethyl)-1-piperidyl]-1-methyl-indazol-3-yl ]piperidine-2,6- dione (100 mg, 249.71 µmol, 1.0 equiv.) in HCOOH (1 mL) was degassed and purged with N2 and stirred at 90 °C for 2 hours under N 2 atmosphere. The residue was concentrated to give 1- [3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]piperidine- 4-carbaldehyde (50 mg, crude) as a brown oil. MS (ESI) m/z: 387.1 [M+H] + . Step D. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[1-[[1-[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6-yl]-4- piperidyl]methyl]-4-piperidyl]propoxy]-2-methyl-phenyl]pyrid ine-2-carboxylic acid To a solution of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- [2-methyl-3-[3-(4-piperidyl)propoxy]phenyl]pyridine-2-carbox ylic acid (100 mg, 151.10 µmol, 1.0 equiv.) in DCM (2 mL) and IPA (2 mL) was added 1-[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]piperidine-4-carbaldehyde (64.26 mg, 181.32 µmol, 1.2 equiv.) at 25 °C for 16 hours. After addition, the mixture was stirred at this temperature, and then NaBH(OAc)3 (96.07 mg, 453.30 µmol, 3.0 equiv.) was added at 0 °C. The mixture was stirred at 25 °C for 1 hour, after which it was concentrated. The crude product was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[1-[[1-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]-4-piperidyl]m ethyl]-4-piperidyl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid (21.7 mg, 21.1 µmol, 14.0% yield, 97.2% purity) as a yellow solid. MS (ESI) m/z: 500.9 [M/2+H] + . 1H NMR (400 MHz, DMSO-d 6 ) δ = 10.84 (s, 1H), 8.18 (s, 1H), 8.01 - 8.01 (m, 1H), 8.01 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 8.0 Hz, 1H), 7.47 - 7.42 (m, 3H), 7.40 - 7.32 (m, 3H), 7.10 - 7.05 (m, 1H), 6.93 - 6.85 (m, 3H), 6.81 (s, 1H), 6.64 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.24 (dd, J = 5.2, 9.2 Hz, 1H), 3.92 (d, J = 5.6 Hz, 2H), 3.87 (s, 3H), 3.78 - 3.74 (m, 2H), 3.02 (t, J = 5.2 Hz, 2H), 2.92 - 2.80 (m, 4H), 2.74 - 2.69 (m, 2H), 2.68 - 2.64 (m, 1H), 2.63 - 2.55 (m, 2H), 2.30 - 2.23 (m, 2H), 2.19 - 2.12 (m, 4H), 1.90 (s, 3H), 1.79 - 1.73 (m, 4H), 1.70 - 1.59 (m, 3H), 1.39 - 1.34 (m, 2H), 1.29 - 1.19 (m, 4H) Example 177. Preparation of Compound 251a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(((1r,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1-yl)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of (1r,4r)-methyl 4-(4-bromo-3- methylphenoxy)cyclohexanecarboxylate To a solution of (1r,4r)-methyl 4-hydroxycyclohexanecarboxylate (2.0 g, 12.64 mmol, 1.1 equiv.) in toluene (20 mL) was added 2-(tributyl-λ5-phosphanylidene)acetonitrile (3.33 g, 13.79 mmol, 1.2 equiv.) and 4-bromo-3-methylphenol (2.15 g, 11.49 mmol, 1.0 equiv.). The mixture was stirred at 120 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=1/0) to give (1r,4r)-methyl 4-(4-bromo-3- methylphenoxy)cyclohexanecarboxylate (2.4 g, crude) as a yellow oil. Step B. Procedure for preparation of ((1r,4r)-4-(4-bromo-3- methylphenoxy)cyclohexyl)methanol To a solution of (1r,4r)-methyl 4-(4-bromo-3-methylphenoxy)cyclohexanecarboxylate (2.4 g, 7.33 mmol, 1 equiv.) in THF (25 mL) was added LAH (278.38 mg, 7.33 mmol, 1 equiv.). The mixture was stirred at 0 °C for 1 hour under N2 atmosphere. The reaction mixture was quenched by addition H2O 0.3 mL, 15% NaOH 0.3 mL and H2O 0.9 mL, the combined mixture was mixed with Na 2 SO 4 , then filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~20% ethyl acetate/petroleum ether) to give compound ((1r,4r)-4-(4-bromo-3- methylphenoxy)cyclohexyl)methanol (1.3 g, 4.3 mmol, 59.2% yield) as a yellow oil. 1 H NMR (400 MHz) δ = 7.40 (d, J = 8.8 Hz, 1H), 6.94 (d, J = 2.8 Hz, 1H), 6.75 - 6.67 (m, 1H), 4.43 (t, J = 5.6 Hz, 1H), 4.26 - 4.15 (m, 1H), 3.23 (t, J = 5.6 Hz, 2H), 2.28 (s, 3H), 2.08 - 2.00 (m, 2H), 1.77 (d, J = 11.6 Hz, 2H), 1.43 - 1.33 (m, 1H), 1.33 - 1.22 (m, 2H), 1.11 - 0.98 (m, 2H) Step C. Procedure for preparation of (1r,4r)-4-(4-bromo-3- methylphenoxy)cyclohexanecarbaldehyde To a solution of oxalyl dichloride (1.10 g, 8.69 mmol, 760.68 µL, 2 equiv.) in DCM (20 mL) was added dropwise a solution of DMSO (1.36 g, 17.38 mmol, 1.36 mL, 4 equiv.) in DCM (2 mL) at -70 °C under N 2 atmosphere. The mixture was stirred at -70 °C for 1 hour. After which time ((1r,4r)-4-(4-bromo-3-methylphenoxy)cyclohexyl)methanol (1.3 g, 4.34 mmol, 1 equiv.) in DCM (2 mL) was added dropwise at -70 °C. The solution was stirred for 1 hour at -70 °C. Then TEA (2.64 g, 26.07 mmol, 3.63 mL, 6 equiv.) was added into the solution. The solution was stirred at -70 °C for 0.5 hour under N 2 atmosphere. DCM (50 mL) and water (50 mL) were added, and the layers were separated. The aqueous layer was extracted with DCM (30 mL × 2). The combined organic extracts were dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum to give (1r,4r)-4-(4-bromo-3- methylphenoxy)cyclohexanecarbaldehyde (1.2 g, 4.04 mmol, 92.9% yield) as a yellow oil. Step D. Procedure for preparation of (E)-ethyl 3-((1r,4r)-4-(4-bromo-3- methylphenoxy)cyclohexyl)acrylate To a solution of ethyl 2-(diethoxyphosphoryl)acetate (1.81 g, 8.08 mmol, 1.60 mL, 2.0 equiv.) in THF (15 mL) was added NaH (339.15 mg, 8.48 mmol, 60% purity, 2.1 equiv.). The mixture was stirred at 0 °C for 1 hour. Then (1r,4r)-4-(4-bromo-3- methylphenoxy)cyclohexanecarbaldehyde (1.2 g, 4.04 mmol, 1 equiv.) was added into the mixture, which was stirred at 25 °C for 0.5 hour under N2. The reaction mixture was quenched by addition water (10 mL) at 0 °C, and then diluted with water (10 mL) and extracted with ethyl acetate (30 mL × 3). The combined organic layers were washed with water (10mL × 3), filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=0/1 to 10/1) to give (E)-ethyl 3-((1r,4r)-4-(4-bromo-3-methylphenoxy)cyclohexyl)acrylate (1 g, 2.7 mmol, 67.4% yield) as a white solid. 1 H NMR (400 MHz) δ = 7.41 (d, J = 8.8 Hz, 1H), 6.96 (d, J = 2.8 Hz, 1H), 6.76 - 6.70 (m, 1H), 6.16 - 6.07 (m, 1H), 5.74 (d, J = 11.6 Hz, 1H), 4.32 - 4.22 (m, 1H), 4.14 - 4.06 (m, 2H), 3.25 - 3.15 (m, 1H), 2.29 (s, 3H), 2.12 - 2.02 (m, 2H), 1.77 - 1.65 (m, 2H), 1.39 - 1.28 (m, 4H), 1.21 (t, J = 7.2 Hz, 3H) Step E. Procedure for preparation of ethyl 3-((1r,4r)-4-(4-bromo-3- methylphenoxy)cyclohexyl)propanoate To a solution of (E)-ethyl 3-((1r,4r)-4-(4-bromo-3-methylphenoxy)cyclohexyl)acrylate (1 g, 2.72 mmol, 1 equiv.) in EtOH (10 mL) was added PtO2 (61.83 mg, 272.27 µmol, 0.1 equiv.) under H 2 atmosphere (15 psi). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a filter cake which was rinsed with a little EtOH. The filter liquor was collected and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=20/1 to 10/1) to give ethyl 3-((1r,4r)-4-(4-bromo-3- methylphenoxy)cyclohexyl)propanoate (700 mg, 1.8 mmol, 65.2% yield, 93.7% purity) as a colorless oil. 1 H NMR (400 MHz) δ = 7.40 (d, J = 8.4 Hz, 1H), 6.94 (d, J = 2.8 Hz, 1H), 6.74 - 6.67 (m, 1H), 4.28 - 4.16 (m, 1H), 4.09 - 4.00 (m, 2H), 2.34 - 2.22 (m, 5H), 2.02 (d, J = 10.0 Hz, 2H), 1.74 (d, J = 12.0 Hz, 2H), 1.51 - 1.40 (m, 2H), 1.34 - 1.22 (m, 3H), 1.21 - 1.15 (m, 3H), 1.11 - 0.98 (m, 2H) Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(((1r,4r)-4-(3-ethoxy-3-ox opropyl)cyclohexyl)oxy)-2- methylphenyl)picolinate To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pic olinate (331.74 mg, 487.42 µmol, 90% purity, 1.2 equiv.) in dioxane (3 mL) was added ethyl 3-((1r,4r)-4-(4-bromo-3- methylphenoxy)cyclohexyl)propanoate (150 mg, 406.18 µmol, 1.0 equiv.), KF (1.5 M, 812.36 µL, 3.0 equiv.), and [2-(2-aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-buty l- phosphane;methanesulfonate (29.58 mg, 40.62 µmol, 0.1 equiv.). The mixture was stirred at 100 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by flash silica gel chromatography (Eluent of 0~30 % ethyl acetate/petroleum ether) to give compound tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(4-(((1r,4r)-4-(3-ethoxy- 3-oxopropyl)cyclohexyl)oxy)-2- methylphenyl)picolinate (360 mg, 371.6 µmol, 91.4% yield, 80% purity) as a yellow solid. MS (ESI) m/z: 775.5 [M+H] + Step G. Procedure for preparation of 3-((1r,4r)-4-(4-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-3- methylphenoxy)cyclohexyl)propanoic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(((1r,4r)-4-(3-ethoxy-3-oxopropyl)cyclohexyl) oxy)-2- methylphenyl)picolinate (360 mg, 371.63 µmol, 80% purity, 1.0 equiv.) in THF (4 mL) and H 2 O (1.3 mL) was added LiOH•H 2 O (46.78 mg, 1.11 mmol, 3.0 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove solvent, to the residue was added H2O (5 mL). The pH of the mixture was adjusted to 4 with 1M HCl. Then the reaction mixture was filtered and concentrated under reduced pressure to give 3-((1r,4r)-4-(4-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-di hydroisoquinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-3-methylphenoxy)cyc lohexyl)propanoic acid (300 mg, 313.2 µmol, 84.3% yield, 78% purity) as a yellow solid. MS (ESI) m/z: 747.2 [M+H] + Step H. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(((1r,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-3-oxopropyl)cyclohexy l)oxy)-2- methylphenyl)picolinate To a solution of 3-((1r,4r)-4-(4-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-3- methylphenoxy)cyclohexyl)propanoic acid (100 mg, 133.88 µmol, 1.0 equiv.) in pyridine (1 mL) was added EDCI (33.37 mg, 174.05 µmol, 1.3 equiv.) and 3-(1-methyl-6-(piperazin-1-yl)- 1H-indazol-3-yl)piperidine-2,6-dione (58.46 mg, 160.66 µmol, 1.2 equiv.). The mixture was stirred at 40 °C for 4 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=4/1 to 0/1, DCM : MeOH = 10:1) to give tert-butyl 6-(8-(benzo[d]thiazol- 2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(4-(((1r,4 r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin-1-yl )-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (90 mg, 85.2 µmol, 63.6% yield) as a yellow solid. MS (ESI) m/z: 1056.4 [M+H] + Step I. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(((1r,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-3-oxopropyl)cyclohexy l)oxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(((1r,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-6- yl)piperazin-1-yl)-3-oxopropyl)cyclohexyl)oxy)-2-methylpheny l)picolinate (90 mg, 85.20 µmol, 1.0 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 158.51 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(4-(((1r,4r)-4-(3-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin -1-yl)-3- oxopropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (20.34 mg, 18.6 µmol, 21.8% yield, 91.6% purity) as an off-white solid. MS (ESI) m/z: 1000.4 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.86 (s, 1H), 12.70 - 12.48 (m, 1H), 10.85 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 7.6 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.53 (d, J = 8.8 Hz, 1H), 7.49 - 7.42 (m, 3H), 7.39 - 7.32 (m, 2H), 6.95 (d, J = 8.8 Hz, 2H), 6.91 - 6.86 (m, 2H), 6.78 (d, J = 2.0 Hz, 1H), 6.73 - 6.68 (m, 1H), 4.97 (s, 2H), 4.29 - 4.25 (m, 1H), 4.24 - 4.17 (m, 1H), 3.93 - 3.86 (m, 5H), 3.63 (s, 2H), 3.23 (s, 2H), 3.18 (s, 2H), 3.02 (t, J = 6.0 Hz, 2H), 2.67 - 2.57 (m, 2H), 2.41 - 2.37 (m, 2H), 2.34 - 2.23 (m, 2H), 2.21 - 2.13 (m, 1H), 2.10 - 2.03 (m, 2H), 2.01 (s, 3H), 1.86 - 1.78 (m, 2H), 1.49 - 1.41 (m, 2H), 1.35 - 1.22 (m, 4H), 1.15 - 1.04 (m, 2H) Example 178. Preparation of Compound 256 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[1-[2-[4-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]-1-piperidyl]e thyl]-4-piperidyl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[2-methyl-3-[3-[1-(2-oxoethyl)-4- piperidyl]propoxy]phenyl]pyridine-2-carboxylic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[1-(2,2-diethoxyethyl)-4-piperidyl]propoxy]-2-me thyl-phenyl]pyridine-2- carboxylate (200 mg, 239.79 µmol, 1.0 equiv.) in HCOOH (2 mL) was stirred at 100 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[2-methyl-3-[3-[1-(2- oxoethyl)-4-piperidyl]propoxy]phenyl]pyridine-2-carboxylic acid (200 mg, crude) as a black oil. MS (ESI) m/z: 772.2 [M+H] + Step B. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[4-[3-(2,6-dioxo-3-piperi dyl)-1-methyl-indazol-7-yl]-1- piperidyl]ethyl]-4-piperidyl]propoxy]-2-methyl-phenyl]pyridi ne-2-carboxylic acid To a solution of NaBH(OAc)3 (73.74 mg, 347.92 µmol, 3.0 equiv.) and 3-[1-methyl-7-(4- piperidyl)indazol-3-yl]piperidine-2,6-dione (37.85 mg, 115.97 µmol, 1 equiv.) in DCM (1 mL) was added 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[2- methyl-3-[3-[1-(2-oxoethyl)-4-piperidyl]propoxy]phenyl]pyrid ine-2-carboxylic acid (81.63 mg, 115.97 µmol, 1.0 equiv.) dropwise at 0 °C. The mixture was stirred at 25 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[4-[3-(2,6-dioxo- 3-piperidyl)-1-methyl-indazol- 7-yl]-1-piperidyl]ethyl]-4-piperidyl]propoxy]-2-methyl-pheny l]pyridine-2-carboxylic acid (17.1 mg, 18.8 µmol, 16.2% yield, 95.3% purity) as a white solid. MS (ESI) m/z: 1014.6 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 10.89 (s, 1H), 8.15 (s, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 6.4 Hz, 1H), 7.54 (d, J = 8.0 Hz, 1H), 7.49 - 7.30 (m, 5H), 7.23 (d, J = 7.2 Hz, 1H), 7.12 - 7.03 (m, 2H), 6.94 - 6.84 (m, 2H), 6.64 (d, J = 7.6 Hz, 1H), 4.96 (s, 2H), 4.42 - 4.31 (m, 1H), 4.20 (s, 2H), 3.98 (s, 3H), 3.89 (s, 2H), 3.20 - 3.09 (m, 6H), 2.99 (s, 2H), 2.84 - 2.73 (m, 3H), 2.71 - 2.62 (m, 4H), 2.44 (s, 2H), 2.38 - 2.29 (m, 4H), 2.20 - 2.13 (m, 1H), 1.93 - 1.83 (m, 5H), 1.77 - 1.65 (m, 4H), 1.37 (s, 3H), 1.23 (s, 1H). Example 179. Preparation of Compound 257 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[1-[[1-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]-4-piperidyl]m ethyl]-4-piperidyl]butoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 4-[4-(3-bromo-2-methyl- phenoxy)butyl]piperidine-1-carboxylate A mixture of 3-bromo-2-methyl-phenol (650 mg, 3.48 mmol, 1.0 equiv.), tert-butyl 4-(4- hydroxybutyl)piperidine-1-carboxylate (1.07g, 4.17 mmol, 1.2 equiv.), and 2-(tributyl-λ5- phosphanylidene)acetonitrile (1.26 g, 5.21 mmol, 1.5 equiv.) in toluene (8 mL) was degassed, purged with N 2 , and stirred at 120 °C for 2 hours under N 2 atmosphere. The residue was concentrated, and the crude product was purified by reverse-phase HPLC to give tert-butyl 4- [4-(3-bromo-2-methyl-phenoxy)butyl]piperidine-1-carboxylate (1.1 g, 2.5 mmol, 74.2% yield) as a brown oil. MS (ESI) m/z: 335.1 [M+H] + . 1H NMR (400 MHz, DMSO-d6) δ = 7.17 - 7.12 (m, 1H), 7.11 - 7.05 (m, 1H), 6.95 (d, J = 8.0 Hz, 1H), 3.97 (t, J = 6.4 Hz, 2H), 3.91 (d, J = 12.0 Hz, 2H), 2.66 (d, J = 2.0 Hz, 2H), 2.22 (s, 3H), 1.75 - 1.67 (m, 2H), 1.62 (d, J = 12.0 Hz, 2H), 1.48 - 1.40 (m, 3H), 1.38 (s, 9H), 1.28 - 1.22 (m, 2H), 1.02 - 0.88 (m, 2H). Step B. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-(1-tert-butoxycarbo nyl-4-piperidyl)butoxy]-2- methyl-phenyl]pyridine-2-carboxylate tert-Butyl 4-[4-(3-bromo-2-methyl-phenoxy)butyl]piperidine-1-carboxylat e (500 mg, 1.17 mmol, 1.0 equiv.), tert-butyl.6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro -1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (718.30 mg, 1.17 mmol, 1.0 equiv.) and Ad2nBuP Pd G3 (85.40 mg, 117.26 µmol, 0.1 equiv.), and K 2 CO 3 (486.20 mg, 3.52 mmol, 3.0 equiv.) were taken up into a microwave tube in dioxane (6 mL) and H 2 O (2 mL).The sealed tube was heated at 100 °C for 1 hour under microwave. The mixture was concentrated, and the resulting residue was purified by flash silica gel chromatography (Eluent of 33% ethyl acetate/petroleum ether) to give tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[4-(1-tert- butoxycarbonyl-4-piperidyl)butoxy]-2-methyl-phenyl]pyridine- 2-carboxylate (700 mg, 841.2 µmol, 71.7% yield) as a brown oil. MS (ESI) m/z: 832.1 [M+H] + . 1H NMR (400 MHz, DMSO-d 6 ) δ = 12.86 (s, 1H), 8.02 (d, J = 7.6 Hz, 1H), 7.80 - 7.75 (m, 1H), 7.60 (d, J = 7.6 Hz, 1H), 7.48 - 7.42 (m, 3H), 7.39 - 7.34 (m, 2H), 7.12 - 7.07 (m, 1H), 6.93 (d, J = 8.4, 14.1 Hz, 2H), 6.57 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.03 (d, J = 7.2 Hz, 3H), 3.03 (t, J = 5.6 Hz, 2H), 1.99 (s, 4H), 1.87 (s, 2H), 1.37 - 1.36 (m, 9H), 1.18 (t, J = 7.2 Hz, 5H), 1.07 (s, 8H), 1.00 (s, 9H). Step C. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[2-methyl-3-[4-(4-piperidyl)butoxy]ph enyl]pyridine-2-carboxylic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[4-(1-tertbutoxycarbonyl-4-piperidyl)butoxy]-2-meth yl-phenyl]pyridine-2- carboxylate (130 mg, 156.24 µmol, 1.0 equiv.) in HCOOH (2 mL) was stirred at 90 °C for 2 hours. The mixture was concentrated to provide 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[2-methyl-3-[4-(4-piperidyl)b utoxy]phenyl]pyridine-2- carboxylic acid (81 mg, 119.8 µmol, 76.7% yield) as a brown oil, which was used in the next step without further purification. MS (ESI) m/z: 676.2 [M+H] + . Step D. Procedure for preparation of 1-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6- yl]piperidine-4-carbaldehyde A mixture of 3-[6-[4-(dimethoxymethyl)-1-piperidyl]-1-methyl-indazol-3-yl ]piperidine-2,6- dione (100 mg, 249.71 µmol, 1 equiv.) in HCOOH (1 mL) was degassed and purged with N2 three times. The mixture was then stirred at 90 °C for 2 hours under N 2 atmosphere. The reaction mixture was then concentrated to provide 1-[3-(2,6-dioxo-3-piperidyl)-1-methyl- indazol-6-yl]piperidine-4-carbaldehyde (74 mg, crude) as a brown oil, which was used in the next step without further purification. MS (ESI) m/z: 373.1 [M+H] + . Step E. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[4-[1-[[1-[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6-yl]-4- piperidyl]methyl]-4-piperidyl]butoxy]-2-methyl-phenyl]pyridi ne-2-carboxylic acid To a solution of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- [2-methyl-3-[4-(4-piperidyl)butoxy]phenyl]pyridine-2-carboxy lic acid (103.29 mg, 152.84 µmol, 1 equiv.) in DCM (0.5 mL) and IPA (0.5 mL) was added 1-[3-(2,6-dioxo-3-piperidyl)- 1-methyl-indazol-6-yl]piperidine-4-carbaldehyde (65 mg, 183.41 µmol, 1.2 equiv.) at 25 °C for 11 hours. The mixture was stirred at this temperature, and then NaBH(OAc)3 (97.18 mg, 458.52 µmol, 3 equiv.) was added at 0 °C. The resulting mixture was stirred at 25 °C for 1 hour. The reaction mixture was concentrated and purified by reverse-phase HPLC to give 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[4-[1-[[1-[3-(2,6-dioxo- 3-piperidyl)-1-methyl-indazol-6-yl]-4-piperidyl]methyl]-4-pi peridyl]butoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid (37.1 mg, 35.7 µmol, 23.3% yield, 97.4% purity) as an off- white solid. MS (ESI) m/z: 507.9 [M/2+H] + . 1H NMR (400 MHz, DMSO-d 6 ) δ = 10.84 (s, 1H), 8.18 (s, 1H), 8.01 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.50 - 7.30 (m, 6H), 7.12 - 7.04 (m, 1H), 6.94 - 6.84 (m, 3H), 6.80 (s, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.96 (s, 2H), 4.24 (dd, J = 4.8, 8.8 Hz, 1H), 3.99 - 3.95 (m, 2H), 3.91 (t, J = 5.2 Hz, 2H), 3.87 (s, 3H), 3.79 - 3.70 (m, 2H), 3.56 (s, 2H), 3.00 (t, J = 5.6 Hz, 2H), 2.87 (d, J = 10.0 Hz, 2H), 2.74 - 2.66 (m, 2H), 2.64 - 2.57 (m, 2H), 2.35 - 2.24 (m, 2H), 2.20 (d, J = 6.4 Hz, 2H), 1.97 - 1.86 (m, 5H), 1.82 - 1.77 (m, 2H), 1.74 - 1.69 (m, 2H), 1.61 (d, J = 11.2 Hz, 2H), 1.44 (d, J = 4.8 Hz, 2H), 1.31 - 1.16 (m, 6H) Example 180. Preparation of Compound 258 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[2-[1-[[1-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]-4-piperidyl]m ethyl]-4-piperidyl]ethoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 4-[2-(3-bromo-2-methyl- phenoxy)ethyl]piperidine-1-carboxylate To a solution of tert-butyl 4-(2-bromoethyl)piperidine-1-carboxylate (1 g, 3.42 mmol, 1.0 equiv.) and 3-bromo-2-methyl-phenol (704.07 mg, 3.76 mmol, 1.1 equiv.) in CH 3 CN (10 mL) was added K2CO3 (1.42 g, 10.27 mmol, 3.0 equiv.). The mixture was stirred at 60 °C for 16 hours. The mixture was filtered to give a filtrate, which was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~4% ethyl acetate/petroleum ether) to give tert-butyl 4-[2-(3-bromo-2-methyl- phenoxy)ethyl]piperidine-1-carboxylate (1.2 g, 2.9 mmol, 86.9% yield, 98.8% purity) as a yellow oil. MS (ESI) m/z: 298.2 [M-100+H] + 1 H NMR (400 MHz, CD3OD) δ = 7.13 - 7.09 (m, 1H), 7.02 (t, J = 8.0 Hz, 1H), 6.89 (d, J = 8.0 Hz, 1H), 4.10 - 4.07 (m, 1H), 4.07 - 4.03 (m, 3H), 2.84 - 2.69 (m, 2H), 2.28 (s, 3H), 1.79 - 1.73 (m, 5H), 1.45 (s, 9H), 1.22 - 1.11 (m, 2H) Step B. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-(1-tert-butoxycarbo nyl-4-piperidyl)ethoxy]-2- methyl-phenyl]pyridine-2-carboxylate tert-Butyl 4-[2-(3-bromo-2-methyl-phenoxy)ethyl]piperidine-1-carboxylat e (1.18 g, 2.96 mmol, 1.0 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (1.81 g, 2.96 mmol, 1.0 equiv.), KF (516.31 mg, 8.89 mmol, 208.19 µL, 3.0 equiv.), and Ad2nBuP Pd G3 (215.74 mg, 296.23 µmol, 0.1 equiv.) were taken up into a microwave tube in dioxane (10 mL) and H 2 O (1 mL). The sealed tube was heated at 100 °C for 2 hours under microwave. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~30% ethyl acetate/petroleum ether) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3- [2-(1-tert-butoxycarbonyl-4-piperidyl)ethoxy]-2-methyl-pheny l]pyridine-2-carboxylate (1.7 g, 2.0 mmol, 69.2% yield, 96.9% purity) was obtained as a yellow solid. MS (ESI) m/z: 804.5 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.86 (s, 1H), 8.01 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.60 (d, J = 7.6 Hz, 1H), 7.49 - 7.41 (m, 3H), 7.39 - 7.31 (m, 2H), 7.09 (t, J = 8.0 Hz, 1H), 6.97 - 6.89 (m, 2H), 6.57 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.04 - 3.97 (m, 2H), 3.96 - 3.83 (m, 4H), 3.07 - 2.99 (m, 2H), 2.76 - 2.57 (m, 2H), 1.87 (s, 3H), 1.66 (s, 5H), 1.37 (s, 9H), 1.11 - 1.03 (m, 2H), 1.00 (s, 9H) Step C. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[2-methyl-3-[2-(4-piperidyl)e thoxy]phenyl]pyridine-2- carboxylic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[2-(1-tert-butoxycarbonyl-4-piperidyl)ethoxy]-2-met hyl-phenyl]pyridine-2- carboxylate (1.7 g, 2.11 mmol, 1.0 equiv.) and HCl/ dioxane (4 M, 17 mL, 32.2 equiv.) was stirred at 25 °C for 2 hours. The mixture was concentrated under reduced pressure to give 6- [8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinol in-2-yl]-3-[2-methyl-3-[2-(4- piperidyl)ethoxy]phenyl]pyridine-2-carboxylic acid (1.5 g, 1.8 mmol, 88.5% yield, 85.4% purity) as a yellow solid. MS (ESI) m/z: 648.4 [M+H] + Step D. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[2-[1-[[1-[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6-yl]-4- piperidyl]methyl]-4-piperidyl]ethoxy]-2-methyl-phenyl]pyridi ne-2-carboxylic acid A solution of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[2- methyl-3-[2-(4-piperidyl)ethoxy]phenyl]pyridine-2-carboxylic acid (150 mg, 231.56 µmol, 1.0 equiv.) and 1-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]piperidin e-4-carbaldehyde (82.06 mg, 231.56 µmol, 1.0 equiv.) in DCM (5 mL) and IPA (0.5 mL) was stirred at 25 °C for 12 hours. Then NaBH(OAc) 3 (147.23 mg, 694.68 µmol, 3.0 equiv.) was added at 0 °C. After the addition, the mixture was stirred at 25 °C for 0.5 hour. The mixture was concentrated under reduced pressure to give a residue, which was diluted with DMF (2 mL) and purified by prep- HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3- [2-[1-[[1-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]- 4-piperidyl]methyl]-4- piperidyl]ethoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid (40 mg, 37.6 µmol, 16.2% yield, 97.0% purity) as an off-white solid. MS (ESI) m/z: 986.5 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.85 (s, 1H), 8.02 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.50 - 7.41 (m, 4H), 7.39 - 7.31 (m, 2H), 7.11 - 7.05 (m, 1H), 6.97 - 6.86 (m, 3H), 6.82 - 6.76 (m, 1H), 6.64 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.26 - 4.22 (m, 1H), 3.99 (t, J = 6.0 Hz, 2H), 3.91 (t, J = 5.6 Hz, 2H), 3.89 - 3.86 (m, 3H), 3.78 - 3.74 (m, 2H), 3.57 - 3.55 (m, 2H), 3.02 (t, J = 6.4 Hz, 2H), 2.88 - 2.83 (m, 2H), 2.74 - 2.68 (m, 2H), 2.63 - 2.58 (m, 2H), 2.19 - 2.15 (m, 2H), 1.95 - 1.84 (m, 5H), 1.82 - 1.75 (m, 2H), 1.74 - 1.64 (m, 5H), 1.55 - 1.44 (m, 1H), 1.29 - 1.18 (m, 4H) Example 181. Preparation of Compound 266b 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4-[(3S)-3-[1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-o xo-ethyl]-4-piperidyl]butoxy]- 2-methyl-phenyl]pyridine-2-carboxylic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(4-((3S)-3-(1- (2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)am ino)-2-oxoethyl)piperidin-4- yl)butoxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of tert-butyl 4-[3-(4-bromo-3-methyl-phenoxy)-1- methyl-propyl]piperidine-1-carboxylate To a solution of tert-butyl 4-(3-hydroxy-1-methyl-propyl)piperidine-1-carboxylate (2.2 g, 8.55 mmol, 1.0 equiv.) and 4-bromo-3-methyl-phenol (1.76 g, 9.40 mmol, 1.1 equiv.) in toluene (20 mL) was added 2-(tributyl-λ5-phosphanylidene)acetonitrile (2.48 g, 10.26 mmol, 1.2 equiv.). The mixture was stirred at 120 °C for 12 hours. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by reverse-phase HPLC (with 0.1% HCOOH) to give tert-butyl 4-[3-(4-bromo-3-methyl-phenoxy)-1-methyl-propyl]piperidine-1 - carboxylate (2.7 g, 6.1 mmol, 72.1% yield, 97.4% purity) as a brown oil. MS (ESI) m/z: 448.2 [M+23] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.42 (d, J = 8.8 Hz, 1H), 6.95 (d, J = 2.8 Hz, 1H), 6.71 (dd, J = 2.8, 8.8 Hz, 1H), 4.06 - 3.89 (m, 4H), 2.72 - 2.54 (m, 2H), 2.29 (s, 3H), 1.84 - 1.74 (m, 1H), 1.58 - 1.45 (m, 4H), 1.38 (s, 10H), 1.15 - 0.97 (m, 2H), 0.85 (d, J = 6.4 Hz, 3H) Step B. Procedure for preparation of tert-butyl 4-[(1S)-3-(4-bromo-3-methyl-phenoxy)-1- methyl-propyl]piperidine-1-carboxylate The compound tert-butyl 4-[3-(4-bromo-3-methyl-phenoxy)-1-methyl-propyl]piperidine-1 - carboxylate (1.4 g, 3.28 mmol, 1.0 equiv.) was purified by prep-HPLC to give tert-butyl 4- [(1R)-3-(4-bromo-3-methyl-phenoxy)-1-methyl-propyl]piperidin e-1-carboxylate (430 mg, 1.00 mmol, 30.51% yield, 99.32% purity) as a brown oil and tert-butyl 4-[(1S)-3-(4-bromo-3- methyl-phenoxy)-1-methyl-propyl]piperidine-1-carboxylate (380 mg, 865.7 µmol, 26.3% yield, 97.1% purity) as a brown oil MS (ESI) m/z: 448.2 [M+23] + Step C. Procedure for preparation of 4-[(1S)-3-(4-bromo-3-methyl-phenoxy)-1-methyl- propyl]piperidine A mixture of tert-butyl 4-[(1S)-3-(4-bromo-3-methyl-phenoxy)-1-methyl-propyl]piperid ine-1- carboxylate (380 mg, 891.21 µmol, 1.0 equiv.) and HCl/EtOAc (4 M, 4.5 mL, 20.2 equiv.) was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give 4-[(1S)-3-(4-bromo-3-methyl-phenoxy)-1-methyl-propyl]piperid ine (320 mg, crude) as a yellow solid. MS (ESI) m/z: 326.0 [M+H] + ( 80 Br). Step D. Procedure for preparation of ethyl 2-[4-[(1S)-3-(4-bromo-3-methyl-phenoxy)-1- methyl-propyl]-1-piperidyl]acetate To a solution of 4-[(1S)-3-(4-bromo-3-methyl-phenoxy)-1-methyl-propyl]piperid ine (320 mg, 882.19 µmol, 1.0 equiv., HCl) and ethyl 2-bromoacetate (132.59 mg, 793.97 µmol, 87.81 µL, 0.9 equiv.) in CH 3 CN (3.5 mL) was added K 2 CO 3 (365.77 mg, 2.65 mmol, 3.0 equiv.). The mixture was stirred at 60 °C for 2.5 hours. The reaction mixture was filtered and concentrated under reduced pressure to give an oil. The oil was purified by column chromatography (SiO2, petroleum ether/ethyl acetate 4/1 to 4/1) to give ethyl 2-[4-[(1S)-3-(4-bromo-3-methyl- phenoxy)-1-methyl-propyl]-1-piperidyl]acetate (228 mg, 519.5 µmol, 58.9% yield, 93.9% purity) as a colorless oil. MS (ESI) m/z: 412.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.42 (d, J = 8.8 Hz, 1H), 6.95 (d, J = 2.4 Hz, 1H), 6.71 (dd, J = 2.4, 8.8 Hz, 1H), 4.07 (q, J = 7.2 Hz, 2H), 4.02 - 3.89 (m, 2H), 3.14 (s, 2H), 2.84 (d, J = 10.0 Hz, 2H), 2.29 (s, 3H), 2.08 (t, J = 11.2 Hz, 2H), 1.85 - 1.75 (m, 1H), 1.58 - 1.43 (m, 4H), 1.32 - 1.21 (m, 2H), 1.21 - 1.15 (m, 4H), 0.86 (d, J = 6.4 Hz, 3H) Step E. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[(3S)-3-[1-(2-ethoxy-2 -oxo-ethyl)-4- piperidyl]butoxy]-2-methyl-phenyl]pyridine-2-carboxylate Ethyl 2-[4-[(1S)-3-(4-bromo-3-methyl-phenoxy)-1-methyl-propyl]-1-p iperidyl]acetate (150 mg, 363.76 µmol, 1.0 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborol an-2-yl)pyridine-2-carboxylate (222.82 mg, 363.76 µmol, 1.0 equiv.), KF (63.40 mg, 1.09 mmol, 25.56 µL, 3 equiv.), and Ad2nBuP Pd G3 (26.49 mg, 36.38 µmol, 0.1 equiv.) were taken up into a microwave tube in dioxane (2 mL) and H 2 O (0.2 mL).The sealed tube was heated at 100 °C for 1 hour under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=3/1 to 1/1) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[4-[(3S)-3-[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]but oxy]-2-methyl-phenyl]pyridine- 2-carboxylate (160 mg, 180.6 µmol, 49.6% yield, 92.4% purity) as a yellow oil. MS (ESI) m/z: 410.0 [M/2+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.85 (s, 1H), 8.02 (d, J = 7.6 Hz, 1H), 7.77 (d, J = 8.0 Hz, 1H), 7.60 (d, J = 7.6 Hz, 1H), 7.49 - 7.41 (m, 3H), 7.39 - 7.32 (m, 2H), 6.92 (d, J = 8.8 Hz, 1H), 6.85 (d, J = 8.4 Hz, 1H), 6.82 (d, J = 2.4 Hz, 1H), 6.72 (dd, J = 2.4, 8.4 Hz, 1H), 4.96 (s, 2H), 4.07 - 3.92 (m, 5H), 3.86 (t, J = 6.0 Hz, 2H), 3.14 (s, 2H), 3.03 (t, J = 6.0 Hz, 2H), 2.84 (d, J = 9.2 Hz, 2H), 2.08 (t, J = 11.2 Hz, 2H), 1.99 (s, 3H), 1.84 - 1.75 (m, 1H), 1.51 (d, J = 12.4 Hz, 4H), 1.31 - 1.22 (m, 3H), 1.19 - 1.17 (m, 3H), 1.03 (s, 9H), 0.86 (d, J = 6.8 Hz, 3H) Step F. Procedure for preparation of 2-[4-[(1S)-3-[4-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-3- methyl-phenoxy]-1-methyl-propyl]-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[4-[(3S)-3-[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]but oxy]-2-methyl-phenyl]pyridine- 2-carboxylate (160 mg, 195.59 µmol, 1.0 equiv.) in THF (1.5 mL) and H2O (0.5 mL)was added LiOH•H2O (14.05 mg, 586.77 µmol, 3 equiv.).The mixture was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to remove THF. The resulting residue was diluted with H2O (10 mL), treated with HCl (1 M) until pH= 2, and filtered to give a filter cake, which was dried to give 2-[4-[(1S)-3-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyr idyl]-3-methyl-phenoxy]-1- methyl-propyl]-1-piperidyl]acetic acid (65 mg, 74.4 µmol, 38.0% yield, 90.5% purity) as a yellow solid. MS (ESI) m/z: 790.5 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.96 - 12.76 (m, 1H), 8.02 (d, J = 7.2 Hz, 1H), 7.77 (d, J = 7.2 Hz, 1H), 7.59 (d, J = 6.8 Hz, 1H), 7.49 - 7.30 (m, 5H), 6.92 (d, J = 8.4 Hz, 1H), 6.86 (d, J = 8.4 Hz, 1H), 6.82 (d, J = 2.0 Hz, 1H), 6.73 (d, J = 2.4 Hz, 1H), 4.96 (s, 2H), 4.08 - 3.91 (m, 2H), 3.86 (t, J = 5.6 Hz, 2H), 3.05-3.02(m, 2H), 3.03 (t, J = 6.8 Hz, 2H), 2.62 - 2.54 (m, 5H), 2.00 (s, 3H), 1.83 - 1.76 (m, 1H), 1.67 - 1.60 (m, 2H), 1.58 - 1.41 (m, 4H), 1.38 - 1.32 (m, 1H), 1.23 (s, 2H), 1.06 - 0.99 (m, 9H), 0.87 (d, J = 6.4 Hz, 3H) Step G. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[(3S)-3-[1-[2-[[3-(2,6 -dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-4-piperidyl]butoxy]-2-methy l-phenyl]pyridine-2- carboxylate To a solution of 2-[4-[(1S)-3-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-3-methyl- phenoxy]-1-methyl-propyl]-1- piperidyl]acetic acid (60 mg, 75.95 µmol, 1.0 equiv.) in DMF (1 mL) was added HATU (34.65 mg, 91.14 µmol, 1.2 equiv.) and DIEA (29.45 mg, 227.85 µmol, 39.69 µL, 3.0 equiv.). The mixture was stirred at 25 °C for 16 hours. The mixture was added to H2O (10 mL), and the resulting mixture was filtered to give a filter cake, which was dried to give tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[4-[(3S)-3-[1-[2-[[3-(2,6- dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl] -4-piperidyl]butoxy]-2-methyl- phenyl]pyridine-2-carboxylate (106 mg, 102.9 µmol, 135.4% yield) as an off-white solid. MS (ESI) m/z: 1030.5 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.85 (s, 1H), 10.88 (s, 1H), 8.02 (d, J = 8.0 Hz, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.70 - 7.62 (m, 1H), 7.60 (d, J = 7.6 Hz, 1H), 7.48 - 7.41 (m, 3H), 7.36 (dd, J = 7.2, 13.6 Hz, 2H), 7.23 - 7.14 (m, 1H), 6.92 (d, J = 8.8 Hz, 1H), 6.86 (d, J = 8.4 Hz, 1H), 6.83 (d, J = 2.4 Hz, 1H), 6.73 (J = 2.8, 8.4 Hz, 1H), 4.96 (s, 2H), 4.33 (dd, J = 5.2, 10.0 Hz, 1H), 4.05 - 3.96 (m, 2H), 3.93 (s, 3H), 3.86 (t, J = 5.6 Hz, 2H), 3.03 (t, J = 6.0 Hz, 2H), 2.63 (dd, J = 4.8, 9.6 Hz, 2H), 2.60 - 2.54 (m, 4H), 2.24 - 2.11 (m, 2H), 2.01 (s, 3H), 1.91 - 1.75 (m, 2H), 1.69 - 1.43 (m, 6H), 1.23 (s, 2H), 1.03 (s, 9H), 0.90 (d, J = 6.4 Hz, 3H) Step H. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[4-[(3S)-3-[1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]butoxy]-2-methyl-phenyl]p yridine-2-carboxylic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(4-((3S)-3- (1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl )amino)-2- oxoethyl)piperidin-4-yl)butoxy)-2-methylphenyl)picolinic acid] A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[4-[(3S)-3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo- ethyl]-4-piperidyl]butoxy]-2-methyl-phenyl]pyridine-2-carbox ylate (100 mg, 97.06 µmol, 1.0 equiv.) in DCM (0.5 mL) and TFA (0.5 mL) was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove DCM. The resulting residue was diluted with DMF (1.5 mL) and purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[(3S)-3-[ 1-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]-4-piper idyl]butoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-((3S)-3-(1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)butoxy)-2-meth ylphenyl)picolinic acid] (22.6 mg, 89.8% purity) as a yellow solid. MS (ESI) m/z: 974.5 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.04 - 12.67 (m, 1H), 10.88 (s, 1H), 9.99 - 9.75 (m, 1H), 8.03 (d, J = 9.2 Hz, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.66 - 7.59 (m, 2H), 7.49 - 7.42 (m, 3H), 7.40 - 7.32 (m, 2H), 7.21 (dd, J = 1.2, 8.8 Hz, 1H), 6.93 (dd, J = 8.4, 14.0 Hz, 2H), 6.80 (d, J = 2.4 Hz, 1H), 6.71 (dd, J = 2.8, 8.8 Hz, 1H), 4.97 (s, 2H), 4.32 (dd, J = 5.2, 9.6 Hz, 1H), 4.02 - 3.95 (m, 2H), 3.94 - 3.88 (m, 5H), 3.21 - 3.15 (m, 2H), 3.02 (t, J = 5.2 Hz, 2H), 3.00 - 2.92 (m, 2H), 2.64 - 2.60 (m, 2H), 2.40 - 2.27 (m, 2H), 2.20 - 2.13 (m, 2H), 2.03 (s, 3H), 1.89 - 1.82 (m, 1H), 1.65 - 1.52 (m, 4H), 1.46 - 1.35 (m, 2H), 1.23 (s, 1H), 0.91 (d, J = 6.4 Hz, 3H) Example 182. Preparation of Compound 270a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pip eridin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of 3-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]propan-1-ol To a solution of ethyl 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]propanoate (600 mg, 1.62 mmol, 1 equiv.) in THF (10 mL) was added LiAlH 4 (55.50 mg, 1.46 mmol, 0.9 equiv.) at 0 °C. The mixture was stirred at 0 °C for 1.5 hours. The reaction mixture was quenched by addition H2O (0.05 mL), 15% NaOH (0.05 mL), and H2O (15mL), and then extracted with ethyl acetate (10 mL × 3). The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was used in the next step without purification. The compound 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]propan-1-ol (500 mg, 1.5 mmol, 94.0% yield) was obtained as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.14 - 7.02 (m, 3H), 4.23 - 4.21 (m, 1H), 3.37 - 3.36 (m, 3H), 3.34 -3.35 (m, 1H), 2.26 - 2.24 (m, 1H), 2.21 -2.19 (m, 2H), 2.06 - 2.04 (m, 2H), 1.76 - 1.72 (m, 2H), 1.41 - 1.42 (m, 2H), 1.40 - 1.39 (m, 2H), 1.38 - 1.36 (m, 2H), 1.20 - 1.15 (m, 3H), 1.03 - 0.99(m, 2H). Step B. Procedure for preparation of 3-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]propanal To a solution of (COCl) 2 (167.23 mg, 1.41 mmol, 101.97 µL, 2 equiv.) in DCM (30 mL) was added dropwise to a solution of DMSO (219.65 mg, 2.81 mmol, 219.65 µL, 4 equiv.) in DCM (30 mL) at -70 °C under N 2 atmosphere. The mixture was stirred at -70 °C for 1 hour. After which time 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]propan-1-ol (230 mg, 702.81 µmol, 1 equiv.) in DCM (30 mL) was added dropwise at -70 °C. The solution was stirred for 1 hour at -70 °C. Then TEA (426.70 mg, 4.22 mmol, 586.94 µL, 6 equiv.) was added into the solution. The solution was stirred at -70 °C for 1 hour under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~43% ethyl acetate/petroleum ether) to give 3-[4- (3-bromo-2-methyl-phenoxy)cyclohexyl]propanal (200 mg, 467.3 µmol, 66.5% yield, 76% purity) as a white solid. Step C. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[2-methyl-3-[4-(3- oxopropyl)cyclohexoxy]phenyl]pyridine-2-carboxylate A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine- 2-carboxylate (414.34 mg, 676.42 µmol, 1.1 eq), 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]propanal (200 mg, 614.93 µmol, 1 eq), KF (1.5 M, 614.93 uL, 1.5 equiv.), and Ad 2 nBuP Pd G 3 (cataCXium® A Pd G 3 ) (134.35 mg, 184.48 µmol, 0.3 equiv.) in 1,4-dioxane (15 mL) was degassed and purged with N 2 and stirred at 100 °C for 1 hour under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~37% ethyl acetate/petroleum ether). The compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[2-methyl-3-[4- (3-oxopropyl)cyclohexoxy]phenyl]pyridine-2-carboxylate (300 mg, 410.4 µmol, 66.7% yield) was obtained as a yellow oil. Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo -3-piperidyl)-1-methyl- indazol-7-yl]-1-piperidyl]propyl]cyclohexoxy]-2-methyl-pheny l]pyridine-2-carboxylate A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[2-methyl-3-[4-(3-oxopropyl)cyclohexoxy]phenyl]pyridin e-2-carboxylate (50 mg, 68.41 µmol, 1 equiv.), 3-[1-methyl-7-(4-piperidyl)indazol-3-yl]piperidine-2,6-dione (33.49 mg, 102.61 µmol, 1.5 equiv.) in DCM (1 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 10 hours under N 2 atmosphere. NaBH(OAc) 3 (29.00 mg, 136.82 µmol, 2 equiv.) was then added to the mixture, which was stirred at 25 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give tert- butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[3-[4- [3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]-1-piperidy l]propyl]cyclohexoxy]-2- methyl-phenyl]pyridine-2-carboxylate (60 mg, 57.6 µmol, 84.2% yield) as a white solid. MS (ESI) m/z: 1042.3 [M+H] + Step E. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperidin-1-yl)propyl)cyclohexyl)oxy) -2-methylphenyl)picolinic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-inda zol-7-yl]-1- piperidyl]propyl]cyclohexoxy]-2-methyl-phenyl]pyridine-2-car boxylate (50.00 mg, 48.02 µmol, 1 equiv.) in DCM (0.2 mL) was added TFA (5.47 mg, 48.02 µmol, 3.56 µL, 1 equiv.). The mixture was stirred at 25 °C for 0.5 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-7-yl)piperidin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (21.9 mg, 21.9 µmol, 45.6% yield, 98.4% purity) as a white solid. MS (ESI) m/z: 493.9 [M/2+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.94 – 10.82 (m, 1H), 8.17 (s, 1H), 8.03 (d, J = 8.4 Hz, 1H), 7.79 (d, J = 8.4 Hz, 1H), 7.62 (d, J = 7.8 Hz, 1H), 7.49 – 7.41 (m, 3H), 7.37 (J = 7.2 Hz, 3H), 7.07 – 7.00 (m, 3H), 6.96 – 6.88 (m, 2H), 6.63 – 6.61 (m, 1H), 4.97 (s, 2H), 4.35 – 4.31 (m, 1H), 4.24 (s, 3H), 4.21 – 4.16 (m, 1H), 3.92 – 3.89 (m, 2H), 3.03 – 3.01 (m, 4H), 2.68 – 2.60 (m, 3H), 2.37 – 2.34 (m, 4H), 2.22 – 2.14 (m, 3H), 2.12 – 2.02 (m, 3H), 1.88 (s, 3H), 1.82 – 1.78 (m, 2H), 1.52 – 1.46 (m, 2H), 1.39 – 1.32 (m, 2H), 1.30 – 1.19 (m, 4H), 1.10 – 1.02 (m, 2H). Example 183. Preparation of Compound 271a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-((2R)-3-(1-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino )-2-oxoethyl)piperidin-4-yl)-2- methylpropoxy)-2-methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(4-((2R)-3-(1- (2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)am ino)-2-oxoethyl)piperidin-4- yl)-2-methylpropoxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of tert-butyl 4-(3-(4-bromo-3-methylphenoxy)-2- methylpropyl)piperidine-1-carboxylate A mixture of tert-butyl 4-(3-hydroxy-2-methylpropyl)piperidine-1-carboxylate (2.2 g, 8.55 mmol, 1 equiv.), 4-bromo-3-methyl-phenol (1.92 g, 10.26 mmol, 1.2 equiv.), and 2-(tributyl- λ5-phosphanylidene)acetonitrile (2.48 g, 10.26 mmol, 1.2 equiv.) in toluene (20 mL) was degassed and purged with N2 and stirred at 120 °C for 4 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (with HCOOH) to give tert-butyl 4-(3-(4-bromo-3- methylphenoxy)-2-methylpropyl)piperidine-1-carboxylate (2.6 g, 6.0 mmol, 70.8% yield, 99.2% purity) as a yellow oil. MS (ESI) m/z: 370.7 [M+H] + . 1 H NMR (400 MHz, CDCl3) δ = 7.39 (d, J = 8.8 Hz, 1H), 6.78 (d, J = 2.8 Hz, 1H), 6.60 (dd, J = 2.8, 5.6 Hz, 1H), 4.09 (d, J = 13.2 Hz, 2H), 3.78 - 3.66 (m, 2H), 2.73 - 2.66 (m, 2H), 2.36 (s, 3H), 2.06 - 2.01 (m, 1H), 1.71 - 1.64 (m, 2H), 1.46 (s, 9H), 1.44 - 1.37 (m, 1H), 1.22 - 1.03 (m, 4H), 1.01 (d, J = 6.4 Hz, 3H). Step B. Procedure for preparation of tert-butyl (R)-4-(3-(4-bromo-3-methylphenoxy)-2- methylpropyl)piperidine-1-carboxylate tert-Butyl 4-(3-(4-bromo-3-methylphenoxy)-2-methylpropyl)piperidine-1-c arboxylate obtained from the preceding step was resolved by SFC to give tert-butyl (S)-4-(3-(4-bromo-3- methylphenoxy)-2-methylpropyl)piperidine-1-carboxylate (1 g, 2.28 mmol, 40.58% yield, 97.39% purity) as a yellow oil and tert-butyl (R)-4-(3-(4-bromo-3-methylphenoxy)-2- methylpropyl)piperidine-1-carboxylate (0.9 g, 2.1 mmol, 37.2% yield, 99.4% purity) as a yellow oil. Step C. Procedure for preparation of (R)-4-(3-(4-bromo-3-methylphenoxy)-2- methylpropyl)piperidine To a solution of tert-butyl (R)-4-(3-(4-bromo-3-methylphenoxy)-2-methylpropyl)piperidine -1- carboxylate (900 mg, 2.11 mmol, 1 equiv.) was added HCl/EtOAc (4 M, 527.69 µL, 1 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give (R)-4-(3-(4-bromo-3-methylphenoxy)-2- methylpropyl)piperidine (700 mg, crude) as a white solid. MS (ESI) m/z: 326.7 [M+H] + . Step D. Procedure for preparation of ethyl (R)-2-(4-(3-(4-bromo-3-methylphenoxy)-2- methylpropyl)piperidin-1-yl)acetate To a solution of ethyl 2-bromoacetate (153.55 mg, 919.48 µmol, 101.69 µL, 1 equiv.) and (R)- 4-(3-(4-bromo-3-methylphenoxy)-2-methylpropyl)piperidine (300.00 mg, 919.48 µmol, 1 equiv.) in CH 3 CN (4 mL) was added K 2 CO 3 (381.23 mg, 2.76 mmol, 3 equiv.). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give ethyl (R)-2-(4-(3-(4-bromo-3-methylphenoxy)-2- methylpropyl)piperidin-1-yl)acetate (300 mg, crude) as a white solid. MS (ESI) m/z: 412.0 [M+H] + . Step E. Procedure for preparation of tert-butyl (R)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(4-(3-(1-(2-ethoxy-2-oxoe thyl)piperidin-4-yl)-2- methylpropoxy)-2-methylphenyl)picolinate A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)picolinat e (222.82 mg, 363.76 µmol, 1 equiv.), ethyl (R)-2-(4-(3-(4-bromo-3-methylphenoxy)-2-methylpropyl)piperid in-1-yl)acetate (150.00 mg, 363.76 µmol, 1 equiv.) in dioxane (3 mL) was added KF (1.5 M, 727.52 µL, 3 equiv.) and Ad2nBuP Pd G3(cataCXium® A Pd G3 ) (26.49 mg, 36.38 µmol, 0.1 equiv.). After addition, the mixture was degassed and purged with N 2 three times, and then the mixture was stirred at 100 °C for 2 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~38% ethyl acetate/petroleum ether) to give compound tert- butyl (R)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquino lin-2(1H)-yl)-3-(4-(3-(1- (2-ethoxy-2-oxoethyl)piperidin-4-yl)-2-methylpropoxy)-2-meth ylphenyl)picolinate (170 mg, 182.3 µmol, 50.1% yield, 87.7% purity) as a yellow solid. MS (ESI) m/z: 819.6 [M+H] + . 1 H NMR (400 MHz, CDCl3) δ = 7.84 (d, J = 7.6 Hz, 1H), 7.56 (t, J = 7.2 Hz, 2H), 7.41 - 7.28 (m, 5H), 7.00 - 6.84 (m, 2H), 6.77 (d, J = 2.4 Hz, 1H), 6.69 (dd, J = 2.8, 5.6 Hz, 1H), 5.04 (s, 2H), 4.22 (q, J = 7.2 Hz, 2H), 4.13 - 4.04 (m, 2H), 3.83 - 3.69 (m, 2H), 3.38 (S, 2H), 3.18 - 2.95 (m, 4H), 2.13 - 1.99 (m, 5H), 1.55 - 1.43 (m, 4H), 1.34 - 1.24 (m, 5H), 1.19 (s, 9H), 1.15 - 1.07 (m, 2H), 1.06 - 0.98 (m, 3H). Step F. Procedure for preparation of (R)-2-(4-(3-(4-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-3- methylphenoxy)-2-methylpropyl)piperidin-1-yl)acetic acid To a solution of tert-butyl (R)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquino lin- 2(1H)-yl)-3-(4-(3-(1-(2-ethoxy-2-oxoethyl)piperidin-4-yl)-2- methylpropoxy)-2- methylphenyl)picolinate (170.00 mg, 207.82 µmol, 1 equiv.) in THF (2 mL) was added LiOH•H 2 O (26.16 mg, 623.45 µmol, 3 equiv.) and H 2 O (0.5 mL). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to remove THF. The aqueous phase was adjusted to pH=4~5 with 1M HCl. The reaction mixture was filtered. After addition, the filter cake was diluted in ethyl acetate. The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue compound (R)-2-(4-(3-(4-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-3-methylpheno xy)-2-methylpropyl)piperidin- 1-yl)acetic acid (160 mg, crude) as a yellow solid. MS (ESI) m/z: 790.4 [M+H] + . Step G. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-((2R)-3-(1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)-2-methylpr opoxy)-2- methylphenyl)picolinate To a solution of (R)-2-(4-(3-(4-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-3-methylpheno xy)-2-methylpropyl)piperidin- 1-yl)acetic acid (160.00 mg, 202.54 µmol, 1 equiv.), 3-(6-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (52.31 mg, 202.54 µmol, 1 equiv.) in pyridine (2 mL) was added EDCI (58.24 mg, 303.81 µmol, 1.5 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was quenched by the addition of H 2 O (2mL). The reaction mixture was filtered, and the filter cake was washed with water (5 mL). Afterwards, the filter cake was washed with DCM (10 mL). The combined organic layers were dried over Na2SO4, filtered, and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(4-((2R)-3-(1-(2-((3-(2,6 -dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)-2-methylpr opoxy)-2- methylphenyl)picolinate (160 mg, crude) as a yellow solid. MS (ESI) m/z: 1030.4 [M+H] + . Step H. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-((2R)-3-(1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)-2-methylpr opoxy)-2- methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-((2R)-3-(1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)-2-methylpr opoxy)-2- methylphenyl)picolinic acid] To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-((2R)-3-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1 -methyl-1H-indazol-6- yl)amino)-2-oxoethyl)piperidin-4-yl)-2-methylpropoxy)-2-meth ylphenyl)picolinate (160.00 mg, 155.30 µmol, 1 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 86.97 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue (100 mg, crude) as a brown oil, which was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-((2R)-3-(1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)-2-methylpropo xy)-2-methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(4-((2R)- 3-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)amino)-2-oxoethyl)piperidin- 4-yl)-2-methylpropoxy)-2-methylphenyl)picolinic acid] (74.6 mg, 70.3 µmol, 68.5% yield, 91.8% purity) as an off white solid. MS (ESI) m/z: 974.4 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 13.11 - 12.52 (m, 1H), 10.89 (s, 1H), 10.49 - 9.96 (m, 1H), 8.14 (s, 1H), 8.07 - 7.99 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.64 (dd, J = 8.8, 8.0 Hz, 2H), 7.50 - 7.41 (m, 3H), 7.41 - 7.32 (m, 2H), 7.18 (d, J = 8.8 Hz, 1H), 6.94 (dd, J = 8.8, 4.8 Hz, 2H), 6.79 (s, 1H), 6.76 - 6.65 (m, 1H), 4.97 (s, 2H), 4.33 (dd, J = 5.2, 4.4 Hz, 1H), 3.98 - 3.89 (m, 5H), 3.88 - 3.65 (m, 4H), 3.02 (t, J = 5.2 Hz, 2H), 2.75 - 2.54 (m, 4H), 2.35 - 2.33 (m, 1H), 2.23 - 2.11 (m, 1H), 2.08 - 1.91 (m, 5H), 1.78 (d, J = 11.6 Hz, 2H), 1.62 - 1.49 (m, 1H), 1.48 - 1.30 (m, 3H), 1.23 - 1.13 (m, 2H), 1.04 - 0.94 (m, 3H). Example 184. Preparation of Compound 282a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1R,4r)-4- ((2R)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 6-yl)piperazin-1-yl)-2- methylpropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1R,4r)-4- ((2R*)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)piperazin-1-yl)-2- methylpropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of ((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)methanol To a mixture of methyl (1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexane-1-carboxylate (40 g, 48.90 mmol, 40% purity, 1 equiv.) in THF (350 mL) was slowly added LiAlH4 (1.95 g, 51.34 mmol, 1.05 equiv.) at 0 °C, and then the mixture was stirred at 25 ℃ for 2 hours. The mixture was quenched by Na 2 SO 4 .10 H 2 O (3 g). The resulting solution was poured onto ice-water (300 mL) slowly. The pH of the resulting mixture was adjusted to pH 4~5 with 1 M HCl. The mixture was then extracted with EtOAc (300 mL × 3), washed with brine (400 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (Eluent of 0~12% ethyl acetate / petroleum ether) to give ((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)methanol (20.05 g, 67.0 mmol, 45.6% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.15 - 7.11 (m, 1H), 7.06 (t, J = 8.0 Hz, 1H), 7.03 - 6.99 (m, 1H), 4.26 - 4.17 (m, 1H), 2.21 (s, 3H), 2.06 (dd, J = 2.4, 11.6 Hz, 2H), 1.98 (s, 1H), 1.78 (d, J = 11.6 Hz, 2H), 1.58 (d, J = 4.8 Hz, 2H), 1.40 - 1.34 (m, 5H) Step B. Procedure for preparation of (1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexane- 1-carbaldehyde To a mixture of (COCl) 2 (8.48 g, 66.84 mmol, 5.85 mL, 2 equiv.) in DCM (80 mL) at -78°C was added DMSO (10.45 g, 133.69 mmol, 10.45 mL, 4 equiv.). The mixture was stirred for 0.5 hours and was then treated with ((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)methanol (10 g, 33.42 mmol, 1 equiv.) in DCM (200 mL) and stirred at-78 °C for 1 hour. Then the mixture was treated with TEA (20.29 g, 200.53 mmol, 27.91 mL, 6 equiv.) and warmed to 25 °C following by stirring for 0.5 hour under N2 atmosphere. The reaction mixture was diluted with H2O (150 mL) and extracted with DCM (100 mL × 3). The combined organic layers were washed with brine (200 mL), filtered, and concentrated under reduced pressure to give (1r,4r)- 4-(3-bromo-2-methylphenoxy)cyclohexane-1-carbaldehyde (33.2 g, crude) as a white solid MS (ESI) m/z: 296.9 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 9.61 (s, 1H), 7.16 - 7.13 (m, 1H), 7.10 - 7.05 (m, 1H), 7.04 - 7.01 (m, 1H), 4.34 - 4.27 (m, 1H), 2.39 - 2.33 (m, 1H), 2.25 - 2.20 (m, 3H), 2.03 - 1.93 (m, 4H), 1.60 - 1.55 (m, 2H), 1.46 (d, J = 9.6 Hz, 2H) Step C. Procedure for preparation of (R)-3-((1r,4R)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2-methylpropan-1-ol A mixture of NaH (1.13 g, 28.27 mmol, 60 % purity, 2.1 equiv.) in THF (100 mL) was purged with N 2 for three times. To the mixture was slowly added ethyl 2- diethoxyphosphorylpropanoate (6.41 g, 26.92 mmol, 5.88 mL, 2 equiv.) at 0 °C. The mixture was stirred for 2 hours under N2 atmosphere, treated with (1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexane-1-carbaldehyde (4.00 g, 13.46 mmol, 1 equiv.) at 0 °C, and warm to 25 °C followed by stirring for 3 hours under N2 atmosphere. The reaction mixture was quenched by addition saturated NH4Cl (100 mL) under 0 °C. The mixture was extracted with DCM (50 mL × 3). The combined organic layers were washed with brine (100 mL × 2), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~5 % ethyl acetate/petroleum ether) to give (R)-3-((1r,4R)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2-meth ylpropan-1-ol (1.1 g, 2.88 mmol, 21.43% yield) as a yellow solid. 1 H NMR (400 MHz, DMSO-d6) δ = 7.18 - 7.00 (m, 3H), 6.53 - 6.46 (m, 1H), 4.34 - 4.24 (m, 1H), 4.16 - 4.06 (m, 2H), 2.43 - 2.37 (m, 1H), 2.22 (s, 3H), 2.10 - 2.03 (m, 2H), 1.81 (s, 3H), 1.72 - 1.64 (m, 2H), 1.52 - 1.42 (m, 2H), 1.38 - 1.30 (m, 2H), 1.21 (br t, J = 7.2 Hz, 3H) Step D. Procedure for preparation of ethyl 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]- 2-methyl-propanoate To a solution of ethyl (E)-3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-2-methyl-prop -2- enoate (1 g, 2.62 mmol, 1 equiv.) in THF (15 mL) was added PtO2 (119.11 mg, 524.52 µmol, 0.2 equiv.) under H2 atmosphere (15 psi). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a little cake which was rinsed with EtOH. The filtrate was collected and concentrated it under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate 0/1 to 80/1) to give ethyl 3-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]-2-methyl-propanoate (950 mg, 2.48 mmol, 94.50% yield) as a yellow oil. 1 H NMR (400 MHz, CDCl3) δ = 7.14 (d, J = 8.0 Hz, 1H), 6.97 (t, J = 8.0 Hz, 1H), 6.87 - 6.76 (m, 1H), 4.20 - 4.03 (m, 3H), 2.58 - 2.47 (m, 1H), 2.36 - 2.20 (m, 3H), 2.18 - 2.08 (m, 2H), 1.94 - 1.86 (m, 1H), 1.84 - 1.76 (m, 1H), 1.69 - 1.61 (m, 1H), 1.50 - 1.38 (m, 2H), 1.36 - 1.24 (m, 5H), 1.15 (d, J = 6.8 Hz, 3H), 1.08 - 0.96 (m, 2H) Step E. Procedure for preparation of 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-2- methyl-propan-1-ol To a solution of ethyl 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-2-methyl-propanoa te (950 mg, 2.48 mmol, 1 equiv.) in THF (15 mL) was added LiAlH4 (94.05 mg, 2.48 mmol, 1 equiv.). The mixture was stirred at 0 °C for 2 hours under N2 atmosphere The reaction mixture was quenched by addition H 2 O (0.1 mL), 15% NaOH (0.1 mL), and H 2 O (0.3 mL), The combined mixture was mixed with Na2SO4, then filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 3-[4-(3-bromo-2- methyl-phenoxy)cyclohexyl]-2-methyl-propan-1-ol (280 mg, 814.4 µmol, 32.8% yield, 99.2% purity) as a yellow oil. MS (ESI) m/z: 342.2 [M+H] + . 1 H NMR (400 MHz, CDCl3) δ = 7.13 (d, J = 8.0 Hz, 1H), 6.96 (t, J = 8.0 Hz, 1H), 6.80 (d, J = 8.0 Hz, 1H), 4.10 - 4.04 (m, 1H), 3.55 - 3.48 (m, 1H), 3.46 - 3.39 (m, 1H), 2.29 (s, 3H), 2.16 - 2.09 (m, 2H), 1.90 - 1.77 (m, 3H), 1.77 - 1.68 (m, 1H), 1.48 - 1.36 (m, 4H), 1.08 - 0.96 (m, 3H), 0.92 (d, J = 6.4 Hz, 3H) Step F. Procedure for preparation of (2R)-3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]- 2-methyl-propan-1-ol The 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-2-methyl-propan-1 -ol residue was separated by prep-HPLC to give (R)-3-((1r,4R)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2- methylpropan-1-ol (100 mg, 293.01 µmol, 35.71% yield) and (2S)-3-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]-2-methyl-propan-1-ol (100 mg, 293.01 µmol, 35.71% yield) as a yellow oil. Step G. Procedure for preparation of (R)-3-((1r,4R)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2-methylpropanal A solution of oxalyl dichloride (74.38 mg, 586.03 µmol, 51.30 µL, 2 equiv.) in DCM (5 mL) was added dropwise to a solution of DMSO (91.57 mg, 1.17 mmol, 91.57 µL, 4 equiv.) in DCM (2 mL) at -70 °C under N 2 atmosphere. The mixture was stirred at -70 °C for 30 minutes. After which time (2R)-3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-2-methyl-pro pan-1-ol (100 mg, 293.01 µmol, 1 equiv.) in DCM (2 mL) was added dropwise at -70 °C. The solution was stirred for 30 minutes at -70 °C. Then TEA (177.90 mg, 1.76 mmol, 244.70 µL, 6 equiv.) was added into the solution. The solution was stirred at -70 °C for 2 hours under N2 atmosphere. The reaction was diluted with water (60 mL) and extracted with DCM (40 mL × 2). The combined organic layers were washed with brine (60 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give (R)-3-((1r,4R)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2-methylpropanal (90 mg, 265.28 µmol, 90.53% yield) as a yellow oil.. Step H. Procedure for preparation of 3-[6-[4-[(2R)-3-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]-2-methyl-propyl]piperazin-1-yl]-1-methyl -indazol-3-yl]piperidine- 2,6-dione To a solution of 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (84.92 mg, 259.38 µmol, 1.1 equiv.) in DCM (1 mL) was added NaBH(OAc) 3 (149.93 mg, 707.41 µmol, 3 equiv.), AcOH (28.32 mg, 471.61 µmol, 26.97 µL, 2 equiv.), and (2R)-3-[4-(3-bromo-2- methyl-phenoxy)cyclohexyl]-2-methyl-propanal (80 mg, 235.80 µmol, 1 equiv.) at 0 °C. The mixture was stirred at 25 °C for 1 hour. The reaction was diluted with water (40 mL) and extracted with DCM (25 mL × 2). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , DCM: MeOH = 20:1) to give 3-[6-[4-[(2R)-3- [4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-2-methyl-propyl]pip erazin-1-yl]-1-methyl- indazol-3-yl]piperidine-2,6-dione (65 mg, 99.90 µmol, 42.37% yield) as a white solid. MS (ESI) m/z: 650.4 [M+H] + . Step I. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[(2R)-3-[4-[3-(2,6-diox o-3-piperidyl)-1-methyl- indazol-6-yl]piperazin-1-yl]-2-methyl-propyl]cyclohexoxy]-2- methyl-phenyl]pyridine-2- carboxylate To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin e-2-carboxylate (60 mg, 97.95 µmol, 1 equiv.) in dioxane (2 mL) and H 2 O (0.5 mL) was added [2-(2- aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-butyl-phos phane;methanesulfonate (7.13 mg, 9.80 µmol, 0.1 equiv.), 3-[6-[4-[(2R)-3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]- 2-methyl-propyl]piperazin-1-yl]-1-methyl-indazol-3-yl]piperi dine-2,6-dione (63.73 mg, 97.95 µmol, 1 equiv.), and KF (17.07 mg, 293.86 µmol, 6.88 µL, 3 equiv.). The mixture was stirred at 100 °C for 1 hour. The reaction was diluted with water (20 mL) and extracted with DCM (10 mL × 2). The combined organic layers were washed with brine (30 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give as a residue. The residue was purified by column chromatography (SiO2, dichloromethane : methanol=50/1 to 20/1) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[(2R)-3-[4- [3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]piperazin-1 -yl]-2-methyl- propyl]cyclohexoxy]-2-methyl-phenyl]pyridine-2-carboxylate (50 mg, 47.33 µmol, 48.32% yield) as a yellow solid. MS (ESI) m/z: 1056.4 [M+H] + . Step J. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-((2R)-3-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-2-methylpropyl)cycloh exyl)oxy)-2- methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-((2R*)-3-(4-(3 -(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-2-methylpropyl)cycloh exyl)oxy)-2- methylphenyl)picolinic acid] To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[4-[(2R)-3-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl -indazol-6-yl]piperazin-1-yl]-2- methyl-propyl]cyclohexoxy]-2-methyl-phenyl]pyridine-2-carbox ylate (50 mg, 47.33 µmol, 1 equiv.) in DCM (0.5 mL) was added TFA (770.00 mg, 6.75 mmol, 0.5 mL, 142.67 equiv.). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1R,4r)-4-((2R)-3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1-yl)-2- methylpropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r) -4-((2R*)-3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin-1-yl )-2- methylpropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] (15.2 mg, 14.9 µmol, 31.6% yield, 98.6% purity) as a yellow solid. MS (ESI) m/z: 1000.4 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.91 - 12.80 (m, 1H), 10.85 (s, 1H), 8.04 (d, J = 8.0 Hz, 1H), 7.80 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 7.6 Hz, 1H), 7.53 - 7.49 (m, 1H), 7.49 - 7.43 (m, 3H), 7.41 - 7.33 (m, 2H), 7.11 - 7.05 (m, 1H), 6.99 - 6.86 (m, 4H), 6.62 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.29 - 4.18 (m, 2H), 3.92 (s, 2H), 3.90 (s, 3H), 3.23 (d, J = 5.2 Hz, 4H), 3.03 (t, J = 5.2 Hz, 2H), 2.66 - 2.55 (m, 8H), 2.35 - 2.30 (m, 1H), 2.18 (d, J = 6.0 Hz, 1H), 2.11 - 2.06 (m, 2H), 1.87 (s, 3H), 1.83 - 1.73 (m, 2H), 1.48 - 1.27 (m, 5H), 1.10 (d, J = 11.2 Hz, 1H), 1.05 - 0.97 (m, 2H), 0.94 - 0.86 (m, 3H) Example 185. Preparation of Compound 282b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1S,4r)-4- ((2S)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 6-yl)piperazin-1-yl)-2- methylpropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1S,4r)-4- ((2S*)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)piperazin-1-yl)-2- methylpropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of (S)-3-((1r,4S)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2-methylpropanal To a solution of oxalyl dichloride (74.38 mg, 586.03 µmol, 51.30 µL, 2 equiv.) in DCM (5 mL) was added a solution of DMSO (91.58 mg, 1.17 mmol, 91.58 µL, 4 equiv.) in DCM (2 mL) dropwise at -70 °C under N 2 atmosphere. The mixture was stirred at -70 °C for 1 hour. After which time (2S)-3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-2-methyl-pro pan-1-ol (100 mg, 293.01 µmol, 1 equiv.) in DCM (2 mL) was added dropwise at -70 °C. The solution was stirred for 1 hour at -70 °C. Then TEA (177.90 mg, 1.76 mmol, 244.70 µL, 6 equiv.) was added into the solution. The solution was stirred at -70 °C for 0.5 hour under N 2 atmosphere, after which water (30 mL) was added, and the reaction was extracted with EtOAc (10 mL × 2), dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum to give (S)-3-((1r,4S)- 4-(3-bromo-2-methylphenoxy)cyclohexyl)-2-methylpropanal (80 mg, crude) as a yellow oil. Step B. Procedure for preparation of 3-(6-(4-((S)-3-((1r,4S)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2-methylpropyl)piperazin-1-yl)-1-m ethyl-1H-indazol-3- yl)piperidine-2,6-dione To a solution of 3-(1-methyl-6-(piperazin-1-yl)-1H-indazol-3-yl)piperidine-2, 6-dione (90.08 mg, 247.59 µmol, 1.2 equiv., HCl) in DCM (2 mL) was added NaBH(OAc) 3 (131.19 mg, 618.98 µmol, 3 equiv.). (S)-3-((1r,4S)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2- methylpropanal (70 mg, 206.33 µmol, 1 equiv.) was then added into the mixture, which was stirred at 0 °C for 1 hour. Then the mixture was stirred at 25 °C for 15 hours. The mixture was filtered and concentrated under vacuum. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=4/1 to DCM: MeOH = 10:1) to give 3-(6-(4-((S)-3- ((1r,4S)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2-methylprop yl)piperazin-1-yl)-1- methyl-1H-indazol-3-yl)piperidine-2,6-dione (70 mg, 107.59 µmol, 52.14% yield) as a yellow solid. MS (ESI) m/z: 650.1 [M+H] + Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-((2S)-3-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-2-methylpropyl)cycloh exyl)oxy)-2- methylphenyl)picolinate A mixture of 3-(6-(4-((S)-3-((1r,4S)-4-(3-bromo-2-methylphenoxy)cyclohexy l)-2- methylpropyl)piperazin-1-yl)-1-methyl-1H-indazol-3-yl)piperi dine-2,6-dione (70 mg, 107.59 µmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pic olinate (79.08 mg, 129.10 µmol, 1.2 equiv.), H2O (0.1 mL), KF (18.75 mg, 322.76 µmol, 7.56 µL, 3 equiv.), and [2-(2- aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-butyl-phos phane;methanesulfonate (7.84 mg, 10.76 µmol, 0.1 equiv.) in dioxane (1 mL)was degassed and purged with N 2 and stirred at 100 °C for 1 hour under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0~25 % ethyl acetate/petroleum ether) to give tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r) -4-((2S)-3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin-1-yl )-2- methylpropyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (60 mg, 56.80 µmol, 52.80% yield) as a yellow solid. MS (ESI) m/z: 1056.7 [M+H] + Step D. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-((2S)-3-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-2-methylpropyl)cycloh exyl)oxy)-2- methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-((2S*)-3-(4-(3 -(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-2-methylpropyl)cycloh exyl)oxy)-2- methylphenyl)picolinic acid] To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1S,4r)-4-((2S)-3-(4-(3-(2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-6- yl)piperazin-1-yl)-2-methylpropyl)cyclohexyl)oxy)-2-methylph enyl)picolinate (60.00 mg, 56.80 µmol, 1 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1.00 mL, 237.78 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by prep-HPLC to give 6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2( 1H)-yl)-3-(3-(((1S,4r)-4-((2S)- 3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)p iperazin-1-yl)-2- methylpropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r) -4-((2S*)-3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin-1-yl )-2- methylpropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] (19.87 mg, 18.78 µmol, 33.07% yield, 98.90% purity, FA) was obtained as a yellow solid. MS (ESI) m/z: 1000.3 [M+H] + 1 H NMR (400 MHz, DMSO) δ = 13.04 - 12.72 (m, 1H), 12.69 - 12.33 (m, 1H), 10.86 (s, 1H), 8.14 (s, 1H), 8.04 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 6.8 Hz, 1H), 7.51 - 7.44 (m, 4H), 7.40 - 7.34 (m, 2H), 7.10 - 7.04 (m, 1H), 6.99 - 6.90 (m, 3H), 6.85 (s, 1H), 6.61 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.29 - 4.17 (m, 2H), 3.96 - 3.90 (m, 2H), 3.89 (s, 3H), 3.23 (s, 4H), 3.03 (t, J = 5.6 Hz, 2H), 2.65 - 2.59 (m, 2H), 2.55 - 2.53 (m, 2H), 2.36 - 2.24 (m, 2H), 2.21 - 2.05 (m, 6H), 1.87 (s, 3H), 1.86 - 1.70 (m, 4H), 1.44 - 1.32 (m, 4H), 1.04 - 0.96 (m, 2H), 0.88 (d, J = 6.4 Hz, 3H) Example 186. Preparation of Compound 285 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[1-[[1-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]-4-piperidyl]m ethyl]-4-piperidyl]butoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[4-[1-[[1-[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-7-yl]-4- piperidyl]methyl]-4-piperidyl]butoxy]-2-methyl-phenyl]pyridi ne-2-carboxylic acid To a solution of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- [2-methyl-3-[4-(4-piperidyl)butoxy]phenyl]pyridine-2-carboxy lic acid (158.28 mg, 234.20 µmol, 1 equiv.) in DCM (1 mL) and IPA (1 mL) was added 1-[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-7-yl]piperidine-4-carbaldehyde (83 mg, 234.20 µmol, 1 equiv.) at 25 °C for 11 hours. The mixture was stirred at this temperature, and then NaBH(OAc)3 (148.91 mg, 702.59 µmol, 3 equiv.) was added at 0 °C. The resulting mixture was stirred at 25 °C for 1 hour. The mixture was concentrated to give a residue. The residue was purified by reverse-phase HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[1- [[1-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]-4-pipe ridyl]methyl]-4- piperidyl]butoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid (27.7 mg, 26.2 µmol, 11.2% yield, 96.0% purity) as an off-white solid. MS (ESI) m/z: 508.1 [M/2+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.87 (s, 1H), 8.18 (s, 1H), 8.01 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.4 Hz, 1H), 7.62 (d, J = 6.8 Hz, 1H), 7.49 - 7.29 (m, 6H), 7.10 - 7.04 (m, 1H), 7.02 - 6.96 (m, 2H), 6.94 - 6.83 (m, 2H), 6.64 (d, J = 7.6 Hz, 1H), 4.96 (s, 2H), 4.33 (d, J = 5.2, 9.6 Hz, 1H), 4.23 (s, 3H), 3.97 (t, J = 6.0 Hz, 2H), 3.90 (t, J = 5.6 Hz, 2H), 3.21 - 3.19 (m, 2H), 3.00 (t, J = 5.6 Hz, 2H), 2.87 (dd, J = 2.0, 8.4 Hz, 2H), 2.68 - 2.61 (m, 4H), 2.33 (s, 1H), 2.23 (d, J = 3.6 Hz, 2H), 2.20 - 2.13 (m, 1H), 1.96 - 1.88 (m, 5H), 1.84 (d, J = 11.6 Hz, 2H), 1.76 - 1.66 (m, 3H), 1.65 - 1.59 (m, 2H), 1.48 - 1.40 (m, 2H), 1.38 - 1.21 (m, 5H), 1.19 - 1.07 (m, 2H). Example 187. Preparation of Compound 106 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[2-[1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-4-piperidyl]ethoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-6-nitro- indazole To a solution of (2,6-dibenzyloxy-3-pyridyl)boronic acid (4.5 g, 13.43 mmol, 1 equiv.) and 3- iodo-1-methyl-6-nitro-indazole (4.48 g, 14.77 mmol, 1.1 equiv.) in dioxane (45 mL) and H 2 O (4.5 mL) was added Na2CO3 (4.27 g, 40.28 mmol, 3 equiv.) and Pd(dppf)Cl2 (982.42 mg, 1.34 mmol, 0.1 equiv.). The mixture was stirred at 100 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=1/0 to 5/1). The compound 3- (2,6-dibenzyloxy-3-pyridyl)-1-methyl-6-nitro-indazole (3.8 g, 8.1 mmol, 60.6% yield) was obtained as a yellow solid. MS (ESI) m/z: 467.1 [M+H] + . Step B. Procedure for preparation of 3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6- amine A mixture of 3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-6-nitro-indazole (3.8 g, 8.15 mmol, 1 equiv.), Pd/C (900 mg, 10% purity), Pd(OH)2 (900 mg, 20% purity) in EtOH (40 mL) and THF (40 mL) was degassed and purged with H2 and stirred at 50 °C for 12 hours under H2 atmosphere (50 psi). The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=10/1 to 1/1). The compound 3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl- indazol-6-amine (1.7 g, 3.8 mmol, 47.8% yield) was obtained as a pink solid. MS (ESI) m/z: 437.1 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 7.86 (d, J = 8.4 Hz, 1H), 7.48 - 7.45 (m, 2H), 7.42 - 7.37 (m, 4H), 7.36 - 7.34 (m, 1H), 7.34 - 7.29 (m, 4H), 6.55 (d, J = 8.0 Hz, 1H), 6.42 - 6.37 (m, 2H), 5.42 (d, J = 10.4 Hz, 4H), 5.32 (s, 2H), 3.84 (s, 3H). Step C. Procedure for preparation of 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6- dione To a solution of 3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-amine (1.7 g, 3.89 mmol, 1 equiv.) in THF (15 mL) and EtOH (15 mL) was added Pd/C (500 mg, 10% purity), Pd(OH)2 (500 mg, 20% purity) and acetic acid (233.88 mg, 3.89 mmol, 222.74 µL, 1 equiv.). The mixture was stirred at 50 °C for 24 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, dichloromethane: methanol=100/1 to 10/1). The compound 3-(6-amino-1-methyl- indazol-3-yl)piperidine-2,6-dione (600 mg, 2.3 mmol, 59.6% yield) was obtained as a pink solid. MS (ESI) m/z: 259.1 [M+H] + 1 H NMR (400 MHz, CDCl 3 ) δ = 7.93 (s, 1H), 7.45 (d, J = 8.8 Hz, 1H), 6.58 (d, J = 8.8 Hz, 1H), 6.50 (s, 1H), 4.24 (t, J = 6.0 Hz, 1H), 3.89 (s, 3H), 3.03 - 2.95 (m, 1H), 2.70 - 2.62 (m, 1H), 2.54 - 2.45 (m, 1H), 2.40 - 2.31 (m, 1H). Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[1-[2-[[3-(2,6-diox o-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-4-piperidyl]ethoxy]-2-methy l-phenyl]pyridine-2- carboxylate To a solution of 2-[4-[2-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]ethyl]-1- piperidyl]acetic acid (100 mg, 131.25 µmol, 1 equiv.) and 3-(6-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (40.68 mg, 157.50 µmol, 1.2 equiv.) in DMF (1 mL) was added DIEA (50.89 mg, 393.74 µmol, 68.58 µL, 3 equiv.) and HATU (64.87 mg, 170.62 µmol, 1.3 equiv.). The mixture was stirred at 25 °C for 2 hours. To the reaction mixture was added H2O (10 mL), and then the reaction mixture was filtered. The residue was dissolved with DCM (10 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[2-[1-[2-[[3-(2,6-dioxo- 3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]-4-pip eridyl]ethoxy]-2-methyl- phenyl]pyridine-2-carboxylate (200 mg, crude) as a yellow solid. MS (ESI) m/z: 501.8 [M+H] + /2 Step E. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[2-[1-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]ethoxy]-2-methyl-phenyl]p yridine-2-carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[2-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-ind azol-6-yl]amino]-2-oxo-ethyl]-4- piperidyl]ethoxy]-2-methyl-phenyl]pyridine-2-carboxylate (105.70 mg, 105.47 µmol, 1 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 128.06 equiv.). The mixture was stirred at 40 °C for 2 hours. The reaction mixture filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[2-[1-[2-[[3-(2,6-dioxo- 3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]-4-pip eridyl]ethoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid (26 mg, 26.1 µmol, 24.7% yield, 95% purity) as a white solid. MS (ESI) m/z: 946.4 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.85 (s, 1H), 9.96 - 9.68 (m, 1H), 8.08 - 7.92 (m, 2H), 7.76 (d, J = 7.2 Hz, 1H), 7.80 - 7.64 (m, 2H), 7.68 – 7.32 (m, 6H), 7.24 - 7.16 (m, 1H), 7.12 - 7.04 (m, 1H), 6.96 (d, J = 8.4 Hz, 1H), 6.92 (d, J = 8.0 Hz, 1H), 6.60 (d, J = 7.2 Hz, 1H), 4.96 (s, 2H), 4.32 (dd, J = 9.2, 5.2 Hz, 1H), 4.04 (s, 2H), 3.92 (s, 5H), 3.24 - 3.08 (m, 1H), 3.06 - 3.01 (m, 3H), 2.65 - 2.57 (m, 2H), 2.28 - 2.12 (m, 4H), 2.08 - 1.96 (m, 1H), 1.92 (s, 3H), 1.76 - 1.68 (m, 4H), 1.60 - 1.52 (m, 1H), 1.48 - 1.29 (m, 3H). Example 188. Preparation of Compound 114 2-[5-[1-(1-adamantylmethyl)-5-methyl-pyrazol-4-yl]-6-[[8-[[3 -(2,6-dioxo-3-piperidyl)-1- methyl-indazol-7-yl]amino]-8-oxo-octyl]carbamoyl]-2-pyridyl] -N-(1,3-benzothiazol-2-yl)- 3,4-dihydro-1H-isoquinoline-8-carboxamide Step A. Procedure for preparation of methyl 8-[[3-[1-(1-adamantylmethyl)-5-methyl- pyrazol-4-yl]-6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihy dro-1H-isoquinolin-2- yl]pyridine-2-carbonyl]amino]octanoate A mixture of 3-[1-(1-adamantylmethyl)-5-methyl-pyrazol-4-yl]-6-[8-(1,3-be nzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]pyridine-2-carb oxylic acid (1 g, 1.52 mmol,1 equiv.), methyl 8-aminooctanoate (636.64 mg, 3.04 mmol, 2 equiv., HCl), and HATU (692.58 mg, 1.82 mmol, 1.2 equiv.) in DMF (16 mL) was degassed and purged with N 2 three times, and the mixture was stirred at 25 °C for 5 minutes. After 5 minutes, DIEA (588.53 mg, 4.55 mmol, 793.17 µL, 3 equiv.) was added, and the mixture was stirred at 25 °C for 1 hour under N2 atmosphere. The reaction mixture was diluted with water (40 mL) and extracted with DCM (50 mL × 3). The combined organic layers were washed with brine (30 mL × 4), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 10/1 to 1/1.5). The compound methyl 8-[[3-[1-(1-adamantylmethyl)-5-methyl-pyrazol-4-yl]-6- [8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinol in-2-yl]pyridine-2- carbonyl]amino]octanoate (710 mg, 846.0 µmol, 55.7% yield, 97% purity) was obtained as a yellow solid. MS (ESI) m/z: 814.3 [M+H] + Step B. Procedure for preparation of 8-[[3-[1-(1-adamantylmethyl)-5-methyl-pyrazol-4- yl]-6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-iso quinolin-2-yl]pyridine-2- carbonyl]amino]octanoic acid A mixture of methyl 8-[[3-[1-(1-adamantylmethyl)-5-methyl-pyrazol-4-yl]-6-[8-(1, 3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] pyridine-2- carbonyl]amino]octanoate (710 mg, 872.19 µmol, 1 equiv.) and LiOH•H2O (109.80 mg, 2.62 mmol, 3 equiv.) in THF (9.0 mL) and H 2 O (3.0 mL) was degassed and purged with N 2 and stirred at 40 °C for 6 hours under N2 atmosphere. The crude product was triturated with petroleum ether at 25 o C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The compound 8-[[3-[1-(1-adamantylmethyl)-5-methyl-pyrazol-4- yl]-6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-iso quinolin-2-yl]pyridine-2- carbonyl]amino]octanoic acid (500 mg, 593.7 µmol, 68.1% yield, 95% purity) was obtained as a yellow solid. MS (ESI) m/z: 800.4 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 13.01 - 11.96 (m, 1H), 8.06 (t, J = 5.6 Hz, 1H), 7.96 (d, J = 7=8.0 Hz, 1H), 7.74 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 6.4 Hz, 1H), 7.47 - 7.27 (m, 6H), 7.23 (s, 1H), 6.94 (d, J = 8.8 Hz, 1H), 5.02 (s, 2H), 3.88 (t, J = 6.0 Hz, 2H), 3.02-2.98 (m, 4H), 2.14 (t, J = 7.2 Hz, 2H), 2.05 (s, 3H), 1.91 (s, J = 5.2 Hz, 3H), 1.69 - 1.60 (m, 4H), 1.58 - 1.50 (m, 10H), 1.44 - 1.39 (m, 2H), 1.30 - 1.25 (m, 2H), 1.16 - 1.06 (m, 6H) Step C. Procedure for preparation of 2-[5-[1-(1-adamantylmethyl)-5-methyl-pyrazol-4- yl]-6-[[8-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl] amino]-8-oxo- octyl]carbamoyl]-2-pyridyl]-N-(1,3-benzothiazol-2-yl)-3,4-di hydro-1H-isoquinoline-8- carboxamide A mixture of 8-[[3-[1-(1-adamantylmethyl)-5-methyl-pyrazol-4-yl]-6-[8-(1, 3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]pyridine-2-carb onyl]amino]octanoic acid (40 mg, 50.00 µmol, 1 equiv.), 3-(7-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (15.50 mg, 60.00 µmol, 1.2 equiv.), and DIPEA (26 mg, 201.17 µmol, 35.04 µL, 4.02 equiv.) in DMF (0.5 mL) was degassed and purged with N2 three times. The mixture was stirred at 25°C for 5 minutes. After 5 minutes, HATU (22.81 mg, 60.00 µmol, 1.2 equiv.) was added to the mixture, and the mixture was stirred at 25 °C for 1 hour under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The crude product was purified by reverse-phase HPLC. The compound 2-[5-[1-(1-adamantylmethyl)-5-methyl- pyrazol-4-yl]-6-[[8-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-ind azol-7-yl]amino]-8-oxo- octyl]carbamoyl]-2-pyridyl]-N-(1,3-benzothiazol-2-yl)-3,4-di hydro-1H-isoquinoline-8- carboxamide (26.2 mg, 25.2 µmol, 50.4% yield, 100% purity) was obtained as a white solid. MS (ESI) m/z: 1040.6 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 12.97 - 12.69 (m, 1H), 10.89 (s, 1H), 9.80 (s, 1H), 8.09 - 8.03 (m, 1H), 8.03 - 7.97 (m, 1H), 7.77 (d, J = 7.6 Hz, 1H), 7.59 (t, J = 7.2 Hz, 2H), 7.49 - 7.42 (m, 2H), 7.41 - 7.37 (m, 1H), 7.37 - 7.30 (m, 2H), 7.23 (s, 1H), 7.12 - 7.03 (m, 2H), 6.94 (d, J = 8.4 Hz, 1H), 5.00 (s, 2H), 4.40 - 4.34 (m, 1H), 4.02 (s, 3H), 3.88 (t, J = 5.6 Hz, 2H), 3.67 (s, 2H), 3.04 - 2.97 (m, 4H), 2.69 - 2.64 (m, 2H), 2.37 - 2.33 (m, 4H), 2.20 - 2.15 (m, 1H), 2.05 (s, 3H), 1.91 (s, 3H), 1.68 - 1.60 (m, 4H), 1.56 (s, 4H), 1.51 (s, 3H), 1.33 - 1.06 (m, 10H) Example 189. Preparation of Compound 240a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[(3R)-1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-o xo-ethyl]pyrrolidin-3- yl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(3R)-1-[2-[[3-(2,6 -dioxo-3-piperidyl)-1-methyl- indazol-7-yl]amino]-2-oxo-ethyl]pyrrolidin-3-yl]propoxy]-2-m ethyl-phenyl]pyridine-2- carboxylate A mixture of 2-[(3R)-3-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]propyl]pyrrolidin-1- yl]acetic acid (120 mg, 157.50 µmol, 1 equiv.), 3-(7-amino-1-methyl-indazol-3-yl)piperidine- 2,6-dione (48.81 mg, 188.99 µmol, 1.2 equiv.), DIPEA (61.06 mg, 472.49 µmol, 82.30 µL, 3 equiv.), and HATU (59.88 mg, 157.50 µmol, 1.5 equiv.) in DMF (3 mL) was degassed and purged with N 2 and stirred at 40 °C for 6 hours under N 2 atmosphere. The reaction mixture was treated with water (3 mL) and then filtered to give a residue which was dried under reduced pressure. The residue was used in the next step without purification. The compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[(3R)-1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-o xo-ethyl]pyrrolidin-3- yl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate (150 mg, crude) was obtained as a white solid. MS (ESI) m/z: 1002.7 [M+H] + Step B. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[(3R)-1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-7- yl]amino]-2-oxo-ethyl]pyrrolidin-3-yl]propoxy]-2-methyl-phen yl]pyridine-2-carboxylic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[(3R)-1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl- indazol-7-yl]amino]-2-oxo- ethyl]pyrrolidin-3-yl]propoxy]-2-methyl-phenyl]pyridine-2-ca rboxylate (150.00 mg, 149.67 µmol, 1 equiv.) in TFA (3 mL) and DCM (3 mL) was degassed and purged with N2 and stirred at 25 °C for 3 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[3-[3-[(3R)-1-[2-[[3-(2,6- dioxo-3-piperidyl)-1-methyl-indazol-7-yl]amino]-2-oxo-ethyl] pyrrolidin-3-yl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid (30.8 mg, 31.8 µmol, 21.2% yield, 97.8% purity) as a white solid, MS (ESI) m/z: 946.4 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.90 - 12.82 (m, 1H), 10.90 (s, 1H), 10.02 - 9.94 (m, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.61 (d, J = 16.8 Hz, 2H), 7.49 - 7.42 (m, 3H), 7.40 - 7.33 (m, 2H), 7.27 (d, J = 7.6 Hz, 1H), 7.11 - 7.06 (m, 2H), 6.97 - 6.95 (m, 1H), 6.89 (d, J = 8.4 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.39 - 4.36 (m, 1H), 4.08 (s, 3H), 4.01 - 3.96 (m, 2H), 3.93 - 3.91 (m, 2H), 3.61 - 3.41 (m, 4H), 3.04 - 3.02 (m, 2H), 2.96 - 2.90 (m, 1H), 2.69 - 2.60 (m, 2H), 2.42 - 2.31 (m, 2H), 2.28 - 2.14 (m, 2H), 2.08 - 2.00 (m, 1H), 1.90 (s, 3H), 1.76 - 1.74 (m, 2H), 1.60 - 1.52 (m, 2H), 1.51 - 1.42 (m, 1H). Example 190. Preparation of Compound 218a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((3R)-4-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino )-2-oxoethyl)-3- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3R)-4-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-2-oxoethyl)-3-(trifluoromethyl)pipera zin-1-yl)propoxy)-2- methylphenyl)picolinate A mixture of (R)-2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)-2- (trifluoromethyl)piperazin-1-yl)acetic acid (75 mg, 88.76 µmol, 1 equiv.), 3-(7-amino-1- methyl-1H-indazol-3-yl)piperidine-2,6-dione (27.51 mg, 106.52 µmol, 1.2 equiv.), and DIEA (34.42 mg, 266.29 µmol, 46.38 µL, 3 equiv.) in DMF (1 mL) was degassed and purged with N2 and stirred at 25°C for 5 minutes. After 5 minutes, HATU (40.50 mg, 106.52 µmol, 1.2 equiv.) was added, and the mixture was stirred at 25 °C for 1 hour under N2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (20 mL × 3). The combined organic layers were washed with brine (20 mL × 6), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-((3R)-4-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino)-2- oxoethyl)-3- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linate(100 mg, crude) as a brown solid. MS (ESI) m/z: 1085.8 [M+H] + Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((3R)-4-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)amino)-2-oxoethyl)-3-(trifluoromethyl)pipera zin-1-yl)propoxy)-2- methylphenyl)picolinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-((3R)-4-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-7-yl)amino)-2- oxoethyl)-3-(trifluoromethyl)piperazin-1-yl)propoxy)-2-methy lphenyl)picolinate (100 mg, 92.15 µmol, 1 equiv.) in TFA (0.5 mL) and DCM (0.5 mL) was degassed and purged with N2 and stirred at 25 °C for 1 hour under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-((3R)-4-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino)-2- oxoethyl)-3- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linic acid (29.2 mg, 26.6 µmol, 28.9% yield, 93.9% purity) as an off-white solid. MS (ESI) m/z: 1029.4 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.03 - 12.32 (m, 2H), 10.90 (s, 1H), 9.79 (s, 1H), 8.14 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.65 - 7.57 (m, 2H), 7.50 - 7.41 (m, 3H), 7.40 - 7.32 (m, 2H), 7.25 - 7.19 (m, 1H), 7.12 - 7.04 (m, 2H), 6.96 (d, J = 8.8 Hz, 1H), 6.86 (d, J = 8.0 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.38 (dd, J = 10.0, 5.2 Hz, 1H), 4.08 (s, 3H), 4.01 (t, J = 5.6 Hz, 2H), 3.91 (t, J = 5.6 Hz, 2H), 3.79 - 3.67 (m, 2H), 3.51 -3.44 (m, 1H), 3.02 (t, J = 5.6 Hz, 3H), 2.87 - 2.75 (m, 2H), 2.75 - 2.62 (m, 2H), 2.61 - 2.53 (m, 3H), 2.48 - 2.41 (m, 2H), 2.40 - 2.31 (m, 1H), 2.21 - 2.12 (m, 1H), 1.96 - 1.85 (m, 5H). Example 191. Preparation of Compound 161a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinate A mixture of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[2- methyl-3-[4-(4-oxobutyl)cyclohexoxy]phenyl]pyridine-2-carbox ylic acid (80 mg, 116.14 µmol, 1 equiv.), 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (38.02 mg, 116.14 µmol, 1 equiv.), and AcOH (6.97 mg, 116.14 µmol, 6.65 µL, 1 equiv.) in DCM (1 mL) was stirred at 25 °C for 1 hour. NaBH(OAc)3 (73.84 mg, 348.42 µmol, 3 equiv.) was added, and then the mixture was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy)-2-methy lphenyl)picolinate (100 mg, crude) as a yellow solid. It was used in the next step without further purification. MS (ESI) m/z: 529.0 [M+H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (100 mg, 94.67 µmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL) was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by reverse-phase HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4- (4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl) piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (27.8 mg, 26.0 µmol, 27.5% yield, 93.6% purity) as a white solid. MS (ESI) m/z: 1000.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.94 (s, 1H), 8.27 (s, 1H), 8.11 (d, J = 7.8 Hz, 1H), 7.87 (d, J = 8.0 Hz, 1H), 7.71 (d, J = 7.2 Hz, 1H), 7.60 - 7.40 (m, 6H), 7.18 - 7.11 (m, 1H), 7.05 - 6.97 (m, 3H), 6.92 (s, 1H), 6.71 (d, J = 7.6 Hz, 1H), 5.06 (s, 2H), 4.37 - 4.24 (m, 2H), 3.97 (s, 3H), 3.30 ( s, 6H), 3.11 ( t, J = 5.4 Hz, 3H), 2.72 - 2.66 (m, 2H), 2.48 - 2.32 (m, 4H), 2.29 - 2.05 (m, 4H), 1.96 (s, 3H), 1.86 ( d, J = 11.4 Hz, 2H), 1.56 - 1.29 (m, 10H), 1.20 - 1.07 (m, 2H) Example 192. Preparation of Compound 184 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4-[3-[1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-4-piperidyl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 4-[3-(4-bromo-3-methyl- phenoxy)propyl]piperidine-1-carboxylate A mixture of tert-butyl 4-(3-bromopropyl)piperidine-1-carboxylate (4.00 g, 13.06 mmol, 1 equiv.), 4-bromo-3-methylphenol (3.66 g, 19.59 mmol, 1.5 equiv.) and K 2 CO 3 (5.42 g, 39.19 mmol, 3.0 equiv.) in CH 3 CN (20 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 60 °C for 12 hours under N2 atmosphere. The reaction was diluted with water (100 mL) and extracted with ethyl acetate (100 mL × 2). The combined organic layers were washed with brine (100 mL × 1), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give tert-butyl 4-[3-(4-bromo-3-methyl-phenoxy)propyl]piperidine- 1-carboxylate (6.7 g, crude) as a colorless oil. MS (ESI) m/z: 312.2 [M+H-100] + . Step B. Procedure for preparation of 4-[3-(4-bromo-3-methyl-phenoxy)propyl]piperidine A mixture of tert-butyl 4-[3-(4-bromo-3-methyl-phenoxy)propyl]piperidine-1-carboxyla te (6.7 g, 16.25 mmol, 1.0 equiv.) in HCl/dioxane (10 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~4% dichloromethane: methanol ether) to give 4-[3-(4-bromo- 3-methyl-phenoxy)propyl]piperidine (2.5 g, 8.0 mmol, 49.2% yield) as a white solid. Step C. Procedure for preparation of ethyl 2-[4-[3-(4-bromo-3-methyl-phenoxy)propyl]- 1-piperidyl]acetate A mixture of 4-[3-(4-bromo-3-methyl-phenoxy)propyl]piperidine (2.5 g, 7.17 mmol, 1.0 equiv., HCl), ethyl 2- bromoacetate (1.20 g, 7.17 mmol, 792.91 µL, 1 equiv.), K2CO3 (990.88 mg, 7.17 mmol, 1.0 equiv.), and KI (238.02 mg, 1.43 mmol, 0.2 equiv.) in CH 3 CN (60 mL) was degassed and purged with N 2 three times. The mixture was then stirred at 25 °C for 1 hour under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~18% ethyl acetate/petroleum ether) to give ethyl 2-[4-[3-(4-bromo-3-methyl-phenoxy)propyl]-1- piperidyl]acetate (2.3 g, 5.7 mmol, 79.9% yield, 99.3% purity) as yellow oil. MS (ESI) m/z: 398.2 [M+H] + . Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[3-[1-(2-ethoxy2-oxo-e thyl)-4-piperidyl]propoxy]- 2-methyl-phenyl]pyridine-2-carboxylate A mixture of ethyl 2-[4-[3-(4-bromo-3-methyl-phenoxy)propyl]-1-piperidyl]acetat e (2.3 g, 5.77 mmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyridine-2-carboxylate (3.18 g, 5.20 mmol, 0.9 equiv.), K 2 CO 3 (1.5 M, 11.55 mL, 3.0 equiv.), and Ad2nBuP Pd G3 (841.01 mg, 1.15 mmol, 0.2 equiv.) in dioxane (20 mL) was degassed and purged with N 2 three times. The mixture was then stirred at 80 °C for 1 hour under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~59% ethyl acetate/petroleum ether) to give tert- butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4-[3-[1-(2- ethoxy2-oxo-ethyl)-4-piperidyl]propoxy]-2-methyl-phenyl]pyri dine-2-carboxylate (3.2 g, 3.9 mmol, 68.9% yield) as a yellow oil. MS (ESI) m/z: 804.5 [M+H] + . Step E. Procedure for preparation of 2-[4-[3-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyr idyl]-3-methyl- phenoxy]propyl]-1-piperidyl]acetic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[4-[3-[1-(2-ethoxy2-oxo-ethyl)-4-piperidyl]propoxy]-2- methyl-phenyl]pyridine-2- carboxylate (3.2 g, 3.98 mmol, 1.0 equiv.) and LiOH•H2O (1 M, 11.94 mL, 3.0 equiv.) in THF (20 mL) was degassed and purged with N2 three times. The mixture was then stirred at 25 °C for 2 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. Then HCl (1 M) was added to the residue to adjust the pH to 4-5. A white precipitate formed, and the solid was collected by filtration and dried to give 2-[4-[3-[4- [6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoqui nolin-2-yl]-2-tert- butoxycarbonyl-3-pyridyl]-3-methyl-phenoxy]propyl]-1-piperid yl]acetic acid (2.6 g, 3.2 mmol, 82.7% yield, 98.3% purity) as a white solid. MS (ESI) m/z: 776.5 [M+H] + . Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(3-(1-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)propoxy)-2-met hylphenyl)picolinate 2-[4-[3-[4-[6-[8-(1,3-Benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H-isoquinolin-2-yl]-2-tert- butoxycarbonyl-3-pyridyl]-3-methyl-phenoxy]propyl]-1-piperid yl]acetic acid (50 mg, 64 µmol, 1 equiv.) was dissolved in DCM (5 mL). To the mixture was added HATU (27 mg, 71 μmol, 1.1 equiv.), followed by 3-(6-amino-1-methyl-1H-indazol-3-yl)piperidine-2,6-dione (20 mg, 77 μmol, 1.2 equiv.) and DIEA (25 mg, 34μL, 0.19 mmol, 3 equiv.). The mixture was then stirred at 25 o C overnight. The solvents were removed. The resulting residue was used in the next step without further purification. Step G. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[4-[3-[1-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]propoxy]-2-methyl-phenyl] pyridine-2-carboxylic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(3-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-meth yl-1H-indazol-6-yl)amino)-2- oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinate (65 mg, 64 µmol, 1 equiv.) in DCM (4 mL) was added TFA (1 mL). The reaction mixture was stirred at 25 °C over night. The solvents were then removed, and the residue was purified by prep-HPLC to give 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[4-[3-[1-[2-[[3-(2,6-dioxo- 3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]-4-pip eridyl]propoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid (32 mg, 33 μmol, 52 % yield) as a white solid. MS (ESI) m/z: 960 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.45 - 12.85 (m, 2H), 10.88 (s, 1H), 8.03 (d, J = 6.4 Hz, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.66 - 7.59 (m, 2H), 7.50 - 7.41 (m, 3H), 7.40 - 7.32 (m, 2H), 7.20 (d, J = 8.8 Hz, 1H), 6.97 - 6.80 (m, 2H), 6.78 (s, 1H), 6.75-6.70 (m, 1H), 4.97 (s, 2H), 4.32 (dd, J = 5.2, 9.6 Hz, 1H), 3.97 - 3.89 (m, 7H), 3.02 (t, J = 6.4 Hz, 2H), 2.65 - 2.63 (m, 2H), 2.51 - 2.48 (m, 4H), 2.38 - 2.29 (m, 2H), 2.20 - 2.12 (m, 1H), 2.03 (s, 3H), 1.76 - 1.73 (m, 4H), 1.39 (s, 5H) Example 193. Preparation of Compound 262b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((2R)-8-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2- oxoethyl)-8- azaspiro[4.5]decan-2-yl)methoxy)-2-methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(3-(((2R)-8- (2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)am ino)-2-oxoethyl)-8- azaspiro[4.5]decan-2-yl)methoxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of tert-butyl 3-[(3-bromo-2-methyl-phenoxy)methyl]- 8-azaspiro[4.5]decane-8-carboxylate A mixture of tert-butyl 3-(hydroxymethyl)-8-azaspiro[4.5]decane-8-carboxylate (1 g, 3.71 mmol, 1.2 equiv.), 3-bromo-2-methyl-phenol (578.60 mg, 3.09 mmol, 1 equiv.), and 2- (tributyl-λ5-phosphanylidene)acetonitrile (1.49 g, 6.19 mmol, 2 equiv.) in toluene (5 mL) was stirred at 120 °C for 12 hours under N2. The mixture was concentrated to remove toluene. The residue was purified by flash silica gel chromatography (eluent of 0 ~ 20% ethyl acetate/petroleum ether) to give tert-butyl 3-[(3-bromo-2-methyl-phenoxy)methyl]-8- azaspiro[4.5]decane-8-carboxylate (1.1 g, 2.3 mmol, 74.6% yield, 92% purity) as a colorless oil. 1 H NMR (400 MHz, CDCl 3 -d) δ = 7.13 (d, J = 8.0 Hz, 1H), 6.98 (t, J = 8.0 Hz, 1H), 6.74 (d, J = 8.0 Hz, 1H), 3.85 (d, J = 6.4 Hz, 2H), 3.43 - 3.33 (m, 4H), 2.55 - 2.45 (m, 1H), 2.34 - 2.30 (m, 3H), 1.95 - 1.78 (m, 2H), 1.62 - 1.50 (m, 6H), 1.46 (s, 9H), 1.30 - 1.19 (m, 2H) Step B. Procedure for preparation of tert-butyl (3R)-3-[(3-bromo-2-methyl- phenoxy)methyl]-8-azaspiro[4.5]decane-8-carboxylate The stereoisomers of tert-butyl 3-[(3-bromo-2-methyl-phenoxy)methyl]-8- azaspiro[4.5]decane-8-carboxylate were separated by SFC to give tert-butyl (3S)-3-[(3-bromo- 2-methyl-phenoxy)methyl]-8-azaspiro[4.5]decane-8-carboxylate (peak 1) (400 mg, 912.41 µmol, 36.36% yield), tert-butyl (3R)-3-[(3-bromo-2-methyl-phenoxy)methyl]-8- azaspiro[4.5]decane-8-carboxylate (peak 2) (300 mg, 684.3 µmol, 27.2% yield) was obtained as a white oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.17 - 7.12 (m, 1H), 7.11 - 7.05 (m, 1H), 6.95 (d, J = 8.0 Hz, 1H), 3.88 (d, J = 6.4 Hz, 2H), 3.28 (d, J = 5.2 Hz, 4H), 2.47 - 2.41 (m, 1H), 2.23 (s, 3H), 1.84 (d, J = 8.0 Hz, 1H), 1.76 (dd, J = 8.0, 12.8 Hz, 1H), 1.53 - 1.45 (m, 3H), 1.38 (s, 13H), 1.19 (dd, J = 9.6, 12.4 Hz, 1H) 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.18 - 7.12 (m, 1H), 7.11 - 7.04 (m, 1H), 6.96 (d, J = 8.0 Hz, 1H), 3.88 (d, J = 6.4 Hz, 2H), 3.31 - 3.26 (m, 4H), 2.47 - 2.41 (m, 1H), 1.89 - 1.81 (m, 1H), 1.80 - 1.72 (m, 1H), 1.54 - 1.46 (m, 3H), 1.41 - 1.34 (m, 13H), 1.19 (dd, J = 9.2, 12.8 Hz, 1H) Step C. Procedure for preparation of (3R)-3-[(3-bromo-2-methyl-phenoxy)methyl]-8- azaspiro[4.5]decane A solution of tert-butyl (3R)-3-[(3-bromo-2-methyl-phenoxy)methyl]-8-azaspiro[4.5]dec ane- 8-carboxylate (300 mg, 684.31 µmol, 1 equiv.) in HCl / dioxane (1.5 mL) and DCM (1.5 mL) was stirred at 25 °C for 0.5 hours. The reaction mixture was concentrated under reduced pressure to give (3R)-3-[(3-bromo-2-methyl-phenoxy)methyl]-8-azaspiro[4.5]dec ane (260 mg, crude) as a white solid. MS (ESI) m/z: 337.8 [M+H] + . Step D. Procedure for preparation of ethyl 2-[(3R)-3-[(3-bromo-2-methyl- phenoxy)methyl]-8-azaspiro[4.5]decan-8-yl]acetate A mixture of (3R)-3-[(3-bromo-2-methyl-phenoxy)methyl]-8-azaspiro[4.5]dec ane (260 mg, 693.81 µmol, 1 equiv. HCl), ethyl 2-bromoacetate (104.28 mg, 624.43 µmol, 69.06 uL, 0.9 equiv.), KI (11.52 mg, 69.38 µmol, 0.1 equiv.), and K 2 CO 3 (287.67 mg, 2.08 mmol, 3 equiv.) in CH3CN (2.6 mL) was stirred at 60 °C for 0.5 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0 ~ 22% ethyl acetate / petroleum ether) to give ethyl 2-[(3R)-3-[(3- bromo-2-methyl-phenoxy)methyl]-8-azaspiro[4.5]decan-8-yl]ace tate (220 mg, 508.0 µmol, 73.2% yield, 98% purity) as a colorless oil. MS (ESI) m/z: 424.2 [M+H] + . 1 H NMR (400 MHz, CDCl3) δ = 7.14 (d, J = 8.0 Hz, 1H), 6.99 (t, J = 8.0 Hz, 1H), 6.75 (d, J = 8.0 Hz, 1H), 4.20 (q, J = 7.2 Hz, 2H), 3.89 - 3.81 (m, 2H), 3.25 (s, 2H), 2.58 (d, J = 1.6 Hz, 4H), 2.49 (s, 1H), 2.32 (s, 3H), 1.91 (d, J = 6.0 Hz, 1H), 1.82 (dd, J = 8.0, 12.8 Hz, 1H), 1.60 - 1.45 (m, 7H), 1.29 (t, J = 7.2 Hz, 3H), 1.23 (dd, J = 9.6, 13.2 Hz, 1H) Step E. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[[(3R)-8-(2-ethoxy-2-o xo-ethyl)-8-azaspiro[4.5] decan-3-yl]methoxy]-2-methyl-phenyl]pyridine-2-carboxylate Ethyl 2-[(3R)-3-[(3-bromo-2-methyl-phenoxy)methyl]-8-azaspiro[4.5] decan-8-yl]acetate (120 mg, 282.77 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (207.85 mg, 339.33 µmol, 1.2 equiv.), [2-(2-aminophenyl)phenyl]palladium(1+);bis(1- adamantyl)-butyl-phosphane;methanesulfonate (20.59 mg, 28.28 µmol, 0.1 equiv.), and KF (49.29 mg, 848.32 µmol, 19.87 uL, 3 equiv.) were taken up into a microwave tube in 1,4- dioxane (2.3 mL) and H 2 O (0.2 mL). The sealed tube was heated at 100 °C for 1 hour under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0 ~ 13 % dichloromethane : methanol) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[[(3R)-8-(2-ethoxy-2-oxo-e thyl)-8-azaspiro[4.5] decan-3- yl]methoxy]-2-methyl-phenyl]pyridine-2-carboxylate (260 mg, crude) as a yellow oil. Step F. Procedure for preparation of 2-[(3R)-3-[[3-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-2- methyl-phenoxy]methyl]-8-azaspiro[4.5]decan-8-yl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[[(3R)-8-(2-ethoxy-2-oxo-ethyl)-8-azaspiro[4.5] decan-3-yl]methoxy]-2-methyl- phenyl]pyridine-2-carboxylate (260 mg, 313.24 µmol, 1 equiv.) in THF (3 mL) and H2O (1 mL) was added LiOH•H2O (65.72 mg, 1.57 mmol, 5 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was treated with water (6 mL), and its pH was adjusted to 3 by progressively adding diluted HCl (1M). The resulting mixture was filtered to give 2-[(3R)-3-[[3-[6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -2-tert-butoxycarbonyl-3- pyridyl]-2-methyl-phenoxy]methyl]-8-azaspiro[4.5]decan-8-yl] acetic acid (170 mg, 165.1 µmol, 52.7% yield, 77.9% purity) as a white solid. MS (ESI) m/z: 802.3 [M+H] + . Step G. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[[(3R)-8-[2-[[3-(2,6-d ioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-8-azaspiro[4.5]decan-3-yl]m ethoxy]-2-methyl- phenyl]pyridine-2-carboxylate To a solution of 2-[(3R)-3-[[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dih ydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]methyl]-8- azaspiro[4.5]decan-8-yl]acetic acid (170 mg, 211.97 µmol, 1 equiv.), and 3-(6-amino-1- methyl-indazol-3-yl)piperidine-2,6-dione (65.70 mg, 254.37 µmol, 1.2 equiv.) in DMF (1.7 mL) was added HATU (120.90 mg, 317.96 µmol, 1.5 equiv.) and DIEA (82.19 mg, 635.92 µmol, 110.76 µL, 3 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was diluted with water (10 mL) and filtered to give tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[[(3R)-8- [2-[[3-(2,6-dioxo-3-piperidyl)- 1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]-8-azaspiro[4.5]dec an-3-yl]methoxy]-2-methyl- phenyl]pyridine-2-carboxylate (170 mg, 117.4 µmol, 55.4% yield, 72% purity) as a pink solid. MS (ESI) m/z: 521.9 [1/2M+H] + Step H. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((2R)-8-(2-((3-(2,6-dioxo piperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)-8-azaspiro[4.5]decan-2-yl)me thoxy)-2- methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((2R)-8-(2-((3-(2,6-dioxo piperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)-8-azaspiro[4.5]decan-2-yl)me thoxy)-2- methylphenyl)picolinic acid] A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[[(3R)-8-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-in dazol-6-yl]amino]-2-oxo-ethyl]- 8-azaspiro[4.5]decan-3-yl]methoxy]-2-methyl-phenyl]pyridine- 2-carboxylate (170.00 mg, 163.11 µmol, 1 equiv.) in TFA (1 mL) and DCM (2 mL) was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((2R)-8-(2-((3-(2,6-dioxopiperidin-3-yl)-1-m ethyl-1H-indazol-6-yl)amino)- 2-oxoethyl)-8-azaspiro[4.5]decan-2-yl)methoxy)-2-methylpheny l)picolinic acid [rel-6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((2R)-8-(2-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2-oxoet hyl)-8-azaspiro[4.5]decan-2- yl)methoxy)-2-methylphenyl)picolinic acid] (36.2 mg, 34.4 µmol, 21.1% yield, 93.9% purity) was obtained as an off-white solid. MS (ESI) m/z: 986.4 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.87 (s, 1H), 9.87 - 9.80 (m, 1H), 8.16 (s, 1H), 8.06 - 8.00 (m, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 8.4 Hz, 2H), 7.49 - 7.41 (m, 3H), 7.36 (d, J = 7.6, 10.2 Hz, 2H), 7.22 (dd, J = 1.6, 8.8 Hz, 1H), 7.11 - 7.05 (m, 1H), 6.95 (d, J = 8.8 Hz, 1H), 6.87 (d, J = 8.4 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.32 (dd, J = 5.2, 9.6 Hz, 1H), 3.92 (s, 4H), 3.91 - 3.83 (m, 3H), 3.13 (s, 4H), 3.02 (t, J = 5.6 Hz, 2H), 2.74 - 2.57 (m, 3H), 2.56 (s, 2H), 2.38 - 2.30 (m, 1H), 2.21 - 2.12 (m, 1H), 1.90 (s, 3H), 1.87 - 1.72 (m, 2H), 1.60 - 1.46 (m, 7H), 1.26 - 1.20 (m, 1H) Example 194. Preparation of Compound 262a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[[(3S)-8-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-o xo-ethyl]-8-azaspiro[4.5]decan- 3-yl]methoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(3-(((2S)-8-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino )-2-oxoethyl)-8- azaspiro[4.5]decan-2-yl)methoxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of (3S)-3-[(3-bromo-2-methyl-phenoxy)methyl]-8- azaspiro[4.5]decane A solution of tert-butyl (3S)-3-[(3-bromo-2-methyl-phenoxy)methyl]-8-azaspiro[4.5]dec ane- 8-carboxylate (400 mg, 912.41 µmol, 1 equiv.)in HCl / dioxane (2 mL) and DCM (2 mL) was stirred at 25 °C for 0.5 hours. The reaction mixture was concentrated under reduced pressure to give (3S)-3-[(3-bromo-2-methyl-phenoxy)methyl]-8-azaspiro[4.5]dec ane (320 mg, crude) as a white solid. MS (ESI) m/z: 338.0 [M+H] + . Step B. Procedure for preparation of ethyl 2-[(3S)-3-[(3-bromo-2-methyl- phenoxy)methyl]-8-azaspiro[4.5]decan-8-yl]acetate A mixture of (3S)-3-[(3-bromo-2-methyl-phenoxy)methyl]-8-azaspiro[4.5]dec ane (320 mg, 853.92 µmol, 1 equiv. HCl.), ethyl 2-bromoacetate (128.34 mg, 768.53 µmol, 85.00 µL, 0.9 equiv.), KI (14.18 mg, 85.39 µmol, 0.1 equiv.), and K2CO3 (354.05 mg, 2.56 mmol, 3 equiv.) in CH 3 CN (3.2 mL) was stirred at 60 °C for 2.5 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0 ~ 22% ethyl acetate / petroleum ether) to give ethyl 2-[(3S)-3-[(3- bromo-2-methyl-phenoxy)methyl]-8-azaspiro[4.5]decan-8-yl]ace tate (230 mg, 536.6 µmol, 62.8% yield, 99% purity) was obtained as a colorless oil. MS (ESI) m/z: 426.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 7.16 - 7.12 (m, 1H), 7.11 - 7.05 (m, 1H), 6.95 (d, J = 8.0 Hz, 1H), 4.10 - 4.03 (m, 2H), 3.92 - 3.83 (m, 2H), 3.16 (s, 2H), 2.43 (d, J = 5.2, 10.4 Hz, 4H), 2.23 (s, 3H), 1.84 - 1.77 (m, 1H), 1.72 (dd, J = 8.0, 12.8 Hz, 1H), 1.50 - 1.38 (m, 7H), 1.20 - 1.14 (m, 4H) Step C. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[[(3S)-8-(2-ethoxy-2-o xo-ethyl)-8- azaspiro[4.5]decan-3-yl]methoxy]-2-methyl-phenyl]pyridine-2- carboxylate Ethyl 2-[(3S)-3-[(3-bromo-2-methyl-phenoxy)methyl]-8-azaspiro[4.5] decan-8-yl]acetate (230 mg, 541.98 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (398.38 mg, 650.37 µmol, 1.2 equiv.), [2-(2-aminophenyl)phenyl]palladium(1+);bis(1- adamantyl)-butyl-phosphane;methanesulfonate (39.47 mg, 54.20 µmol, 0.1 equiv.), and KF (94.46 mg, 1.63 mmol, 38.09 µL, 3 equiv.) were taken up into a microwave tube in 1,4-dioxane (2.3 mL) and H2O (0.2 mL). The sealed tube was heated at 100 °C for 1 hour under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0 ~ 13 % dichloromethane : methanol) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[[(3S)-8-(2-ethoxy-2-oxo-ethyl)-8-aza spiro[4.5]decan-3-yl]methoxy]- 2-methyl-phenyl]pyridine-2-carboxylate (480 mg, 414.6 µmol, 76.5% yield, 71.7% purity) as a yellow oil. MS (ESI) m/z: 426.0 [M+H] + . Step D. Procedure for preparation of 2-[(3S)-3-[[3-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-2- methyl-phenoxy]methyl]-8-azaspiro[4.5]decan-8-yl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[[(3S)-8-(2-ethoxy-2-oxo-ethyl)-8-azaspiro[4.5]de can-3-yl]methoxy]-2-methyl- phenyl]pyridine-2-carboxylate (200.00 mg, 240.95 µmol, 1 equiv.) in THF (1 mL) and H2O (0.3 mL) was added LiOH•H 2 O (50.56 mg, 1.20 mmol, 5 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was treated with water (6 mL). The pH was adjusted to around 3 by progressively adding dilute HCl. The mixture was filtered to give 2-[(3S)-3-[[3-[6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -2-tert-butoxycarbonyl-3- pyridyl]-2-methyl-phenoxy]methyl]-8-azaspiro[4.5]decan-8-yl] acetic acid (100 mg, 89.8 µmol, 37.2% yield, 72% purity) as a white solid. MS (ESI) m/z: 802.6 [M+H] + . Step E. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[[(3S)-8-[2-[[3-(2,6-d ioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-8-azaspiro[4.5]decan-3-yl]m ethoxy]-2-methyl- phenyl]pyridine-2-carboxylate To a solution of 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (38.65 mg, 149.63 µmol, 1.2 equiv.), 2-[(3S)-3-[[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dih ydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]methyl]-8- azaspiro[4.5]decan-8-yl]acetic acid (100.00 mg, 124.69 µmol, 1 equiv.) in DMF (1 mL) was added HATU (71.12 mg, 187.04 µmol, 1.5 equiv.) and DIEA (48.35 mg, 374.08 µmol, 65.16 µL, 3 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was diluted with water (10 mL) and filtered to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[[(3S)-8-[2-[[3-(2,6-dioxo -3-piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-8-azaspiro[4.5]decan-3-yl]methoxy]- 2-methyl-phenyl]pyridine-2- carboxylate (160 mg, 89.3 µmol, 71.6% yield, 58.2% purity) as a pink solid. MS (ESI) m/z: 521.9 [1/2M+H] + Step F. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[[(3S)-8-[2-[[3-(2,6-dioxo-3-piper idyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-8-azaspiro[4.5]decan-3-yl]methoxy]-2- methyl-phenyl]pyridine-2- carboxylic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)- yl)-3-(3-(((2S)-8-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2- oxoethyl)-8-azaspiro[4.5]decan-2-yl)methoxy)-2-methylphenyl) picolinic acid] To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[[(3S)-8-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo- ethyl]-8-azaspiro[4.5]decan-3-yl]methoxy]-2-methyl-phenyl]py ridine-2-carboxylate (160.00 mg, 153.51 µmol, 1 equiv.) in TFA (1 mL) and DCM (2 mL). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[[(3S)-8-[2-[[3-(2,6-dioxo -3-piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-8-azaspiro[4.5]decan-3-yl]methoxy]- 2-methyl-phenyl]pyridine-2- carboxylic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)- 3-(3-(((2S)-8-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-6-yl)amino)-2-oxoethyl)- 8-azaspiro[4.5]decan-2-yl)methoxy)-2-methylphenyl)picolinic acid] (20.9 mg, 20.5 µmol, 13.3% yield, 96.8% purity) was obtained as an off-white solid. MS (ESI) m/z: 493.9 [1/2M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.87 (s, 1H), 9.87 (s, 1H), 8.05 - 8.00 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.65 - 7.59 (m, 2H), 7.50 - 7.41 (m, 3H), 7.40 - 7.32 (m, 2H), 7.21 (dd, J = 1.6, 8.8 Hz, 1H), 7.11 - 7.05 (m, 1H), 6.96 (d, J = 8.8 Hz, 1H), 6.87 (d, J = 8.4 Hz, 1H), 6.62 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.32 (dd, J = 5.2, 9.6 Hz, 1H), 3.92 (s, 4H), 3.91 - 3.84 (m, 3H), 3.17 (s, 4H), 3.02 (t, J = 5.6 Hz, 2H), 2.71 - 2.60 (m, 3H), 2.55 - 2.53 (m, 2H), 2.37 - 2.30 (m, 1H), 2.22 - 2.12 (m, 1H), 1.90 (s, 3H), 1.87 - 1.75 (m, 2H), 1.60 - 1.45 (m, 7H), 1.23 (t, J = 11.2 Hz, 1H) Example 195. Preparation of Compound 249 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3, 4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[1-[2-[4- [3-(2, 6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]-1-piperidyl]ethy l]-4-piperidyl]ethoxy]-2- methyl-phenyl] pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 4-[3-(2, 6-dibenzyloxy-3-pyridyl)-1- methyl-indazol-7-yl]-3,6-dihydro-2H-pyridine-1-carboxylate A mixture of 7-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (3.0 g, 6.00 mmol, 1.0 equiv.), tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydro- 2H-pyridine-1- carboxylate (1.85 g, 6.00 mmol, 1.0 equiv.), and Ad2nBuP Pd G3 (436.63 mg, 599.54 µmol, 0.1 equiv.) and K3PO4 (3.82 g, 17.99 mmol, 3.0 equiv.) in dioxane (30 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 80 °C for 16 hours under N 2 atmosphere. The reaction was quenched by H 2 O (120 mL) and extracted by ethyl acetate (150 mL × 2). The organic phase was washed with brine (100 mL × 2), dried with Na2SO4, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~30% ethyl acetate/petroleum ether) to give tert-butyl 4-[3-(2, 6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-7-yl]-3,6-dihydro- 2H-pyridine-1-carboxylate (2.4 g, 3.8 mmol, 63.6% yield, 95.0% purity) as yellow oil. MS (ESI) m/z: 603.4 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 7.88 (d, J = 8.4 Hz, 1H), 7.57 (dd, J = 0.8, 8.4 Hz, 1H), 7.50 - 7.46 (m, 2H), 7.43 - 7.29 (m, 8H), 7.09 (dd, J = 0.8, 6.8 Hz, 1H), 7.00 (d, J = 8.4 Hz, 1H), 6.60 (d, J = 8.4 Hz, 1H), 5.79 (s, 1H), 5.44 (d, J = 4.8 Hz, 2H), 4.11 - 4.02 (m, 7H), 3.63 (t, J = 5.2 Hz, 2H), 1.45 (s, 9H). Step B. Procedure for preparation of tert-butyl 4-[3-(2, 6-dioxo-3-piperidyl)-1-methyl- indazol-7-yl] piperidine-1-carboxylate A mixture of tert-butyl 4-[3-(2, 6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-7-yl]-3,6-dihydro- 2H-pyridine-1-carboxylate (2.4 g, 3.98 mmol, 1.0 equiv.), Pd/C (2 g, 10% purity, 1.0 equiv.), Pd(OH)2 (2.0 g, 2.85 mmol, 20% purity) and AcOH (717.37 mg, 11.95 mmol, 683.21 µL, 3.0 equiv.) in THF (15 mL) and EtOH (15 mL) was degassed and purged with H 2 three times, and then the mixture was stirred at 25 °C for 16 hours under H2 (15Psi) atmosphere. The mixture solution was filtered and concentrated under reduced pressure to give tert-butyl 4-[3-(2, 6- dioxo-3-piperidyl)-1-methyl-indazol-7-yl] piperidine-1-carboxylate (1.6 g, crude) as a purple solid. MS (ESI) m/z: 427.3 [M+H] + Step C. Procedure for preparation of 3-[1-methyl-7-(4-piperidyl)indazol-3-yl]piperidine- 2,6-dione To a solution of tert-butyl 4-[3-(2, 6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl] piperidine-1- carboxylate (1.6 g, 3.75 mmol, 1.0 equiv.) inn HCl/dioxane (10 mL). The mixture was stirred at 25 °C for 1 hour. The mixture was filtered to give 3-[1-methyl-7-(4-piperidyl)indazol-3- yl]piperidine-2,6-dione (1.1 g, crude) as a white solid. Step D. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[2-methyl-3-[2-[1-(2-oxoethyl)-4- piperidyl]ethoxy]phenyl]pyridine-2-carboxylic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[2-[1-(2,2-diethoxyethyl)-4-piperidyl]ethoxy]-2-met hyl-phenyl]pyridine-2- carboxylate (60 mg, 73.17 µmol, 1.0 equiv.) in HCOOH (1 mL) was stirred at 85 C for 1.5 hours. The reaction mixture was concentrated under reduced pressure to remove HCOOH. The residue was diluted with H 2 O (10 mL) and extracted with ethyl acetate (10 mL × 3). Then the mixture was dried, filtered, and concentrated under reduced pressure to give 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[2-methyl-3-[2-[1-(2- oxoethyl)-4-piperidyl]ethoxy]phenyl]pyridine-2-carboxylic acid (45 mg, crude) as a yellow oil and it was used into the next step without further purification. MS (ESI) m/z: 708.4 [M+H2O+H] + Step E. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3, 4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[2-[1-[2-[4-[3-(2,6-dioxo-3-piperi dyl)-1-methyl-indazol-7-yl]-1- piperidyl]ethyl]-4-piperidyl]ethoxy]-2-methyl-phenyl] pyridine-2-carboxylic acid A solution of 3-[1-methyl-7-(4-piperidyl)indazol-3-yl]piperidine-2,6-dione (14.19 mg, 43.49 µmol, 1.0 equiv.) and NaBH(OAc)3 (27.65 mg, 130.47 µmol, 3.0 equiv.) in DCM (0.5 mL) and IPA (0.5 mL) was stirred at 0 °C. 6-[8-(1,3-Benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[2-methyl-3-[2-[1-(2-oxoethyl)-4-piperid yl]ethoxy]phenyl]pyridine-2- carboxylic acid (30 mg, 43.49 µmol, 1.0 equiv.) was added dropwise slowly at 0 °C. After addition, the mixture was stirred at 0 °C for 1 hour, warmed to 25 °C, and stirred for 2 hours. The reaction mixture was concentrated under reduced pressure to remove DCM (0.5 mL). The residue was diluted with H2O (10 mL) and extracted with ethyl acetate (10 mL × 3). The reaction mixture was then filtered, and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3, 4-dihydro-1H-isoquinolin-2-yl]-3-[3-[2-[1-[2-[4-[3-(2,6-diox o-3-piperidyl)- 1-methyl-indazol-7-yl]-1-piperidyl]ethyl]-4-piperidyl]ethoxy ]-2-methyl-phenyl] pyridine-2- carboxylic acid (7.3 mg, 7.2 µmol, 16.7% yield, 99.2% purity) as a yellow solid. MS (ESI) m/z: 500.5 [M/2+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 10.92 - 10.86 (m, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 7.6 Hz, 1H), 7.53 (d, J = 8.0 Hz, 1H), 7.50 - 7.28 (m, 3H), 7.39 - 7.32 (m, 2H), 7.24 (d, J = 7.2 Hz, 1H), 7.10 - 7.05 (m, 2H), 6.94 - 6.87 (m, 2H), 6.66 (d, J = 6.8 Hz, 1H), 4.98 (s, 2H), 4.39 - 4.33 (m, 1H), 4.20 (s, 2H), 4.02 - 3.97 (m, 3H), 3.91 ( t, J = 5.2 Hz, 2H), 3.08 - 3.01 (m, 6H), 2.98 - 2.85 (m, 2H), 2.75 - 2.61 (m, 5H), 2.23 - 2.14 (m, 3H), 2.09 - 1.95 (m, 3H), 1.91 (s, 3H), 1.87 - 1.81 (m, 2H), 1.79 - 1.76 (m, 1H), 1.75 - 1.60 (m, 5H), 1.55 - 1.41 (m, 1H), 1.29 - 1.21 (m, 2H) Example 196. Preparation of Compound 148b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4s)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of 3-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]propanal A solution of DMSO (95.50 mg, 1.22 mmol, 95.50 µL, 4 equiv.) in DCM (5 mL) was added dropwise to a solution of oxalyl dichloride (77.57 mg, 611.14 µmol, 53.50 µL, 2 equiv.) in DCM (1 mL) at -70 °C under N 2 atmosphere. The mixture was stirred at -70°C for 30 minutes. After which time 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]propan-1-ol (100 mg, 305.57 µmol, 1 equiv.) in DCM (1 mL) was added dropwise at -70 °C. The solution was stirred for 30 minutes at -70 °C. Then TEA (185.52 mg, 1.83 mmol, 255.19 µL, 6 equiv.) was added into the solution. The solution was stirred at 25 °C for 2 hour under N2 atmosphere. The reaction mixture was quenched by addition H2O (15 mL) at 25°C and then extracted with DCM (20 mL × 3). The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~10% ethyl acetate/petroleum ether) to give 3-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]propanal (100 mg, crude) as a yellow oil. Step B. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[2-methyl-3-[4-(3- oxopropyl)cyclohexoxy]phenyl]pyridine-2-carboxylate 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]propanal (100 mg, 307.47 µmol, 1 equiv.), tert- butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-2-carboxylate (188.34 mg, 307.47 µmol, 1 equiv.), KF (53.59 mg, 922.40 μmol, 21.61 μL, 3 equiv.), and Ad2nBuP Pd G3(cataCXium® A Pd G3 ) (22.39 mg, 30.75 μmol, 0.1 equiv.) were taken up into a microwave tube in dioxane (1 mL) and H 2 O (0.1 mL). The sealed tube was heated at 100°C for 60 min under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~8% dichloromethane / methanol ether) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[2-methyl-3-[4-(3-oxopropyl)cyclohexoxy]phenyl]pyridin e-2-carboxylate (110 mg, 138.3 µmol, 44.9% yield, 91.9% purity) as a yellow oil. MS (ESI) m/z: 731.6[M+H] + . Step C. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo -3-piperidyl)-1-methyl- indazol-6-yl]piperazin-1-yl]propyl]cyclohexoxy]-2-methyl-phe nyl]pyridine-2- carboxylate To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[2-methyl-3-[4-(3-oxopropyl)cyclohexoxy]phenyl]pyrid ine-2-carboxylate (110 mg, 150.50 µmol, 1 equiv.) and 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (60.23 mg, 165.55 µmol, 1.1 equiv. HCl) in DCM (1.1 mL) was added AcOH (903.76 ug, 15.05 µmol, 0.861 µL, 0.1 equiv.). The mixture was stirred at 25 °C for 15.5 hours. NaBH(OAc) 3 (95.69 mg, 451.49 µmol, 3 equiv.) was then added. The mixture was stirred at 25 °C for 0.5 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~10% dichloromethane / methanol) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo-3-piperidyl)-1 -methyl-indazol-6-yl]piperazin-1- yl]propyl]cyclohexoxy]-2-methyl-phenyl]pyridine-2-carboxylat e (110 mg, 77.5 µmol, 51.5% yield, 73.5% purity) as a yellow solid. MS (ESI) m/z:521.9[1/2 M+H] + . Step D. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4s)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)propyl)cyclohexyl)oxy) -2-methylphenyl)picolinic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-inda zol-6-yl]piperazin-1- yl]propyl]cyclohexoxy]-2-methyl-phenyl]pyridine-2-carboxylat e (110 mg, 105.54 µmol, 1 equiv.) in TFA (0.6 mL) was added DCM (0.6 mL). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4s)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1-yl)propyl)cyclohexyl)oxy)-2-meth ylphenyl)picolinic acid (23.2 mg, 23.0 µmol, 21.8% yield, 97.8% purity) as a grey solid. MS (ESI) m/z: 986.4 [M+H] + . 1H NMR (400 MHz, DMSO-d 6 ) δ = 10.85 (s, 1H), 8.15 (s, 1H), 8.07 - 7.98 (m, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.65 - 7.59 (m, 1H), 7.51 - 7.42 (m, 4H), 7.40 - 7.31 (m, 2H), 7.07 (t, J = 8.0 Hz, 1H), 6.98 - 6.90 (m, 2H), 6.90 - 6.87 (m, 1H), 6.83 (s, 1H), 6.60 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.61 (s, 1H), 4.25 (dd, J = 5.2, 9.2 Hz, 1H), 3.93 - 3.86 (m, 5H), 3.21 (s, 4H), 3.02 (t, J = 5.2 Hz, 2H), 2.62 - 2.58 (m, 2H), 2.54 (s, 4H), 2.35 - 2.31 (m, 2H), 2.20 - 2.12 (m, 1H), 1.93 (s, 5H), 1.61 - 1.46 (m, 6H), 1.40 - 1.17 (m, 6H) Example 197. Preparation of Compound 254a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(((1s,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of (1r,4r)-methyl 4-(4-bromo-3- methylphenoxy)cyclohexanecarboxylate To a solution of (1s,4s)-methyl 4-hydroxycyclohexanecarboxylate (2.0 g, 12.64 mmol, 1.1 equiv.) in toluene (20 mL) was added 2-(tributyl-λ5-phosphanylidene)acetonitrile (3.33 g, 13.79 mmol, 1.2 equiv.) and 4-bromo-3-methylphenol (2.15 g, 11.49 mmol, 1.0 equiv.). The mixture was stirred at 120 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=1/0) to give (1r,4r)-methyl 4-(4-bromo-3- methylphenoxy)cyclohexanecarboxylate (2.4 g, crude) as a yellow oil. Step B. Procedure for preparation of ((1r,4r)-4-(4-bromo-3- methylphenoxy)cyclohexyl)methanol To a solution of (1r,4r)-methyl 4-(4-bromo-3-methylphenoxy)cyclohexanecarboxylate (2.4 g, 7.33 mmol, 1 equiv.) in THF (25 mL) was added LiAlH4 (278.38 mg, 7.33 mmol, 1 equiv.). The mixture was stirred at 0 °C for 1 hour under N2 atmosphere. The reaction mixture was quenched by addition H 2 O (0.3 mL), 15% NaOH (0.3 mL), and H 2 O (0.9 mL). The combined mixture was mixed with Na2SO4, and then filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~20% ethyl acetate/petroleum ether) to give compound ((1r,4r)-4-(4-bromo-3- methylphenoxy)cyclohexyl)methanol (1.3 g, 4.3 mmol, 59.2% yield) as a yellow oil. 1 H NMR (400 MHz) δ = 7.40 (d, J = 8.8 Hz, 1H), 6.94 (d, J = 2.8 Hz, 1H), 6.75 - 6.67 (m, 1H), 4.43 (t, J = 5.6 Hz, 1H), 4.26 - 4.15 (m, 1H), 3.23 (t, J = 5.6 Hz, 2H), 2.28 (s, 3H), 2.08 - 2.00 (m, 2H), 1.77 (d, J = 11.6 Hz, 2H), 1.43 - 1.33 (m, 1H), 1.33 - 1.22 (m, 2H), 1.11 - 0.98 (m, 2H) Step C. Procedure for preparation of (1r,4r)-4-(4-bromo-3- methylphenoxy)cyclohexanecarbaldehyde A solution of oxalyl dichloride (1.10 g, 8.69 mmol, 760.68 µL, 2 equiv.) in DCM (20 mL) was added dropwise to a solution of DMSO (1.36 g, 17.38 mmol, 1.36 mL, 4 equiv.) in DCM (2 mL) at -70 °C under N 2 atmosphere. The mixture was stirred at -70 °C for 1 hour. After which time ((1r,4r)-4-(4-bromo-3-methylphenoxy)cyclohexyl)methanol (1.3 g, 4.34 mmol, 1 equiv.) in DCM (2 mL) was added dropwise at -70 °C. The solution was stirred for 1 hour at -70 °C. Then TEA (2.64 g, 26.07 mmol, 3.63 mL, 6 equiv.) was added into the solution. The solution was stirred at -70 °C for 0.5 hour under N 2 atmosphere. DCM (50 mL) and water (50 mL) were added, and the layers were separated. The aqueous phase was washed with DCM (30 mL × 2), dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum to give (1r,4r)- 4-(4-bromo-3-methylphenoxy)cyclohexanecarbaldehyde (1.2 g, 4.0 mmol, 92.9% yield) as a yellow oil. Step D. Procedure for preparation of (E)-ethyl 3-((1r,4r)-4-(4-bromo-3- methylphenoxy)cyclohexyl)acrylate To a solution of ethyl 2-(diethoxyphosphoryl)acetate (1.81 g, 8.08 mmol, 1.60 mL, 2.0 equiv.) in THF (15 mL) was added NaH (339.15 mg, 8.48 mmol, 60% purity, 2.1 equiv.). The mixture was stirred at 0 °C for 1 hour. Then (1r,4r)-4-(4-bromo-3- methylphenoxy)cyclohexanecarbaldehyde (1.2 g, 4.04 mmol, 1 equiv.) was added into the mixture and stirred at 25 °C for 0.5 hour under N2. The reaction mixture was quenched by addition water (10 mL) at 0 °C and extracted with ethyl acetate (30 mL × 3). The combined organic layers were washed with water (10mL × 3), filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=0/1 to 10/1) to give (E)-ethyl 3-((1r,4r)-4-(4-bromo-3- methylphenoxy)cyclohexyl)acrylate (1 g, 2.7 mmol, 67.4% yield) as a white solid. 1 H NMR (400 MHz) δ = 7.41 (d, J = 8.8 Hz, 1H), 6.96 (d, J = 2.8 Hz, 1H), 6.76 - 6.70 (m, 1H), 6.16 - 6.07 (m, 1H), 5.74 (d, J = 11.6 Hz, 1H), 4.32 - 4.22 (m, 1H), 4.14 - 4.06 (m, 2H), 3.25 - 3.15 (m, 1H), 2.29 (s, 3H), 2.12 - 2.02 (m, 2H), 1.77 - 1.65 (m, 2H), 1.39 - 1.28 (m, 4H), 1.21 (t, J = 7.2 Hz, 3H) Step E. Procedure for preparation of ethyl 3-((1r,4r)-4-(4-bromo-3- methylphenoxy)cyclohexyl)propanoate To a solution of (E)-ethyl 3-((1r,4r)-4-(4-bromo-3-methylphenoxy)cyclohexyl)acrylate (1 g, 2.72 mmol, 1 equiv.) in EtOH (10 mL) was added PtO2 (61.83 mg, 272.27 µmol, 0.1 equiv.) under H2 atmosphere (15 psi). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a filter cake, rinsing with EtOH. The filter cake was dried under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=20/1 to 10/1) to give ethyl 3-((1r,4r)-4-(4-bromo-3-methylphenoxy)cyclohexyl)propanoate (700 mg, 1.7 mmol, 65.2% yield, 93.7% purity) as a colorless oil. 1 H NMR (400 MHz) δ = 7.40 (d, J = 8.4 Hz, 1H), 6.94 (d, J = 2.8 Hz, 1H), 6.74 - 6.67 (m, 1H), 4.28 - 4.16 (m, 1H), 4.09 - 4.00 (m, 2H), 2.34 - 2.22 (m, 5H), 2.02 (d, J = 10.0 Hz, 2H), 1.74 (d, J = 12.0 Hz, 2H), 1.51 - 1.40 (m, 2H), 1.34 - 1.22 (m, 3H), 1.21 - 1.15 (m, 3H), 1.11 - 0.98 (m, 2H) Step F. Procedure for preparation of 3-((1r,4s)-4-(4-bromo-3- methylphenoxy)cyclohexyl)propan-1-ol To a solution of ethyl 3-((1r,4r)-4-(4-bromo-3-methylphenoxy)cyclohexyl)propanoate (500 mg, 1.35 mmol, 1 equiv.) in THF (10 mL) was added LiAlH4 (51.38 mg, 1.35 mmol, 1 equiv.)The mixture was stirred at 0 °C for 1 hour under N 2 atmosphere. The reaction mixture was quenched by addition of H 2 O (0.1 mL), 15% NaOH (0.1 mL), and H 2 O (0.3 mL). The combined mixture was mixed with Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~20% ethyl acetate/petroleum ether) to give 3-((1r,4s)-4-(4-bromo-3- methylphenoxy)cyclohexyl)propan-1-ol (400 mg, 1.2 mmol, 90.2% yield) as a yellow oil. 1 H NMR (400 MHz,) δ = 7.40 (d, J = 8.8 Hz, 1H), 6.94 (d, J = 2.8 Hz, 1H), 6.74 - 6.69 (m, 1H), 4.34 (t, J = 5.2 Hz, 1H), 4.26 - 4.17 (m, 1H), 3.39 - 3.33 (m, 2H), 2.28 (s, 3H), 2.02 (d, J = 10.0 Hz, 2H), 1.79 - 1.72 (m, 2H), 1.47 - 1.38 (m, 2H), 1.34 - 1.25 (m, 2H), 1.24 - 1.16 (m, 3H), 1.10 - 0.96 (m, 2H) Step G. Procedure for preparation of 3-((1r,4r)-4-(4-bromo-3- methylphenoxy)cyclohexyl)propanal A solution of oxalyl dichloride (310.29 mg, 2.44 mmol, 213.99 µL, 2.0 equiv.) in DCM (20 mL) was added dropwise to a solution of DMSO (381.99 mg, 4.89 mmol, 381.99 µL, 4.0 equiv.) in DCM (2 mL) at -70 °C under N 2 atmosphere. The mixture was stirred at -70 °C for 1 hour. After which time 3-((1r,4s)-4-(4-bromo-3-methylphenoxy)cyclohexyl)propan-1-ol (400 mg, 1.22 mmol, 1 equiv.) in DCM (2 mL) was added dropwise at -70 °C. The solution was stirred for 1 hour at -70 °C. Then TEA (742.10 mg, 7.33 mmol, 1.02 mL, 6 equiv.) was added into the solution. The solution was stirred at -70 °C for 0.5 hour under N2 atmosphere. DCM (50 mL) and water (50 mL) were added, and layers were separated. The aqueous layer was extracted with DCM (30 mL × 2). The combined organic extracts were dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum to give 3-((1r,4r)-4-(4- bromo-3-methylphenoxy)cyclohexyl)propanal (270 mg, 830.1 µmol, 67.9% yield) as a yellow oil. Step H. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(2-methyl-4-(((1r,4r)-4-(3- oxopropyl)cyclohexyl)oxy)phenyl)picolinate To a solution of 3-((1r,4r)-4-(4-bromo-3-methylphenoxy)cyclohexyl)propanal (270 mg, 830.16 µmol, 1.0 equiv.) in dioxane (5 mL) was added KF (1.5 M, 1.66 mL, 3.0 equiv.), tert- butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)picolinate (508.51 mg, 830.16 µmol, 1.0 equiv.), and [2- (2-aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-butyl-p hosphane;methanesulfonate (60.46 mg, 83.02 µmol, 0.1 equiv.). The mixture was stirred at 100 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by flash silica gel chromatography (eluent of 0~50% ethyl acetate/petroleum ether) to give tert- butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2-methyl-4- (((1r,4r)-4-(3-oxopropyl)cyclohexyl)oxy)phenyl)picolinate (230 mg, 314.6 µmol, 37.9% yield) as a yellow solid. MS (ESI) m/z: 731.5[M+H] + Step I. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)propyl)cyclohexyl)oxy) -2- methylphenyl)picolinate To a solution of 3-(1-methyl-6-(piperazin-1-yl)-1H-indazol-3-yl)piperidine-2, 6-dione (59.73 mg, 164.18 µmol, 1.2 equiv.) in DCM (0.5 mL) was added NaBH(OAc) 3 (86.99 mg, 410.45 µmol, 3.0 equiv.). Then tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(2-methyl-4-(((1r,4r)-4-(3- oxopropyl)cyclohexyl)oxy)phenyl)picolinate (100 mg, 136.82 µmol, 1.0 equiv.) was added into the mixture at 0 °C. The mixture was stirred at 25°C for 1 hour. The reaction mixture was concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(4-(((1s,4r)-4-(3-(4-(3-( 2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)propyl)cyclohexyl)oxy) -2-methylphenyl)picolinate (120 mg, 115.1 µmol, 84.1% yield) as a yellow solid. MS (ESI) m/z: 1042.4 [M+H] + Step J. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)propyl)cyclohexyl)oxy) -2-methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-6- yl)piperazin-1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl)pico linate (120 mg, 115.13 µmol, 1.0 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 117.31 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(4-(((1s,4r)-4-(3-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin -1-yl)propyl)cyclohexyl)oxy)- 2-methylphenyl)picolinic acid (43.4 mg, 43.7 µmol, 37.9% yield, 99.2% purity) as a white solid. MS (ESI) m/z: 986.4 [M+H] + 1 H NMR (400 MHz) δ = 12.92 - 12.82 (m, 1H), 12.67 - 12.46 (m, 1H), 10.87 (s, 1H), 9.55 - 9.43 (m, 1H), 8.04 (d, J = 8.0 Hz, 1H), 7.80 (d, J = 8.0 Hz, 1H), 7.65 - 7.55 (m, 2H), 7.49 - 7.44 (m, 2H), 7.41 - 7.33 (m, 2H), 7.01 - 6.94 (m, 3H), 6.91 (d, J = 8.4 Hz, 1H), 6.78 (d, J = 2.3 Hz, 1H), 6.74 - 6.69 (m, 1H), 4.98 (s, 2H), 4.32 - 4.21 (m, 2H), 3.93 (s, 3H), 3.92 - 3.89 (m, 2H), 3.62 ( d, J = 10.4 Hz, 2H), 3.21 - 3.13 (m, 4H), 3.09 - 3.01 (m, 4H), 2.69 - 2.60 (m, 2H), 2.55 - 2.52 (m, 2H), 2.34 - 2.31 (m, 1H), 2.20 - 2.14 (m, 1H), 2.09 ( d, J = 11.6 Hz, 2H), 2.03 (s, 3H), 1.86 - 1.79 (m, 2H), 1.72 ( d, J = 4.0 Hz, 2H), 1.38 - 1.22 (m, 5H), 1.16 - 1.04 (m, 2H) Example 198. Preparation of Compound 269a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip eridin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of 3-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]propan-1-ol To a solution of ethyl 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]propanoate (600 mg, 1.62 mmol, 1 equiv.) in THF (10 mL) was added LiAlH4 (55.50 mg, 1.46 mmol, 0.9 equiv.) at 0 °C. The mixture was stirred at 0 °C for 1.5 hours. The reaction mixture was quenched by addition H 2 O (0.05 mL), 15% NaOH (0.05 mL), H 2 O (15mL), and then extracted with ethyl acetate 30 mL (10 mL × 3). The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was used in the next step without purification. The compound 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]propan-1- ol (500 mg, 1.5 mmol, 94.0% yield) was obtained as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ = 7.14 - 7.02 (m, 3H), 4.23 - 4.21 (m, 1H), 3.37 - 3.36 (m, 3H), 3.34 -3.35 (m, 1H), 2.26 - 2.24 (m, 1H), 2.21 -2.19 (m, 2H), 2.06 - 2.04 (m, 2H), 1.76 - 1.72 (m, 2H), 1.41 - 1.42 (m, 2H), 1.40 - 1.39 (m, 2H), 1.38 - 1.36 (m, 2H), 1.20 - 1.15 (m, 3H), 1.03 - 0.99(m, 2H). Step B. Procedure for preparation of 3-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]propanal A solution of (COCl)2 (167.23 mg, 1.41 mmol, 101.97 µL, 2 equiv.) in DCM (30 mL) was added dropwise to a solution of DMSO (219.65 mg, 2.81 mmol, 219.65 µL, 4 equiv.) in DCM (30 mL) at -70 °C under N 2 atmosphere. The mixture was stirred at -70 °C for 1 hour. After which time 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]propan-1-ol (230 mg, 702.81 µmol, 1 equiv.) in DCM (30 mL) was added dropwise at -70 °C. The solution was stirred for 1 hour at -70 °C. Then TEA (426.70 mg, 4.22 mmol, 586.94 µL, 6 equiv.) was added into the solution. The solution was stirred at -70 °C for 1 hour under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~43% ethyl acetate/petroleum ether gradient) to give 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]propanal (200 mg, 467.3 µmol, 66.5% yield, 76% purity) as a white solid. Step C. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[2-methyl-3-[4-(3- oxopropyl)cyclohexoxy]phenyl]pyridine-2-carboxylate A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine- 2-carboxylate (414.34 mg, 676.42 µmol, 1.1 equiv.), 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]propanal (200 mg, 614.93 µmol, 1 equiv.), KF (1.5 M, 614.93 µL, 1.5 equiv.), and Ad 2 nBuP Pd G 3 (cataCXium® A Pd G3 ) (134.35 mg, 184.48 µmol, 0.3 equiv.) in 1,4-dioxane (15 mL) was degassed and purged with N2 three times. The mixture was then stirred at 100 °C for 1 hour under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~37% ethyl acetate/petroleum ether). The compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[2-methyl-3-[4-(3- oxopropyl)cyclohexoxy]phenyl]pyridine-2-carboxylate (300 mg, 410.4 µmol, 66.7% yield) was obtained as a yellow oil. Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo -3-piperidyl)-1-methyl- indazol-6-yl]-1-piperidyl]propyl]cyclohexoxy]-2-methyl-pheny l]pyridine-2-carboxylate A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[2-methyl-3-[4-(3-oxopropyl)cyclohexoxy]phenyl]pyridin e-2-carboxylate (50 mg, 68.41 µmol, 1 equiv.), and 3-[1-methyl-6-(4-piperidyl)indazol-3-yl]piperidine-2,6-dione (33.49 mg, 102.61 µmol, 1.5 equiv.) in DCM (1 mL) was degassed and purged with N2 three times. The mixture was then stirred at 25 °C for 10 hours under N 2 atmosphere. NaBH(OAc) 3 (29.00 mg, 136.82 µmol, 2 equiv.) was then added to the mixture, which was stirred at 25 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-inda zol-6-yl]-1- piperidyl]propyl]cyclohexoxy]-2-methyl-phenyl]pyridine-2-car boxylate (60 mg, 57.6 µmol, 84.2% yield) was obtained as a white solid, which was used in the next step without further purification. MS (ESI) m/z: 986.4 [M+H-56] + Step E. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperidin-1-yl)propyl)cyclohexyl)oxy) -2-methylphenyl)picolinic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[4-[3-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-inda zol-6-yl]-1- piperidyl]propyl]cyclohexoxy]-2-methyl-phenyl]pyridine-2-car boxylate (50 mg, 48.02 µmol, 1 equiv.) in DCM (0.2 mL) was added TFA (769.98 mg, 6.75 mmol, 499.99 µL, 140.64 equiv.). The mixture was stirred at 25 °C for 0.5 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-6-yl)piperidin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (16.6 mg, 16.2 µmol, 33.7% yield, 95.9% purity) as a brown solid. MS (ESI) m/z: 986.4 [M+H-56] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.91 - 10.74 (m, 1H), 8.30 (s, 2H), 7.99 (d, J = 6.1 Hz, 1H), 7.76 (d, J = 7.2 Hz, 1H), 7.63 (d, J = 7.2 Hz, 1H), 7.49 (d, J = 8.8 Hz, 1H), 7.45 - 7.38 (m, 2H), 7.34 (d, J = 5.6 Hz, 2H), 7.07 - 6.98 (m, 1H), 6.91 (d, J = 9.2 Hz, 1H), 6.85 - 6.81 (m, 2H), 6.69 - 6.67 (m, 1H), 5.04 - 4.91 (m, 2H), 4.26 - 4.24 (m, 1H), 3.91 - 3.89 (m, 5H), 3.22- 3.20 (m, 4H), 3.02 - 3.00 (m, 4H), 2.64 - 2.58 (m, 2H), 2.35 - 2.29 (m, 4H), 2.21 - 2.12 (m, 2H), 2.09 - 2.07 (m, 3H), 1.89 (s, 3H), 1.82 - 1.80 (m, 2H), 1.49 - 1.47 (m, 2H), 1.39 - 1.33 (m, 2H), 1.27 - 1.16 (m, 4H), 1.10 - 1.03 (m, 2H). Example 199. Preparation of Compound 263b 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[(2S)-3-[1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-o xo-ethyl]-4-piperidyl]-2- methyl-propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(3-((2S)-3-(1- (2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)am ino)-2-oxoethyl)piperidin-4- yl)-2-methylpropoxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of ethyl 2-[4-[(2S)-3-(3-bromo-2-methyl-phenoxy)-2- methyl-propyl]-1-piperidyl]acetate The stereoisomers of ethyl 2-[4-[3-(3-bromo-2-methyl-phenoxy)-2-methyl-propyl]-1- piperidyl]acetate (1 g, 2.43 mmol, 1.00 equiv.) were separated by SFC to give ethyl 2-[4-[(2S)- 3-(3-bromo-2-methyl-phenoxy)-2-methyl-propyl]-1-piperidyl]ac etate (0.3 g, 727.5 µmol, 30.0% yield) as a colorless oil. Step B. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[(2S)-3-[1-(2-ethoxy-2 -oxo-ethyl)-4-piperidyl]-2- methyl-propoxy]-2-methyl-phenyl]pyridine-2-carboxylate A mixture of ethyl 2-[4-[(2S)-3-(3-bromo-2-methyl-phenoxy)-2-methyl-propyl]-1- piperidyl]acetate (150.00 mg, 363.76 µmol, 1.00 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tet ramethyl-1,3,2-dioxaborolan-2- yl)pyridine-2-carboxylate (222.82 mg, 363.76 µmol, 1.00 equiv.), Ad2nBuP Pd G3 (26.49 mg, 36.38 µmol, 0.10 equiv.), and K 3 PO 4 (231.64 mg, 1.09 mmol, 3.00 equiv.) in dioxane (2 mL) and H2O (0.2 mL) was degassed and purged with N2 three times. The mixture was then stirred at 80 °C for 16 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , petroleum ether/ethyl acetate = 1:1) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[(2S)-3-[1-(2-ethoxy-2-oxo -ethyl)-4-piperidyl]-2-methyl- propoxy]-2-methyl-phenyl]pyridine-2-carboxylate (140 mg, 171.1 µmol, 47.0% yield) was obtained as a yellow solid. MS (ESI) m/z: 409.9 [M/2+H] + . Step C. Procedure for preparation of 2-[4-[(2S)-3-[3-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-2- methyl-phenoxy]-2-methyl-propyl]-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[(2S)-3-[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]-2- methyl-propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (140 mg, 171.14 µmol, 1.00 equiv.) in THF (2 mL) and H 2 O (0.5 mL) was added LiOH•H2O (21.55 mg, 513.43 µmol, 3.00 equiv.).The mixture was stirred at 25 °C for 3 minutes. The reaction mixture was concentrated under reduced pressure to give crude 2-[4-[(2S)-3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H-isoquinolin-2- yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]-2-met hyl-propyl]-1-piperidyl]acetic acid (100 mg, 126.5 µmol, 73.9% yield) as a yellow solid. MS (ESI) m/z 790.4 [M+H] + . Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[(2S)-3-[1-[2-[[3-(2,6 -dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-4-piperidyl]-2-methyl-propo xy]-2-methyl- phenyl]pyridine-2-carboxylate To a solution of 2-[4-[(2S)-3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]-2-methyl-propyl]-1- piperidyl]acetic acid (80 mg, 101.27 µmol, 1.00 equiv.) 3-(6-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (28.77 mg, 111.40 µmol, 1.10 equiv.) in DMF (1 mL) was added HATU (46.21 mg, 121.52 µmol, 1.20 equiv.) and TEA (30.74 mg, 303.81 µmol, 42.29 µL, 3.00 equiv.). The mixture was stirred at 40 °C for 16 hours. The solution was then poured into water (2 mL) and filtered to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[(2S)-3-[1-[2-[[3-(2,6-dio xo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-4-piperidyl]-2-methyl-propo xy]-2-methyl-phenyl]pyridine- 2-carboxylate (80 mg, 77.6 µmol, 76.6% yield) as a pink solid. MS (ESI) m/z: 516.0 [M/2+H] + . Step E. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[(2S)-3-[1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]-2-methyl-propoxy]-2-meth yl-phenyl]pyridine-2- carboxylic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)- yl)-3-(3-((2S)-3-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-6-yl)amino)-2- oxoethyl)piperidin-4-yl)-2-methylpropoxy)-2-methylphenyl)pic olinic acid] To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[(2S)-3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methy l-indazol-6-yl]amino]-2-oxo- ethyl]-4-piperidyl]-2-methyl-propoxy]-2-methyl-phenyl]pyridi ne-2-carboxylate (80 mg, 77.65 µmol, 1.00 equiv.) in DCM (1 mL) was added TFA (616.00 mg, 5.40 mmol, 400.00 µL, 69.57 equiv.). The mixture was stirred at 40 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8- (1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin- 2-yl]-3-[3-[(2S)-3-[1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-4-piperidyl]-2-methyl- propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid [rel-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-((2S)-3-( 1-(2-((3-(2,6-dioxopiperidin-3- yl)-1-methyl-1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-4-y l)-2-methylpropoxy)-2- methylphenyl)picolinic acid] (18.8 mg, 17.8 µmol, 22.9% yield, 96.4% purity) as a yellow solid MS (ESI) m/z: 488.0 [M/2+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.88 (s, 1H), 9.90 - 9.75 (m, 1H), 8.19 (s, 1H), 8.08 - 7.97 (m, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 8.4 Hz, 2H), 7.48 - 7.40 (m, 3H), 7.39 - 7.31 (m, 2H), 7.21 (dd, J = 1.6, 8.8 Hz, 1H), 7.11 - 7.04 (m, 1H), 6.92 (d, J = 8.4 Hz, 1H), 6.86 (d, J = 8.4 Hz, 1H), 6.64 (d, J = 7.5 Hz, 1H), 5.01 - 4.94 (m, 2H), 4.32 (dd, J = 5.2, 9.6 Hz, 1H), 3.93 - 3.88 (m, 5H), 3.83 - 3.74 (m, 2H), 3.12 (s, 2H), 3.02 (t, J = 5.2 Hz, 2H), 2.90 - 2.83 (m, 2H), 2.66 - 2.60 (m, 2H), 2.19 - 2.14 (m, 2H), 2.03 (s, 2H), 1.94 - 1.89 (m, 3H), 1.71 - 1.63 (m, 2H), 1.49 - 1.39 (m, 2H), 1.32 - 1.13 (m, 4H), 1.03 - 0.98 (m, 3H) Example 200. Preparation of Compound 264 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4-[3-[1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-4-methyl-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 4-[3-(4-bromo-3-methyl- phenoxy)propyl]-4-methyl-piperidine-1-carboxylate To a solution of tert-butyl 4-(3-hydroxypropyl)-4-methyl-piperidine-1-carboxylate (250 mg, 971.37 µmol, 1.0 equiv.) in toluene (3 mL) and 4-bromo-3-methyl-phenol (218.01 mg, 1.17 mmol, 1.2 equiv.) was added 2-(tributyl-λ5-phosphanylidene)acetonitrile (351.67 mg, 1.46 mmol, 1.5 equiv.). The mixture was stirred at 120 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=1/0 to 10/1) to give tert-butyl 4-[3-(4- bromo-3-methyl-phenoxy)propyl]-4-methyl-piperidine-1-carboxy late (300 mg, 703.5 µmol, 72.4% yield) as a yellow oil. Step B. Procedure for preparation of 4-[3-(4-bromo-3-methyl-phenoxy)propyl]-4-methyl- piperidine A solution of tert-butyl 4-[3-(4-bromo-3-methyl-phenoxy)propyl]-4-methyl-piperidine-1 - carboxylate (300 mg, 703.59 µmol, 1.0 equiv.) in HCl/dioxane (3 mL) was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give 4-[3-(4-bromo- 3-methyl-phenoxy)propyl]-4-methyl-piperidine (180 mg, 551.6 µmol, 78.4% yield) as a white solid. Step C. Procedure for preparation of ethyl 2-[4-[3-(4-bromo-3-methyl-phenoxy)propyl]- 4-methyl-1-piperidyl]acetate To a solution of 4-[3-(4-bromo-3-methyl-phenoxy)propyl]-4-methyl-piperidine (180 mg, 551.69 µmol, 1.0 equiv.) and ethyl 2-bromoacetate (92.13 mg, 551.69 µmol, 61.01 µL, 1.0 equiv.) in CH3CN (2 mL) was added K2CO3 (228.74 mg, 1.66 mmol, 3.0 equiv.). The mixture was stirred at 60 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give ethyl 2-[4-[3-(4-bromo-3-methyl-phenoxy)propyl]-4-methyl-1- piperidyl]acetate (190 mg, 460.7 µmol, 83.5% yield) as a yellow oil. MS (ESI) m/z: 412.0 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 7.42 (d, J = 8.4 Hz, 1H), 6.94 (d, J = 2.8 Hz, 1H), 6.74 - 6.68 (m, 1H), 4.11 - 4.04 (m, 2H), 3.92 (t, J = 6.4 Hz, 2H), 3.18 (s, 2H), 2.46 - 2.37 (m, 3H), 2.29 (s, 3H), 1.69 - 1.58 (m, 2H), 1.41 - 1.25 (m, 7H), 1.20 - 1.16 (m, 3H), 0.86 (s, 3H). Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[3-[1-(2-ethoxy-2-oxo- ethyl)-4-methyl-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate Ethyl 2-[4-[3-(4-bromo-3-methyl-phenoxy)propyl]-4-methyl-1-piperid yl]acetate (190 mg, 460.76 µmol, 1.0 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (338.69 mg, 552.92 µmol, 1.2 equiv.), [2-(2-aminophenyl)phenyl]palladium(1+);bis(1- adamantyl)-butyl-phosphane;methanesulfonate (33.56 mg, 46.08 µmol, 0.1 equiv.), and KF (1.5 M, 921.53 µL, 3.0 equiv.) were taken up into a microwave tube in dioxane (3 mL). The sealed tube was heated at 100 °C for 1 hour under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate=10/1 to 1/1) to give tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[4-[3-[1-(2-ethoxy-2-oxo- ethyl)-4-methyl-4-piperidyl]propoxy]-2-methyl-phenyl]pyridin e-2-carboxylate (230 mg, 281.1 µmol, 61.0% yield) as a yellow solid. MS (ESI) m/z: 818.4 [M+H] + Step E. Procedure for preparation of 2-[4-[3-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyr idyl]-3-methyl- phenoxy]propyl]-4-methyl-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[4-[3-[1-(2-ethoxy-2-oxo-ethyl)-4-methyl-4-piperidyl ]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (230 mg, 281.16 µmol, 1.0 equiv.) in THF (1 mL) and H2O (1 mL) was added LiOH.H 2 O (67.34 mg, 2.81 mmol, 10.0 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was adjusted pH to 3~ 4 with HCl (1 M), and extracted with DCM (2 mL × 3). The combined organic layers were concentrated under reduced pressure to give 2-[4-[3-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H-isoquinolin-2-yl]- 2-tert-butoxycarbonyl-3-pyridyl]-3-methyl-phenoxy]propyl]-4- methyl-1-piperidyl]acetic acid (200 mg, 253.1 µmol, 90.0% yield) as a white solid. Step F. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[3-[1-[2-[[3-(2,6-diox o-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-4-methyl-4-piperidyl]propox y]-2-methyl- phenyl]pyridine-2-carboxylate To a solution of 2-[4-[3-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-3-methyl- phenoxy]propyl]-4-methyl-1- piperidyl]acetic acid (100 mg, 126.59 µmol, 1.0 equiv.) and 3-(6-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (39.23 mg, 151.90 µmol, 1.2 equiv.) in pyridine (1 mL) was added EDCI (36.40 mg, 189.88 µmol, 1.5 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was quenched by adding water (5 mL), and then filtered and concentrated under reduced pressure to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[4-[3-[1-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-methyl-4-piperidyl]propoxy]-2-methy l-phenyl]pyridine-2- carboxylate (100 mg, 97.0 µmol, 76.6% yield) as a yellow solid. MS (ESI) m/z: 516.0 [M/2+H] + Step G. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[4-[3-[1-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-methyl-4-piperidyl]propoxy]-2-methy l-phenyl]pyridine-2- carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[4-[3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-ind azol-6-yl]amino]-2-oxo-ethyl]-4- methyl-4-piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carbo xylate (100 mg, 97.06 µmol, 1.0 equiv.) in DCM (1 mL) and TFA (1 mL). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[4-[3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)- 1-methyl-indazol-6-yl]amino]-2- oxo-ethyl]-4-methyl-4-piperidyl]propoxy]-2-methyl-phenyl]pyr idine-2-carboxylic acid (37.8 mg, 37.4 µmol, 38.6% yield, 96.6% purity) as a white solid. MS (ESI) m/z: 974.4 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.86 (s, 1H), 9.82 (s, 1H), 8.17 (s, 1H), 8.07 - 7.99 (m, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 8.8 Hz, 2H), 7.49 - 7.40 (m, 3H), 7.39 - 7.31 (m, 2H), 7.25 - 7.19 (m, 1H), 6.92 (d, J = 8.4 Hz, 2H), 6.79 (d, J = 1.6 Hz, 1H), 6.74 - 6.66 (m, 1H), 4.97 (s, 2H), 4.37 - 4.27 (m, 1H), 3.97 - 3.88 (m, 7H), 3.17 - 3.15 (m, 2H), 3.05 - 3.00 (m, 2H), 2.66 - 2.55 (m, 6H), 2.35 - 2.31 (m, 1H), 2.21 - 2.14 (m, 1H), 2.03 (s, 3H), 1.74 - 1.64 (m, 2H), 1.55 - 1.46 (m, 2H), 1.44 - 1.35 (m, 4H), 0.92 (s, 3H). Example 201. Preparation of Compound 265 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3, 4-dihydroisoquinolin-2(1H)-yl)-3-(4-((7-(2-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2-oxoet hyl)-7-azaspiro[3.5]nonan-2- yl)methoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 2-((4-bromo-3-methylphenoxy)methyl)-7- azaspiro[3.5]nonane-7-carboxylate A mixture of tert-butyl 2-(hydroxymethyl)-7-azaspiro[3.5]nonane-7-carboxylate (600 mg, 2.35 mmol, 1 equiv.), 4-bromo-3-methyl-phenol (571.31 mg, 3.06 mmol, 1.3 equiv.), and 2- (tributyl-λ 5 -phosphanylidene)acetonitrile (850.66 mg, 3.53 mmol, 1.5 equiv. ) in toluene (12 mL) was degassed and purged with N 2 three times. The mixture was then stirred at 120 °C for 5 hours under N2 atmosphere. The reaction mixture was diluted with water (10 mL) and extracted with EtOAc (25 mL × 3). The combined organic layers were washed with brine (25 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, petroleum ether: ethyl acetate = 10: 1) to give tert-butyl 2-((4-bromo-3-methylphenoxy)methyl)-7- azaspiro[3.5]nonane-7-carboxylate (693 mg, 1.6 mmol, 69.4% yield) as yellow oil. Step B. Procedure for preparation of 2-[(4-bromo-3-methyl-phenoxy)methyl]-7- azaspiro[3.5]nonane A mixture of tert-butyl 2-((4-bromo-3-methylphenoxy)methyl)-7-azaspiro[3.5]nonane-7- carboxylate (693 mg, 1.63 mmol, 1 equiv.) in HCl/dioxane (6 mL) and DCM (6 mL) was degassed and purged with N2 three times. The mixture was then stirred at 25 °C for 16 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give 2-[(4-bromo-3-methyl-phenoxy)methyl]-7-azaspiro[3.5]nonane (1.54 g, crude) as white solid. 1 HNMR (400 MHz, CDCl3) δ = 9.42 - 9.36 (m, 1H), 7.40 (d, J = 8.4 Hz, 1H), 6.78 (d, J = 2.8 Hz, 1H), 6.59 (dd, J = 8.4, 2.8 Hz, 1H), 3.88 (d, J = 5.6 Hz, 2H), 3.18 - 3.12 (m, 2H), 3.12 - 3.05 (m, 2H), 2.76 - 2.69 (m, 1H), 2.36 (s, 3H), 2.09 - 2.02 (m, 2H), 2.02 - 1.99 (m, 2H), 1.94 - 1.88 (m, 2H), 1.81 - 1.75 (m, 2H) Step C. Procedure for preparation of ethyl 2-[2-[(4-bromo-3-methyl-phenoxy)methyl]-7- azaspiro[3.5]nonan-7-yl]acetate A mixture of 2-[(4-bromo-3-methyl-phenoxy)methyl]-7-azaspiro[3.5]nonane (1.54 g, 4.27 mmol, 1 equiv., HCl), ethyl 2-bromoacetate (712.97 mg, 4.27 mmol, 472.17 µL, 1 equiv.), and K2CO3 (1.77 g, 12.81 mmol, 3 equiv. ) in DMF (23 mL) was degassed and purged with N2 three times. The mixture was then stirred at 60 °C for 2 hours under N2 atmosphere. The reaction mixture was diluted with water (25 mL) and extracted with EtOAc (25 mL × 3). The combined organic layers were washed with brine (25 mL) and dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, petroleum ether : ethyl acetate = 10 : 1) to give ethyl 2-[2-[(4- bromo-3-methyl-phenoxy)methyl]-7-azaspiro[3.5]nonan-7-yl]ace tate (252 mg, 614.1 µmol, 14.3% yield) as a colorless oil. MS (ESI) m/z: 410.2 [M+H] + 1 HNMR (400 MHz, CDCl3) δ = 7.39 (d, J = 8.8 Hz, 1H), 6.78 (d, J = 2.8 Hz, 1H), 6.60 (dd, J = 8.8, 3.2 Hz, 1H), 4.25 - 4.15 (m, 2H), 3.88 (d, J = 6.4 Hz, 2H), 3.58 - 3.41 (m, 1H), 3.35 - 3.10 (m, 2H), 2.76 - 2.61 (m, 2H), 2.36 (s, 3H), 2.01 - 1.95 (m, 2H), 1.83 - 1.74 (m, 2H), 1.74 - 1.67 (m, 2H), 1.67 - 1.64 (m, 2H), 1.64 - 1.60 (m, 2H), 1.36 - 1.34 (m, 1H), 1.35 (s, 1H), 1.29 (t, J = 7.2 Hz, 3H) Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-((7-(2-ethoxy-2-oxoethyl)- 7-azaspiro[3.5]nonan-2- yl)methoxy)-2-methylphenyl)picolinate Ethyl 2-[2-[(4-bromo-3-methyl-phenoxy)methyl]-7-azaspiro[3.5]nonan -7-yl]acetate (100 mg, 243.70 µmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)picolinate (149.28 mg, 243.70 µmol, 1 equiv.), Ad 2 nBuP Pd G 3 (cataCXium® A Pd G 3 ) (17.75 mg, 24.37 µmol, 0.1 equiv.), and KF (1.5 M, 487.40 µL, 3 equiv.) were taken up into a microwave tube in dioxane (1 mL). The sealed tube was heated at 100 °C for 1 hour under microwave. The reaction mixture was diluted with water (25 mL) and extracted with EtOAc (25 mL × 3). The combined organic layers were washed with brine (25mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , petroleum ether : ethyl acetate =1: 1) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(4-((7-(2-ethoxy-2- oxoethyl)-7-azaspiro[3.5]nonan-2-yl)methoxy)-2-methylphenyl) picolinate(147 mg, 180.1 µmol, 73.9% yield) as a light yellow solid. MS (ESI) m/z: 816.3 [M+H] + 1 HNMR (400 MHz, CDCl3) δ = 7.86 (d, J = 7.6 Hz, 1H), 7.76 (d, J = 8.4 Hz, 1H), 7.58 (d, J =11.2 Hz, 1H), 7.48 - 7.42 (m, 1H), 7.40 (d, J = 8.8 Hz, 1H), 7.36 (d, J = 5.2 Hz, 1H), 7.35 (s, 1H), 7.30 (d, J = 6.0 Hz, 1H), 7.00 - 6.96 (m, 1H), 6.88 (d, J = 8.8 Hz, 1H), 6.77 (d, J = 2.8 Hz, 1H), 6.71 - 6.67 (m, 1H), 5.07 - 4.98 (m, 2H), 4.28 - 4.14 (m, 2H), 4.11 - 4.08 (m, 2H), 3.93 (d, J = 6 Hz, 1H), 3.10 - 3.05 (m, 2H), 2.78 - 2.69 (m, 1H), 2.10 (s, 3H), 2.02 - 1.94 (m, 1H), 1.94 - 1.84 (m, 2H), 1.76 (d, J = 3.6 Hz, 2H), 1.74 - 1.68 (m, 2H), 1.32 - 1.29 (m, 1H), 1.32 - 1.29 (m, 1H), 1.28 - 1.26 (m, 2H), 1.25 (s, 2H), 1.19 (s, 9H), 1.09 (s, 2H) Step E. Procedure for preparation of 2-[2-[[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl ]-3-methyl- phenoxy]methyl]-7-azaspiro[3.5]nonan-7-yl] acetic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4-((7-(2-ethoxy-2-oxoethyl)-7-azaspiro[3.5]nonan-2-yl )methoxy)-2- methylphenyl)picolinate(147 mg, 180.14 µmol, 1 equiv.) and LiOH•H2O (37.80 mg, 900.72 µmol, 5 equiv. ) in THF (1.5 mL) and H 2 O (1.5 mL) was degassed and purged with N 2 three times. The mixture was then stirred at 25 °C for 16 hours under N2 atmosphere. The mixture was concentrated under reduced pressure to give a residue. To this residue, HCl (1 M,1 mL) was slowly added dropwise to until pH=3~4, when a yellow solid was precipitated and collected by filtration. The yellow solid was dissolved in DCM (10 mL) and concentrated under reduced pressure to give 2-[2-[[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-3-methyl- phenoxy]methyl]-7- azaspiro[3.5]nonan-7-yl] acetic acid (101 mg, crude). 1 HNMR (400 MHz, DMSO) δ = 13.18 - 12.53 (m, 1H), 8.01 (d, J = 8.0 Hz, 1H), 7.77 (d, J = 8.0 Hz, 1H), 7.59 (d, J = 7.6 Hz, 1H), 7.49 - 7.45 (m, 1H), 7.43 (d, J = 2.8 Hz, 1H), 7.39 (d, J = 7.2 Hz, 1H), 7.37 - 7.34 (m, 1H), 7.34 - 7.31 (m, 1H), 6.97 - 6.90 (m, 1H), 6.856 (d, J = 8.4 Hz, 1H), 6.81 (d, J = 1.6 Hz, 1H), 6.71 (d, J = 8.0 Hz, 1H), 4.96 (s, 2H), 3.92 (d, J = 6 Hz, 2H), 3.87 - 3.82 (m, 2H), 3.24 (s, 2H), 3.06-2.99 (m, 2H), 2.93 (s, 2H), 2.85 (s, 2H), 2.67 - 2.61 (m, 1H), 2.00 (s, 3H), 1.94 - 1.89 (m, 2H), 1.75 (s, 2H), 1.67 (s, 2H), 1.64 - 1.55 (m, 2H), 1.06 - 0.99 (m, 9H) Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-((7-(2-((3-(2,6-dioxopiper idin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)-7-azaspiro[3.5]nonan-2-yl)me thoxy)-2- methylphenyl)picolinate A mixture of 2-[2-[[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2- yl]-2-tert-butoxycarbonyl-3-pyridyl]-3-methyl-phenoxy]methyl ]-7-azaspiro[3.5]nonan-7-yl] acetic acid (101 mg, 128.18 µmol, 1 equiv. ), 3-(6-amino-1H-indazol-3-yl)piperidine-2,6- dione(39.73 mg, 153.81 µmol, 1.2 equiv. ), and DIEA (49.70 mg, 384.54 µmol, 66.98 µL, 3 equiv.) in DMF (1 mL) was degassed and purged with N2 three times. The mixture was stirred at 25°C for 5 minutes. After 5 minutes, HATU (73.11 mg, 192.27 µmol, 1.5 equiv.) was added, and the mixture was stirred at 25 °C for 2 hours under N 2 atmosphere. To the mixture was added water (2 mL) dropwise, and a pink solid was precipitated and collected by filtration. The pink solid was dissolved in DCM (10 mL) and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-((7- (2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)am ino)-2-oxoethyl)-7- azaspiro[3.5]nonan-2-yl)methoxy)-2-methylphenyl)picolinate (116 mg, crude). MS (ESI) m/z: 1028.6 [M+H] + 1 HNMR (400 MHz, DMSO) δ = 12.90 - 12.79 (m, 1H), 10.89 (s, 1H), 10.53 - 10.18 (m, 1H), 8.02 (d, J = 4.0 Hz, 1H), 8.01 (s, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.67 (d, J = 8.4 Hz, 1H), , 7.60 (d, J = 7.6 Hz, 1H), 7.47 (d, J = 8.4 Hz, 1H), 7.44 - 7.43 (m, 1H), 7.40 (d, J = 7.6 Hz, 1H), 7.36 (d, J = 5.6 Hz, 1H), 7.33 (d, J = 7.6 Hz, 1H), 7.17 (d, J = 8.8 Hz, 1H), 6.92 (d, J =9.2 Hz, 1H), 6.86 (d, J = 8.4 Hz, 1H), 6.82 (s, 1H), 6.73 (d, J = 8.4 Hz, 1H), 4.97 (s, 2H), 4.33 (dd, J = 9.6, 5.2 Hz, 1H), 3.87- 3.84 (m, 2H), 3.05- 3.02 (m, 2H), 2.66 (dd, J = 9.6, 4.8 Hz, 2H), 2.39 - 2.33 (m, 1H), 2.25 - 2.11 (m, 2H), 2.01 (s, 3H), 1.94 (d, J = 10.4 Hz, 2H), 1.89 (s, 2H), 1.81 (s, 2H), 1.73 (s, 2H), 1.68 (s, 2H), 1.66 - 1.59 (m, 2H), 1.23 (s, 2H), 1.20 - 1.14 (m, 2H), 1.04 (s, 9H) Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3, 4- dihydroisoquinolin-2(1H)-yl)-3-(4-((7-(2-((3-(2,6-dioxopiper idin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)-7-azaspiro[3.5]nonan-2-yl)me thoxy)-2- methylphenyl)picolinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4-((7-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-in dazol-6-yl)amino)-2-oxoethyl)- 7-azaspiro[3.5]nonan-2-yl)methoxy)-2-methylphenyl)picolinate (116 mg, 112.82 µmol, 1 equiv.) in DCM (1 mL) and TFA (1 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 16 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3, 4-dihydroisoquinolin- 2(1H)-yl)-3-(4-((7-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl -1H-indazol-6-yl)amino)-2- oxoethyl)-7-azaspiro[3.5]nonan-2-yl)methoxy)-2-methylphenyl) picolinic acid (31.3 mg, 32.2 µmol, 28.5% yield) as a yellow solid. MS (ESI) m/z: 487.1 [M/2+H] + 1 HNMR (400 MHz, DMSO) δ = 10.87 (s, 1H), 9.83 (s, 1H), 8.20 - 8.14 (m, 1H), 8.024 (d, J = 8.8 Hz, 1H), 7.78 (d, J = 8 Hz, 1H), 7.63 (d, J = 4.4 Hz, 1H), 7.61 (d, J = 2.8 Hz, 1H), 7.47 (d, J = 7.6 Hz, 1H), 7.45 (s, 1H), 7.43 (s, 1H), 7.39 (d, J = 6.8 Hz, 1H), 7.36 (s, 1H), 7.33 (d, J = 6.8 Hz, 1H), 7.23 (d, J = 10 Hz, 1H), 6.93 (d, J = 4.0 Hz, 1H), 6.91 (d, J = 3.6 Hz, 1H), 6.78 (s, 1H), 6.69 (d, J = 8.8 Hz, 1H), 4.96 (s, 2H), 4.32 (dd, J = 9.6, 4.8 Hz, 1H), 3.95 - 3.87 (m, 8H), 3.01 (t, J = 9.6 Hz,2H), 2.68 - 2.64 (m, 2H), 2.64 - 2.60 (m, 2H), 2.43 - 2.37 (m, 2H), 2.33 (s, 2H), 2.24 - 2.11 (m, 2H), 2.02 (s, 3H), 1.92 (t, J = 9.6 Hz, 2H), 1.66 - 1.72 (m, 2H), 1.63 - 1.57 (m, 4H) Example 202. Preparation of Compound 277 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(2-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2- oxoethyl)-2- azaspiro[3.3]heptan-6-yl)ethoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(2-methoxy-2-oxoethyl)-2- azaspiro[3.3]heptane-2-carboxylate To a mixture of 2-(2-(tert-butoxycarbonyl)-2-azaspiro[3.3]heptan-6-yl)acetic acid (500 mg, 1.96 mmol, 1 equiv.) and Cs2CO3 (765.71 mg, 2.35 mmol, 1.2 equiv.) in DMF (1 mL) was added MeI (333.57 mg, 2.35 mmol, 146.30 uL, 1.2 equiv.). The mixture was stirred at 25 °C for 2 hours, diluted with water (10 mL), and extracted with EtOAc (5 mL × 3). The combined organic layers were washed with brine (5 mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0 ~ 36 % ethyl acetate / petroleum ether) to give tert-butyl 6-(2- methoxy-2-oxoethyl)-2-azaspiro[3.3]heptane-2-carboxylate (400 mg, 1.4 mmol, 75.8% yield) as a white solid. MS (ESI) m/z: 214.2 [M+H] + . Step B. Procedure for preparation of tert-butyl 6-(2-hydroxyethyl)-2- azaspiro[3.3]heptane-2-carboxylate A mixture of tert-butyl 6-(2-methoxy-2-oxoethyl)-2-azaspiro[3.3]heptane-2-carboxylat e (400 mg, 1.49 mmol, 1 equiv.), LiAlH4 (67.63 mg, 1.78 mmol, 1.2 equiv.) in THF (1 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 0 °C for 1 hour under N 2 atmosphere. The reaction mixture was quenched by the addition of H 2 O (0.135 mL), 15% NaOH (0.405 mL), and H2O 0.135 mL). The resulting mixture was then diluted with water (3 mL) and extracted with ethyl acetate (2 mL × 3). The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0 ~ 8% DCM / MeOH) to give tert- butyl 6-(2-hydroxyethyl)-2-azaspiro[3.3]heptane-2-carboxylate (350 mg, 1.4 mmol, 97.6% yield) as a white solid. MS (ESI) m/z: 186.3 [M+H] + . Step C. Procedure for preparation of tert-butyl 6-(2-(3-bromo-2-methylphenoxy)ethyl)-2- azaspiro[3.3]heptane-2-carboxylate To a solution of tert-butyl 6-(2-hydroxyethyl)-2-azaspiro[3.3]heptane-2-carboxylate (350 mg, 1.45 mmol, 1 equiv.) and 3-bromo-2-methyl-phenol (325.51 mg, 1.74 mmol, 1.2 equiv.) in toluene (15 mL) was added 2-(tributyl-λ 5 -phosphanylidene)acetonitrile (700.08 mg, 2.90 mmol, 2 equiv.). The mixture was stirred at 120 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0 ~ 18 % ethyl acetate / petroleum ether) to give tert- butyl 6-(2-(3-bromo-2-methylphenoxy)ethyl)-2-azaspiro[3.3]heptane- 2-carboxylate (300 mg, 731.0 µmol, 50.4% yield) as a white solid. MS (ESI) m/z: 354.9 [M+H] + . Step D. Procedure for preparation of 6-(2-(3-bromo-2-methylphenoxy)ethyl)-2- azaspiro[3.3]heptane A solution of tert-butyl 6-(2-(3-bromo-2-methylphenoxy)ethyl)-2-azaspiro[3.3]heptane- 2- carboxylate (200 mg, 487.40 µmol, 1 equiv.) in TFA (1 mL) and DCM (3 mL) was stirred at 25 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0 ~ 68 % ethyl acetate / petroleum ether) to give 6-(2-(3-bromo-2-methylphenoxy)ethyl)-2- azaspiro[3.3]heptane (250 mg, crude) as a white oil. MS (ESI) m/z: 310.0 [M+H] + . Step E. Procedure for preparation of ethyl 2-(6-(2-(3-bromo-2-methylphenoxy)ethyl)-2- azaspiro[3.3]heptan-2-yl)acetate A mixture of 6-(2-(3-bromo-2-methylphenoxy)ethyl)-2-azaspiro[3.3]heptane (250 mg, 805.86 µmol, 1 equiv.) and ethyl 2-oxoacetate (98.72 mg, 967.03 µmol, 1.2 equiv.) in DCM (4 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 25 °C for 8 hours under N2 atmosphere. NaBH(OAc)3 (170.79 mg, 805.86 µmol, 1 equiv.) was added to the mixture, which was stirred at 25 °C for 1 hour. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0 ~ 67% ethyl acetate / petroleum ether) to give ethyl 2-(6-(2-(3-bromo-2-methylphenoxy)ethyl)-2-azaspiro[3.3]hepta n-2-yl)acetate (250 mg, 630.8µmol, 78.2% yield) as a white oil. MS (ESI) m/z: 396.2 [M+H] + . Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(2-(2-ethoxy-2-oxoethyl )-2-azaspiro[3.3]heptan-6- yl)ethoxy)-2-methylphenyl)picolinate A mixture of ethyl 2-(6-(2-(3-bromo-2-methylphenoxy)ethyl)-2-azaspiro[3.3]hepta n-2- yl)acetate (210 mg, 529.88 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3, 2-dioxaborolan-2-yl)pyridine-2- carboxylate (421.95 mg, 688.84 µmol, 1.3 equiv.), [2-(2- aminophenyl)phenyl]palladium(1 + );bis(1-adamantyl)-butyl-phosphane;methanesulfonate (38.59 mg, 52.99 µmol, 0.1 equiv.), and K 2 CO 3 (1.5 M, 529.88 µL, 1.5 equiv.) in dioxane (3 mL) was degassed and purged with N2 three times. The mixture was stirred at 100 °C for 1 hour under N2 atmosphere under microwave. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0 ~ 65 % ethyl acetate / petroleum ether) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(2-(2-(2-ethoxy-2- oxoethyl)-2-azaspiro[3.3]heptan-6-yl)ethoxy)-2-methylphenyl) picolinate (200 mg, 249.3 µmol, 47.0% yield) as a yellow oil. MS (ESI) m/z: 802.3 [M+H] + . Step G. Procedure for preparation of 2-(6-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyri din-3-yl)-2- methylphenoxy)ethyl)-2-azaspiro[3.3]heptan-2-yl)acetic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(2-(2-(2-ethoxy-2-oxoethyl)-2-azaspiro[3.3]heptan-6 -yl)ethoxy)-2- methylphenyl)picolinate (200 mg, 249.38 µmol, 1 equiv.), and LiOH•H 2 O (52.32 mg, 1.25 mmol, 5 equiv.) in THF (2 mL) and H2O (1 mL) was stirred at 25 °C for 12 hours. The reaction mixture was diluted with H2O (5 mL) and concentrated as a turbid liquid. Then the pH of the turbid liquid was adjusted to 3 with 1 N HCl (5 mL), and the residue was extracted with DCM / MEOH (20:1), The combined organic layers were filtered and concentrated. The residue was used in the next step without further purification. The compound 2-(6-(2-(3-(6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)ethyl)-2-azaspi ro[3.3]heptan-2-yl)acetic acid (196 mg, crude) was obtained as a yellow solid. MS (ESI) m/z: 774.3 [M+H] + . Step H. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(2-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)-2-azaspiro[3.3]heptan-6-yl)e thoxy)-2- methylphenyl)picolinate A mixture of 2-(6-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)eth yl)-2-azaspiro[3.3]heptan-2- yl)acetic acid (176 mg, 227.41 µmol, 1 equiv.), 3-(6-amino-1-methyl-indazol-3-yl)piperidine- 2,6-dione (70.48 mg, 272.89 µmol, 1.2 equiv.), and EDCI (65.39 mg, 341.11 µmol, 1.5 equiv.) in pyridine (1 mL) was stirred at 25 °C for 2 hours. The reaction mixture was diluted with H2O (10 mL × 3) and extracted with ethyl acetate (8 mL × 3). The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was used in the next step without further purification. The compound tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(2-(2- ((3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)-2-azaspiro[3.3]hep tan-6-yl)ethoxy)-2- methylphenyl)picolinate (150 mg, 147.9 µmol, 65.0% yield) was obtained as a yellow oil. MS (ESI) m/z: 1014.3 [M+H] + . Step I. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(2-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)-2-azaspiro[3.3]heptan-6-yl)e thoxy)-2- methylphenyl)picolinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(2-(2-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2- oxoethyl)-2-azaspiro[3.3]heptan-6-yl)ethoxy)-2-methylphenyl) picolinate (150 mg, 147.90 µmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL) was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(2-(2-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2- oxoethyl)-2-azaspiro[3.3]heptan-6-yl)ethoxy)-2-methylphenyl) picolinic acid (11.8 mg, 12.3 µmol, 8.3% yield, 99.6% purity) as a yellow solid. MS (ESI) m/z: 958.3 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.87 (s, 1H), 9.84 (s, 1H), 8.04 - 7.99 (m, 2H), 7.80 - 7.75 (m, 1H), 7.64 - 7.59 (m, 2H), 7.47 - 7.32 (m, 5H), 7.19 - 7.15 (m, 1H), 7.10 - 7.04 (m, 1H), 6.92 - 6.82 (m, 2H), 6.69 - 6.59 (m, 1H), 5.04 - 4.93 (m, 2H), 4.31 (dd, J = 4.8, 9.6 Hz, 1H), 3.92 - 3.89 (m, 6H), 3.23 - 3.21 (m, 4H), 3.04 - 3.00 (m, 2H), 2.69 - 2.62 (m, 4H), 2.34 - 2.27 (m, 4H), 2.23 - 2.10 (m, 2H), 1.89 (s, 3H), 1.85 - 1.78 (m, 4H) Example 203. Preparation of Compound 286 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[2-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-2-azaspiro[3.3]heptan-6- yl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 6-[(E)-3-ethoxy-3-oxo-prop-1-enyl]-2- azaspiro[3.3]heptane-2-carboxylate A mixture of tert-butyl 6-formyl-2-azaspiro[3.3]heptane-2-carboxylate (300 mg, 1.33 mmol, 1 equiv.) in THF (1 mL) was added LiHMDS (1 M, 1.60 mL, 1.2 equiv.) at -78°C for 0.5 hour under N2 atmosphere, and then ethyl 2-diethoxyphosphorylacetate (358.25 mg, 1.60 mmol, 317.04 µL, 1.2 equiv.) in THF (0.5 mL) was added dropwise at -78°C. The resulting mixture was stirred at 20 °C for 1.5 hours under N 2 atmosphere. The reaction mixture was quenched by addition of saturated NH4Cl (5 mL) at 0°C, diluted with EtOAc (1 mL), and extracted with (1 mL × 3). The combined organic layers were washed with H2O (3 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0 ~ 20 % ethyl acetate/petroleum ether) to give tert-butyl 6-[(E)-3-ethoxy-3-oxo-prop-1-enyl]-2-azaspiro[3.3]heptane-2- carboxylate (230 mg, 778.6 µmol, 58.4% yield) as a colorless oil. MS (ESI) m/z: 240.1 [M-56+H] + . Step B. Procedure for preparation of tert-butyl 6-(3-ethoxy-3-oxo-propyl)-2- azaspiro[3.3]heptane-2-carboxylate A mixture of tert-butyl 6-[(E)-3-ethoxy-3-oxo-prop-1-enyl]-2-azaspiro[3.3]heptane-2- carboxylate (230 mg, 778.68 µmol, 1 equiv.), Pd/C (50 mg, 80% purity), in MeOH (2 mL) was stirred at 25 °C for 2 hours under H 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give tert-butyl 6-(3-ethoxy-3-oxo-propyl)-2- azaspiro[3.3]heptane-2-carboxylate (300 mg, crude) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 4.03 (q, J = 7.2 Hz, 2H), 3.85 - 3.66 (m, 4H), 2.22 - 2.14 (m, 4H), 2.03 (d, J = 7.6, 15.4 Hz, 1H), 1.76 - 1.69 (m, 2H), 1.60 - 1.52 (m, 2H), 1.35 (s, 9H), 1.17 (t, J = 7.2 Hz, 3H). Step C. Procedure for preparation of tert-butyl 6-(3-hydroxypropyl)-2- azaspiro[3.3]heptane-2-carboxylate To a mixture of tert-butyl 6-(3-ethoxy-3-oxo-propyl)-2-azaspiro[3.3]heptane-2-carboxyla te (300 mg, 1.01 mmol, 1 equiv.) in THF (3 mL) was added LiAlH4 (32.54 mg, 857.46 µmol, 0.85 equiv.). The reaction mixture was stirred at 0 °C for 2 hours and was quenched by addition Na 2 SO 4 •10H 2 O (1 g) under 0 °C and N 2 atmosphere. The resulting mixture was filtered, and the filtrate was concentrated under reduced pressure to give a residue. The residue was then purified by flash silica gel chromatography (eluent of 0 ~ 40 % ethyl acetate/petroleum ether) to give tert-butyl 6-(3-hydroxypropyl)-2-azaspiro[3.3]heptane-2-carboxylate (210 mg, 822.3 µmol, 81.5% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 4.31 (t, J = 5.2 Hz, 1H), 3.85 - 3.68 (m, 4H), 3.37 - 3.32 (m, 2H), 2.24 - 2.16 (m, 2H), 2.07 - 1.98 (m, 1H), 1.74 - 1.65 (m, 2H), 1.35 (s, 9H), 1.33 - 1.28 (m, 4H) Step D. Procedure for preparation of tert-butyl 6-[3-(3-bromo-2-methyl- phenoxy)propyl]-2-azaspiro[3.3]heptane-2-carboxylate A mixture of tert-butyl 6-(3-hydroxypropyl)-2-azaspiro[3.3]heptane-2-carboxylate (210.00 mg, 822.3 µmol, 1.2 equiv.), 3-bromo-2-methyl-phenol (128.18 mg, 685.33 µmol, 1 equiv.), and 2-(tributyl-λ5-phosphanylidene)acetonitrile (248.11 mg, 1.03 mmol, 1.5 equiv.) in toluene (1 mL) was stirred at 120 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The mixture was purified by flash silica gel chromatography (eluent of 0 ~ 20 % ethyl acetate/petroleum ether) to give tert-butyl 6-[3-(3-bromo-2-methyl- phenoxy)propyl]-2-azaspiro[3.3]heptane-2-carboxylate (290 mg, 683.3 µmol, 99.7% yield) as a colorless oil. MS (ESI) m/z: 446.0[M+23] +. Step E. Procedure for preparation of 6-[3-(3-bromo-2-methyl-phenoxy)propyl]-2- azaspiro[3.3]heptane A mixture of tert-butyl 6-[3-(3-bromo-2-methyl-phenoxy)propyl]-2-azaspiro[3.3]heptan e-2- carboxylate (290 mg, 683.36 µmol, 1 equiv.) in TFA (0.5 mL) and DCM (1.5 mL) was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give 6- [3-(3-bromo-2-methyl-phenoxy)propyl]-2-azaspiro[3.3]heptane (200 mg, 616.8 µmol, 90.2% yield, crude) as a yellow oil. MS (ESI) m/z: 325.0[M+H] + ( 80 Br) . Step F. Procedure for preparation of ethyl 2-[6-[3-(3-bromo-2-methyl-phenoxy)propyl]- 2-azaspiro[3.3]heptan-2-yl]acetate A mixture of 6-[3-(3-bromo-2-methyl-phenoxy)propyl]-2-azaspiro[3.3]heptan e (180 mg, 555.12 µmol, 1 equiv.) and ethyl 2-oxoacetate (136.01 mg, 666.14 µmol, 50% purity, 1.2 in DCM (2 mL) was stirred at 25 °C for 2 hours. To the mixture was added NaBH(OAc)3 (352.96 mg, 1.67 mmol, 3 equiv.). The mixture was then stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give ethyl 2-[6-[3-(3-bromo-2- methyl-phenoxy)propyl]-2-azaspiro[3.3]heptan-2-yl]acetate (200 mg, crude) as a yellow oil. MS (ESI) m/z: 411.0[M+H] +. ( 80 Br) Step G. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[2-(2-ethoxy-2-oxo- ethyl)-2- azaspiro[3.3]heptan-6-yl]propoxy]-2-methyl-phenyl]pyridine-2 -carboxylate Ethyl 2-[6-[3-(3-bromo-2-methyl-phenoxy)propyl]-2-azaspiro[3.3]hep tan-2-yl]acetate (35 mg, 85.29 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (73.76 mg, 102.35 µmol, 1.2 equiv.), KF (14.87 mg, 255.88 µmol, 5.99 µL, 3 equiv.), and [2- (2-aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-butyl-p hosphane;methanesulfonate (12.42 mg, 17.06 µmol, 0.2 equiv.) were taken up into a microwave tube in dioxane (0.3 mL) and H 2 O (0.03 mL) .The sealed tube was heated at 100 °C for 1 hour under microwave. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, petroleum ether: ethyl acetate = 1:1). The compound tert-butyl 6- [8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinol in-2-yl]-3-[3-[3-[2-(2-ethoxy- 2-oxo-ethyl)-2-azaspiro[3.3]heptan-6-yl]propoxy]-2-methyl-ph enyl]pyridine-2-carboxylate (50 mg, 53.8 μmol, 63.1% yield, 87.9% purity) was obtained as a yellow oil. MS (ESI) m/z: 816.3[M+H] + . 1 H NMR (400 MHz, CDCl 3 -d) δ = 7.88 - 7.82 (m, 1H), 7.57 (d, J = 8.0 Hz, 1H), 7.47 - 7.40 (m, 1H), 7.38 - 7.33 (m, 3H), 7.08 (t, J = 8.0 Hz, 1H), 6.91 - 6.88 (m, 1H), 6.78 (d, J = 7.6 Hz, 1H), 6.69 (d, J = 6.4 Hz, 1H), 5.30 (s, 1H), 4.18 (d, J = 6.4 Hz, 1H), 4.12 - 4.07 (m, 2H), 3.94 (t, J = 6.0 Hz, 2H), 3.10 - 3.00 (m, 2H), 2.39 - 2.25 (m, 2H), 2.21 - 2.11 (m, 1H), 2.08 - 2.00 (m, 1H), 1.98 (s, 3H), 1.84 - 1.74 (m, 2H), 1.73 - 1.67 (m, 3H), 1.29 - 1.24 (m, 6H), 1.14 (s, 9H), 0.93 - 0.79 (m, 2H), 0.07 (s, 6H) Step H. Procedure for preparation of 2-[6-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyr idyl]-2-methyl- phenoxy]propyl]-2-azaspiro[3.3]heptan-2-yl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[3-[2-(2-ethoxy-2-oxo-ethyl)-2-azaspiro[3.3]hepta n-6-yl]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (50 mg, 61.27 µmol, 1 equiv.) in THF (1 mL) was added LiOH•H 2 O (12.86 mg, 306.37 µmol, 5 equiv.) and H 2 O (0.3 mL). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was treated with water (6 mL). The pH of the resulting mixture was adjusted to around 3 by progressively adding dilute HCl (1M). The mixture was filtered to give 2-[6-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H-isoquinolin-2-yl]-2-tert- butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]propyl]-2-azaspir o[3.3]heptan-2-yl]acetic acid (25 mg, 30.6 µmol, 50.0% yield, 96.7% purity) as a yellow solid. MS (ESI) m/z: 788.6 [M+H] + . Step I. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[2-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-2-azaspiro[3.3]heptan-6-yl]propoxy] -2-methyl- phenyl]pyridine-2-carboxylate To a solution of 2-[6-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]propyl]-2- azaspiro[3.3]heptan-2-yl]acetic acid (20 mg, 25.38 µmol, 1 equiv.) and 3-(6-amino-1-methyl- indazol-3-yl)piperidine-2,6-dione (7.87 mg, 30.46 µmol, 1.2 equiv.) in pyridine (0.3 mL) was added EDCI (5.84 mg, 30.46 µmol, 1.2 eq). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was diluted with water (10 mL) and filtered to give tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[2-[2- [[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]amino]-2-oxo-ethyl]-2-azaspiro[3.3]hepta n-6-yl]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (20 mg, 17.0 µmol, 67.1% yield, 87.6% purity) as a white solid. MS (ESI) m/z: 514.9[1/2M+H] +. Step J. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[2-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-2-azaspiro[3.3]heptan-6-yl]propoxy]-2 -methyl-phenyl]pyridine- 2-carboxylic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[2-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indaz ol-6-yl]amino]-2-oxo-ethyl]-2- azaspiro[3.3]heptan-6-yl]propoxy]-2-methyl-phenyl]pyridine-2 -carboxylate (20 mg, 19.45 µmol, 1 equiv.) in TFA (0.2 mL) and DCM (1 mL) was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[3-[2-[2-[[3-(2,6-dioxo-3-piperidyl)- 1-methyl-indazol-6-yl]amino]-2- oxo-ethyl]-2-azaspiro[3.3]heptan-6-yl]propoxy]-2-methyl-phen yl]pyridine-2-carboxylic acid (7.9 mg, 7.7 µmol, 39.5% yield, 94.4% purity) as a yellow solid. MS (ESI) m/z: 486.8 [M/2+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.95 - 10.80 (m, 1H), 9.89 - 9.77 (m, 1H), 8.30 (s, 1H), 8.05 - 7.95 (m, 2H), 7.76 (d, J = 8.0 Hz, 1H), 7.66 - 7.55 (m, 2H), 7.48 - 7.38 (m, 2H), 7.38 - 7.29 (m, 3H), 7.17 (dd, J = 1.2, 8.8 Hz, 1H), 7.08 - 7.00 (m, 1H), 6.82 (dd, J = 4.0, 8.4 Hz, 2H), 6.67 (d, J = 7.6 Hz, 1H), 4.96 (s, 2H), 4.31 (dd, J = 5.2, 9.6 Hz, 1H), 3.90 (s, 3H), 3.34 (s, 2H), 3.22 (d, J = 10.4 Hz, 4H), 3.00 (t, J = 5.2 Hz, 3H), 2.70 - 2.55 (m, 3H), 2.38 - 2.27 (m, 2H), 2.26 - 2.13 (m, 4H), 2.13 - 2.07 (m, 1H), 1.91 (s, 3H), 1.75 - 1.68 (m, 2H), 1.67 - 1.60 (m, 2H), 1.54 - 1.46 (m, 2H) Example 204. Preparation of Compound 268a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[[(3S)-8-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-o xo-ethyl]-8-azaspiro[4.5]decan- 3-yl]oxy]-2-methyl-phenyl]pyridine-2-carboxylic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(3-(((2S)-8- (2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)am ino)-2-oxoethyl)-8- azaspiro[4.5]decan-2-yl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of tert-butyl 3-hydroxy-8-azaspiro[4.5]decane-8- carboxylate To a solution of tert-butyl 3-oxo-8-azaspiro[4.5]decane-8-carboxylate (1 g, 3.95 mmol, 1 equiv.) in EtOH (10 mL) was added NaBH 4 (179.20 mg, 4.74 mmol, 1.2 equiv.) at 0 °C. The mixture was stirred at 20 °C for 2 hours. The reaction mixture was quenched by addition saturated NH4Cl (5 mL) at 0 °C, and then diluted with ethyl acetate (1 mL) and extracted with 3 mL (1 mL × 3). The combined organic layers were washed with H 2 O (3 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was used in the next step without further purification. The compound tert-butyl 3-hydroxy-8- azaspiro[4.5]decane-8-carboxylate (1 g, crude) was obtained as a white oil. Step B. Procedure for preparation of tert-butyl 3-(3-bromo-2-methyl-phenoxy)-8- azaspiro[4.5]decane-8-carboxylate A mixture of 3-bromo-2-methyl-phenol (610.38 mg, 3.26 mmol, 1 equiv.), tert-butyl 3- hydroxy-8-azaspiro[4.5]decane-8-carboxylate (1 g, 3.92 mmol, 1.2 equiv.), and 2-(tributyl-λ 5 - phosphanylidene)acetonitrile (1.18 g, 4.89 mmol, 1.5 equiv.) in toluene (10 mL) was stirred at 120 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~10% ethyl acetate / petroleum ether) to give tert-butyl 3-(3-bromo-2-methyl-phenoxy)-8- azaspiro[4.5]decane-8-carboxylate (600 mg, 1.3 mmol, 39.9% yield, 92% purity) as a yellow oil. MS (ESI) m/z: 367.9[M-56+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 7.15 - 7.11 (m, 1H), 7.10 - 7.05 (m, 1H), 6.93 (d, J = 7.6 Hz, 1H), 4.88 - 4.84 (m, 1H), 3.30 - 3.24 (m, 4H), 2.20 (s, 3H), 2.16 - 2.02 (m, 2H), 1.94 - 1.88 (m, 1H), 1.80 - 1.73 (m, 1H), 1.69 - 1.63 (m, 2H), 1.58 - 1.49 (m, 2H), 1.48 - 1.45 (m, 2H), 1.38 (s, 9H) Step C. Procedure for preparation of tert-butyl (S)-2-(3-bromo-2-methylphenoxy)-8- azaspiro[4.5]decane-8-carboxylate The tert-butyl 3-(3-bromo-2-methyl-phenoxy)-8-azaspiro[4.5]decane-8-carboxy late residue was purified by prep-HPLC to give tert-butyl (R)-2-(3-bromo-2-methylphenoxy)-8- azaspiro[4.5]decane-8-carboxylate (200 mg, 471.29 µmol, 33.33% yield) and tert-butyl (S)-2- (3-bromo-2-methylphenoxy)-8-azaspiro[4.5]decane-8-carboxylat e (220 mg, 518.4 µmol, 36.7% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.14 - 7.11 (m, 1H), 7.10 - 7.05 (m, 1H), 6.93 (d, J = 8.0 Hz, 1H), 4.88 - 4.84 (m, 1H), 3.31 - 3.23 (m, 4H), 2.20 (s, 3H), 2.12 - 2.01 (m, 1H), 1.93 - 1.88 (m, 1H), 1.81 - 1.74 (m, 1H), 1.69 - 1.63 (m, 2H), 1.58 - 1.44 (m, 4H), 1.38 (s, 10H) 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.14 - 7.11 (m, 1H), 7.10 - 7.05 (m, 1H), 6.93 (d, J = 8.0 Hz, 1H), 4.88 - 4.84 (m, 1H), 3.30 - 3.24 (m, 4H), 2.20 (s, 3H), 2.10 - 2.04 (m, 1H), 1.93 - 1.88 (m, 1H), 1.82 - 1.73 (m, 1H), 1.69 - 1.63 (m, 2H), 1.58 - 1.46 (m, 4H), 1.38 (s, 10H) Step D. Procedure for preparation of (S)-2-(3-bromo-2-methylphenoxy)-8- azaspiro[4.5]decane A solution of tert-butyl (S)-2-(3-bromo-2-methylphenoxy)-8-azaspiro[4.5]decane-8- carboxylate (200 mg, 471.29 µmol, 1 equiv.) in HCl / dioxane (2 mL) was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was used in the next step without further purification. The compound (S)-2-(3-bromo- 2-methylphenoxy)-8-azaspiro[4.5]decane (150 mg, crude) was obtained as a colorless oil. MS (ESI) m/z: 324.1[M+H] + Step E. Procedure for preparation of ethyl (S)-2-(2-(3-bromo-2-methylphenoxy)-8- azaspiro[4.5]decan-8-yl)acetate A mixture of (S)-2-(3-bromo-2-methylphenoxy)-8-azaspiro[4.5]decane (140 mg, 431.76 µmol, 1 equiv.), ethyl 2-bromoacetate (72.10 mg, 431.76 µmol, 47.75 µL, 1 equiv.), K2CO3 (179.01 mg, 1.30 mmol, 3 equiv.), and KI (35.84 mg, 215.88 µmol, 0.5 equiv.) in CH 3 CN (2 mL) was stirred at 60 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~40% ethyl acetate / petroleum ether) to give ethyl (S)-2-(2-(3-bromo-2-methylphenoxy)- 8-azaspiro[4.5]decan-8-yl)acetate (110 mg, 268.0 µmol, 62.0% yield) as a yellow oil. MS (ESI) m/z: 410.0[M+H] +1 H NMR (400 MHz, DMSO-d6) δ = 7.14 - 7.11 (m, 1H), 7.10 - 7.05 (m, 1H), 6.93 (d, J = 8.0 Hz, 1H), 4.87 - 4.82 ( m, 1H), 4.09 - 4.04 (m, 2H), 3.16 (s, 2H), 2.48 - 2.35 (m, 4H), 2.19 (s, 3H), 2.10 - 2.00 (m, 1H), 1.88 - 1.83 (m, 1H), 1.81 - 1.71 (m, 1H), 1.65 - 1.51 (m, 5H), 1.45 - 1.43 (m, 2H), 1.17 (t, J = 7.2 Hz, 3H) Step F. Procedure for preparation of tert-butyl (S)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-((8-(2-ethoxy-2-oxoeth yl)-8-azaspiro[4.5]decan-2- yl)oxy)-2-methylphenyl)picolinate A mixture of ethyl (S)-2-(2-(3-bromo-2-methylphenoxy)-8-azaspiro[4.5]decan-8-yl )acetate (100 mg, 243.70 µmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)pyridine-2- carboxylate (179.13 mg, 292.44 µmol, 1.2 equiv.), [2-(2- aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-butyl-phos phane;methanesulfonate (35.50 mg, 48.74 µmol, 0.2 equiv.), and K2CO3 (1.5 M, 487.40 uL, 3 equiv.) in dioxane (2 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 100 °C for 1 hour under N 2 atmosphere under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The mixture was purified by flash silica gel chromatography (eluent of 0~52% ethyl acetate / petroleum ether) to give tert-butyl (S)-6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2( 1H)-yl)-3-(3-((8-(2-ethoxy-2- oxoethyl)-8-azaspiro[4.5]decan-2-yl)oxy)-2-methylphenyl)pico linate (152 mg, 186.2 µmol, 76.4% yield) as a yellow oil. MS (ESI) m/z: 816.3 [M+H] + Step G. Procedure for preparation of (S)-2-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyri din-3-yl)-2- methylphenoxy)-8-azaspiro[4.5]decan-8-yl)acetic acid A mixture of tert-butyl (S)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquino lin- 2(1H)-yl)-3-(3-((8-(2-ethoxy-2-oxoethyl)-8-azaspiro[4.5]deca n-2-yl)oxy)-2- methylphenyl)picolinate (142 mg, 174.02 µmol, 1 equiv.) and LiOH•H2O (36.51 mg, 870.08 µmol, 5 equiv.) in THF (2 mL) and H 2 O (1 mL) was stirred at 25 °C for 16 hours. The reaction mixture was diluted with H2O (5 mL) and concentrated as a turbid liquid. Then the pH of the turbid liquid was adjusted to 3 with 1 N HCl (5 mL), and the residue was extracted with DCM / MeOH (20:1). The combined organic layers were filtered and concentrated. The residue was used in the next step without further purification. The compound (S)-2-(2-(3-(6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)-8-azaspiro[4.5 ]decan-8-yl)acetic acid (140 mg, crude) was obtained as a yellow solid. MS (ESI) m/z: 788.3 [M+H] + Step H. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((2S)-8-(2-((3-(2,6-dioxo piperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)-8-azaspiro[4.5]decan-2-yl)ox y)-2- methylphenyl)picolinate A mixture of (S)-2-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydr oisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)-8-azaspiro[4.5]decan-8- yl)acetic acid (100 mg, 126.91 µmol, 1 equiv.), 3-(6-amino-1-methyl-indazol-3-yl)piperidine- 2,6-dione (39.33 mg, 152.29 µmol, 1.2 equiv.), and EDCI (36.49 mg, 190.36 µmol, 1.5 equiv.) in pyridine (1 mL) was stirred at 25 °C for 2 hours. The mixture was diluted with H2O mL (10 mL × 3) and extracted with ethyl acetate 24 mL (8 mL × 3). The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was used in the next step without further purification. The compound tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((2S)-8- (2-((3-(2,6-dioxopiperidin-3- yl)-1-methyl-1H-indazol-6-yl)amino)-2-oxoethyl)-8-azaspiro[4 .5]decan-2-yl)oxy)-2- methylphenyl)picolinate (100 mg, 97.2 µmol, 76.6% yield) was obtained as a yellow solid. MS (ESI) m/z: 1028.4[M+H] + . Step I. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[[(3S)-8-[2-[[3-(2,6-dioxo-3-piper idyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-8-azaspiro[4.5]decan-3-yl]oxy]-2-meth yl-phenyl]pyridine-2- carboxylic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)- yl)-3-(3-(((2S)-8-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2- oxoethyl)-8-azaspiro[4.5]decan-2-yl)oxy)-2-methylphenyl)pico linic acid] A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((2S)-8-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2- oxoethyl)-8-azaspiro[4.5]decan-2-yl)oxy)-2-methylphenyl)pico linate (90 mg, 87.53 µmol, 1 equiv.) in TFA (0.8 mL) and DCM (0.8 mL) was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[[(3S)-8-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-in dazol-6-yl]amino]-2-oxo-ethyl]- 8-azaspiro[4.5]decan-3-yl]oxy]-2-methyl-phenyl]pyridine-2-ca rboxylic acid [rel-6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((2S)-8-(2-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2-oxoet hyl)-8-azaspiro[4.5]decan-2- yl)oxy)-2-methylphenyl)picolinic acid] (24.2 mg, 24.1 µmol, 27.5% yield, 96.0% purity) as a white solid. MS (ESI) m/z: 972.3[M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.95 - 10.82 (m, 1H), 9.89 - 9.79 (m, 1H), 8.07 - 7.99 (m, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 ( d, J = 8.8 Hz, 2H), 7.50 - 7.42 (m, 3H), 7.40 - 7.32 (m, 2H), 7.22 ( d, J = 8.8 Hz, 1H), 7.08 ( t, J = 8.0 Hz, 1H), 6.94 (d, J = 8.4 Hz, 1H), 6.85 ( d, J = 8.4 Hz, 1H), 6.61 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.79 - 4.70 (m, 1H), 4.32 (dd, J = 5.2, 9.6 Hz, 1H), 3.95 - 3.88 (m, 5H), 3.13 (s, 2H), 3.06 - 2.99 (m, 2H), 2.69 - 2.59 (m, 4H), 2.54 (s, 2H), 2.39 - 2.27 (m, 2H), 2.21 - 2.14 (m, 1H), 2.11 - 2.03 (m, 2H), 1.96 - 1.90 (m, 1H), 1.86 (s, 3H), 1.71 - 1.63 (m, 4H), 1.55 ( d, J = 5.2 Hz, 2H) Example 205. Preparation of Compound 268b 6-(8-(benzo[d]thiazol-2-yl-carbamoyl)-3,4dihydroisoquinolin- 2(1H)-yl)-3-(3-(((2R)-8-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2- oxoethyl)-8- azaspiro[4.5]decan-2-yl)oxy)-2-methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(3-(((2R)-8- (2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)am ino)-2-oxoethyl)-8- azaspiro[4.5]decan-2-yl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of (3R)-3-(3-bromo-2-methyl-phenoxy)-8- azaspiro[4.5]decane A mixture of tert-butyl (3R)-3-(3-bromo-2-methyl-phenoxy)-8-azaspiro[4.5]decane-8- carboxylate (200 mg, 471.29 µmol, 1 equiv.) in HCl / dioxane (2 mL) was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was used in the next step without purification. The compound (3R)-3-(3-bromo-2- methyl-phenoxy)-8-azaspiro[4.5]decane (150 mg, crude) was obtained as a white solid. MS (ESI) m/z: 325.9 [M+H] + Step B. Procedure for preparation of ethyl (R)-2-(2-(3-bromo-2-methylphenoxy)-8- azaspiro[4.5]decan-8-yl)acetate A solution of (3R)-3-(3-bromo-2-methyl-phenoxy)-8-azaspiro[4.5]decane (150 mg, 462.60 µmol, 1 equiv.), ethyl 2-bromoacetate (77.25 mg, 462.60 µmol, 51.16 µL, 1 equiv.), K2CO3 (191.80 mg, 1.39 mmol, 3 equiv.), and KI (38.40 mg, 231.30 µmol, 0.5 equiv.) in CH 3 CN (2 mL) was stirred at 60 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was then purified by flash silica gel chromatography (eluent of 0 ~ 40% ethyl acetate / petroleum ether) to give ethyl (R)-2-(2-(3- bromo-2-methylphenoxy)-8-azaspiro[4.5]decan-8-yl)acetate (150 mg, 365.5 µmol, 79.0% yield) as a yellow oil. MS (ESI) m/z: 411.9 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.14 - 7.04 (m, 2H), 6.93 (d, J = 8.0 Hz, 1H), 4.84 (dd, J = 2.8, 6.0 Hz, 1H), 4.06 (d, J = 7.2 Hz, 2H), 3.16 (s, 2H), 2.46 - 2.39 (m, 4H), 2.21 - 2.16 (m, 1H), 2.19 (s, 3H), 2.06 - 2.00 (m, 1H), 1.88 - 1.82 (m, 1H), 1.65 - 1.59 (m, 2H), 1.58 - 1.49 (m, 4H), 1.47 - 1.42 (m, 2H), 1.19 - 1.15 (m, 3H) Step C. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[[(3R)-8-(2-ethoxy-2-o xo-ethyl)-8- azaspiro[4.5]decan-3-yl]oxy]-2-methyl-phenyl]pyridine-2-carb oxylate A mixture of ethyl (R)-2-(2-(3-bromo-2-methylphenoxy)-8-azaspiro[4.5]decan-8-yl )acetate (150 mg, 365.55 µmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)picolinate (268.70 mg, 438.66 µmol, 1.2 equiv.), K 2 CO 3 (1.5 M, 731.09 µL, 3 equiv.), and [2-(2- aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-butyl-phos phane;methane sulfonate (53.24 mg, 73.11 µmol, 0.2 equiv.) in dioxane (1.5 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 100 °C for 1 hour under N 2 atmosphere under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~52% ethyl acetate / petroleum ether) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[[(3R)-8-(2-ethoxy-2-oxo-e thyl)-8-azaspiro[4.5]decan-3- yl]oxy]-2-methyl-phenyl]pyridine-2-carboxylate (200 mg, 242.6 µmol, 66.3% yield, 99% purity) as a yellow oil. MS (ESI) m/z: 816.7 [M+H] + . Step D. Procedure for preparation of (R)-2-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyri din-3-yl)-2- methylphenoxy)-8-azaspiro[4.5]decan-8-yl)acetic acid A mixture of give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[[(3R)-8-(2-ethoxy-2-oxo-ethyl)-8-aza spiro[4.5]decan-3-yl]oxy]-2- methyl-phenyl]pyridine-2-carboxylate (140 mg, 171.57 µmol, 1 equiv.), LiOH•H 2 O (36.00 mg, 857.83 µmol, 5 equiv.) in THF (2 mL) and H2O (1 mL) was stirred at 25 °C for 16 hours. The reaction mixture was diluted with H2O (5 mL) and concentrated as a turbid liquid. Then the pH of the turbid liquid was adjusted to 3 with 1 N HCl (5 mL), the residue was extracted with DCM / MeOH (20:1), the combined organic layers were filtered and concentrated. The residue was used in the next step without further purification. The compound (R)-2-(2-(3-(6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)-8-azaspiro[4.5 ]decan-8-yl)acetic acid (200 mg, crude) was obtained as a yellow solid. MS (ESI) m/z: 788.4 [M+H] + . Step E. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1h)-yl)-3-(3-(((2r)-8-(2-((3-(2,6-dioxo piperidin-3-yl)-1-methyl-1h- indazol-6-yl)amino)-2-oxoethyl)-8-azaspiro[4.5]decan-2-yl)ox y)-2- methylphenyl)picolinate A mixture of (R)-2-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydr oisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)-8-azaspiro[4.5]decan-8- yl)acetic acid (120 mg, 152.29 µmol, 1 equiv.), 3-(6-amino-1-methyl-1H-indazol-3- yl)piperidine-2,6-dione (47.20 mg, 182.75 µmol, 1.2 equiv.), and EDCI (43.79 mg, 228.44 µmol, 1.5 equiv.) in pyridine (1.2 mL) was stirred at 25 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was used in the next step without further purification. The compound tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1h)-yl)-3-(3-(((2r)-8- (2-((3-(2,6-dioxopiperidin-3-yl)- 1-methyl-1h-indazol-6-yl)amino)-2-oxoethyl)-8-azaspiro[4.5]d ecan-2-yl)oxy)-2- methylphenyl)picolinate (100 mg, crude) was obtained as a yellow oil. MS (ESI) m/z: 1028.4[M+H] + . Step F. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-yl-carbamoyl)- 3,4dihydroisoquinolin-2(1H)-yl)-3-(3-(((2R)-8-(2-((3-(2,6-di oxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-8-azaspiro[4.5]decan-2-yl )oxy)-2- methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((2R)-8-(2-((3-(2,6-dioxo piperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)-8-azaspiro[4.5]decan-2-yl)ox y)-2-methylphenyl)picolinic acid] A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1h)- yl)-3-(3-(((2r)-8-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl- 1h-indazol-6-yl)amino)-2- oxoethyl)-8-azaspiro[4.5]decan-2-yl)oxy)-2-methylphenyl)pico linate (90 mg, 87.53 µmol, 1 equiv.) in DCM (0.8 mL) and TFA (0.8 mL) was stirred at 25 °C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-yl-carbamoyl)- 3,4dihydroisoquinolin-2(1H)-yl)-3-(3-(((2R)-8-(2-((3-(2,6-di oxopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)-8-azaspiro[4.5]decan-2-yl)ox y)-2-methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(3-(((2R)- 8-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl) amino)-2-oxoethyl)-8- azaspiro[4.5]decan-2-yl)oxy)-2-methylphenyl)picolinic acid] as a white solid. MS (ESI) m/z: 972.5[M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.90 - 10.84 (m, 1H), 9.82 (s, 1H), 8.16 (s, 1H), 8.06 - 8.01 (m, 2H), 7.78 (d, J = 8.4 Hz, 1H), 7.62 (d, J = 8.8 Hz, 2H), 7.48 - 7.42 (m, 1H), 7.35 (td, J = 8.0, 10.4 Hz, 2H), 7.24 - 7.20 (m, 1H), 7.11 - 7.05 (m, 1H), 6.94 ( J = 8.4 Hz, 1H), 6.87 - 6.83 (m, 1H), 6.64 - 6.60 (m, 1H), 5.01 - 4.95 (m, 2H), 4.86 - 4.80 (m, 1H), 4.32 (dd, J = 5.2, 10.0 Hz, 1H), 3.92 (s, 5H), 3.29 - 3.23 (m, 4H), 3.13 (s, 2H), 3.05 - 3.00 (m, 2H), 2.68 - 2.63 (m, 2H), 2.40 - 2.28 (m, 2H), 2.21 - 2.14 (m, 1H), 2.12 - 2.02 (m, 2H), 1.86 (s, 3H), 1.75 - 1.63 (m, 4H), 1.65 - 1.47 (m, 4H) Example 206. Preparation of Compound 272b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro-isoquinolin -2(1h)-yl)-3-(3-(2-((2r)-8-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1h-indazol-6-yl)amino )-2-oxoethyl)-8- azaspiro[4.5]decan-2-yl)ethoxy)-2-methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(3-(2-((2R)-8- (2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)am ino)-2-oxoethyl)-8- azaspiro[4.5]decan-2-yl)ethoxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of tert-butyl (R)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(8-(2- ethoxy-2-oxoethyl)-8- azaspiro[4.5]decan-2-yl)ethoxy)-2-methylphenyl) picolinate A mixture of ethyl (R)-2-(2-(2-(3-bromo-2-methylphenoxy)ethyl)-8-azaspiro[4.5]d ecan-8-yl) acetate (97.81 mg, 159.67 µmol, 1.4 equiv.), ethyl 2-[(3R)-3-[2-(3-bromo-2- methylphenoxy)ethyl]-8-azaspiro[4.5] decan-8-yl]acetate(50 mg, 114.05 µmol,1equiv.), tert- butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)picolinate (16.61 mg, 22.81 µmol, 0.2 equiv.), and K2CO3 (1.5 M, 114.05 µL, 1.5 equiv.) in dioxane(0.5 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 100 °C for 1 hour under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0 ~ 50% ethyl acetate / petroleum ether). The compound tert-butyl (R)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(8-(2-ethoxy-2-oxoethyl )-8-azaspiro[4.5]decan-2- yl)ethoxy)-2-methylphenyl) picolinate (95 mg, 112.5 µmol, 98.6% yield) was obtained as a colorless oil. MS (ESI) m/z: 844.4 [M+H] + Step B. Procedure for preparation of (r)-2-(2-(2-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1h)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)ethyl)-8-azaspiro[4.5]decan-8-yl)acetic acid A mixture of tert-butyl (R)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquino lin- 2(1H)-yl)-3-(3-(2-(8-(2-ethoxy-2-oxoethyl)-8-azaspiro[4.5]de can-2-yl)ethoxy)-2- methylphenyl)picolinate (95 mg, 112.55 µmol, 1 equiv.) and LiOH·H 2 O(14.17 mg, 337.65 µmol, 3 equiv.) in THF (3 mL) and H2O (1 mL) was stirred at 25 °C for 10 hours. The mixture was concentrated and the pH was adjusted to 3, then triturated and filtered to obtain a residue. The residue was used in next step without further purification. The compound (r)-2-(2-(2-(3- (6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1h)-yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-2-methylphenoxy)ethyl)-8-azaspi ro[4.5]decan-8-yl)acetic acid (80 mg, 98.0 µmol, 87.1% yield) was obtained as a white solid. MS (ESI) m/z: 816.4 [M+H] + Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((2R)-8-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-8-azaspiro[4.5]decan-2-yl )ethoxy)-2- methylphenyl)picolinate A mixture of (r)-2-(2-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1h)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)ethyl)-8- azaspiro[4.5]decan-8-yl)acetic acid (80.00 mg, 98.04 µmol, 1 equiv.), 3-(6-amino-1-methyl- 1H-indazol-3-yl)piperidine-2,6-dione (30.38 mg, 117.64 µmol, 1.2 equiv.), and EDCI (28.19 mg, 147.06 µmol, 1.5 equiv.) in pyridine (1mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 2 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was used in the next step without further purification. The compound tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((2R)- 8-(2-((3-(2,6-dioxopiperidin-3- yl)-1-methyl-1H-indazol-6-yl)amino)-2-oxoethyl)-8-azaspiro[4 .5]decan-2-yl)ethoxy)-2- methylphenyl)picolinate (80 mg, 75.7 µmol, 77.2% yield) was obtained as a colorless oil. MS (ESI) m/z: 1056.4[M+H] + Step D. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro- isoquinolin-2(1h)-yl)-3-(3-(2-((2r)-8-(2-((3-(2,6-dioxopiper idin-3-yl)-1-methyl-1h- indazol-6-yl)amino)-2-oxoethyl)-8-azaspiro[4.5]decan-2-yl)et hoxy)-2- methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((2R)-8-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-8-azaspiro[4.5]decan-2-yl )ethoxy)-2- methylphenyl)picolinic acid] A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(2-((2R)-8-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-6-yl)amino)-2- oxoethyl)-8-azaspiro[4.5]decan-2-yl)ethoxy)-2-methylphenyl)p icolinate (80 mg, 75.74 µmol, 1 equiv.) in DCM (0.8 mL) and TFA (0.8 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 25 °C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC. The compound 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro-isoquinolin -2(1h)-yl)-3-(3- (2-((2r)-8-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1h-inda zol-6-yl)amino)-2-oxoethyl)-8- azaspiro[4.5]decan-2-yl)ethoxy)-2-methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(2-((2R)- 8-(2-((3-(2,6-dioxopiperidin-3- yl)-1-methyl-1H-indazol-6-yl)amino)-2-oxoethyl)-8-azaspiro[4 .5]decan-2-yl)ethoxy)-2- methylphenyl)picolinic acid] (22.69 mg, 22.5 µmol, 29.7% yield, 99.1% purity) was obtained as a yellow solid. MS (ESI) m/z: 1000.4 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.91 - 12.81 (m, 1H), 10.88 (s, 1H), 9.89 - 9.75 (m, 1H), 8.02 ( d, J = 7.2 Hz, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.66 - 7.60 (m, 2H), 7.49 - 7.42 (m, 3H), 7.40 - 7.32 (m, 2H), 7.24 - 7.17 (m, 1H), 7.09 (t, J = 8.0 Hz, 1H), 6.97 (d, J = 9.2 Hz, 1H), 6.89 (d, J = 8.0 Hz, 1H), 6.63 (d, J = 8.0 Hz, 1H), 4.98 (s, 2H), 4.32 (dd, J = 5.2, 9.6 Hz, 1H), 3.99 - 3.89 (m, 7H), 3.30 - 3.28 (m, 4H), 3.05 - 3.00 (m, 2H), 2.67 - 2.58 (m, 4H), 2.37 - 2.31 (m, 2H), 1.91 - 1.76 (m, 7H), 1.42 - 1.42 (m, 1H), 1.64 - 1.42 (m, 5H), 1.35 - 1.19 (m, 1H), 1.19 - 1.18 (m, 1H), 1.10 - 1.03 (m, 1H) Example 207. Preparation of Compound 281a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1R,4r)-4- ((3S)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 6-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1R,4r)-4- ((3S*)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of ((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)methanol To a mixture of methyl (1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexane-1-carboxylate (40 g, 48.90 mmol, 40% purity, 1 equiv.) in THF (350 mL) was slowly added LAH (1.95 g, 51.34 mmol, 1.05 equiv.) at 0 °C and then the mixture was stirred at 25 ℃ for 2 hours. The mixture was quenched by Na2SO4.10 H2O (3 g). The solution was slowly poured into ice-water (300 mL) and the pH was acidized to pH 4~5 with 1 M HCl, extracted with EtOAc 900 mL (300 mL × 3), washed with brine (400 mL), dried by sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (eluent of 0~12% ethyl acetate / petroleum ether) to give ((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)methanol (20.5 g, 67.0 mmol, 45.6% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ = 7.15 - 7.11 (m, 1H), 7.06 (t, J = 8.0 Hz, 1H), 7.03 - 6.99 (m, 1H), 4.26 - 4.17 (m, 1H), 2.21 (s, 3H), 2.06 (dd, J = 2.4, 11.6 Hz, 2H), 1.98 (s, 1H), 1.78 (d, J = 11.6 Hz, 2H), 1.58 (d, J = 4.8 Hz, 2H), 1.40 - 1.34 (m, 5H) Step B. Procedure for preparation of (1s,4s)-4-(3-bromo-2-methylphenoxy)cyclohexane- 1-carbaldehyde To a mixture of (COCl) 2 (8.48 g, 66.84 mmol, 5.85 mL, 2 equiv.) in DCM (80 mL) at -78°C was added DMSO (10.45 g, 133.69 mmol, 10.45 mL, 4 equiv.) and the mixture was stirred for 0.5 hours. To the solution was added ((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)methanol (10 g, 33.42 mmol, 1 equiv.) in DCM (200 mL) and the mixture was stirred at-78 °C for 1 hour. Then to the mixture was added TEA (20.29 g, 200.53 mmol, 27.91 mL, 6 equiv.) and the mixture was warmed to 25 °C and stirred for 0.5 hour under N 2 atmosphere. The reaction mixture was diluted with H 2 O (150 mL) and extracted with DCM 300 mL (100 mL × 3). The combined organic layers were washed with saturated NaCl (200 mL), filtered and concentrated under reduced pressure to give (1s,4s)-4-(3-bromo-2- methylphenoxy)cyclohexane-1-carbaldehyde (33.2 g, crude) as a white solid. MS (ESI) m/z: 296.9 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 9.61 (s, 1H), 7.16 - 7.13 (m, 1H), 7.10 - 7.05 (m, 1H), 7.04 - 7.01 (m, 1H), 4.34 - 4.27 (m, 1H), 2.39 - 2.33 (m, 1H), 2.25 - 2.20 (m, 3H), 2.03 - 1.93 (m, 4H), 1.60 - 1.55 (m, 2H), 1.46 (d, J = 9.6 Hz, 2H) Step C. Procedure for preparation of ethyl (E)-3-((1s,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)acrylate A mixture of NaH (4.95 g, 123.83 mmol, 60% purity, 1.6 equiv.) in THF (200 mL) was purged with N2 three times and ethyl 2-(diethoxyphosphoryl)acetate (24.29 g, 108.35 mmol, 21.50 mL, 1.4 equiv.) was slowly added at 0 °C, and the mixture was stirred for 1.5 hours under N2 atmosphere. To the solution was added (1s,4s)-4-(3-bromo-2-methylphenoxy)cyclohexane-1- carbaldehyde (23 g, 77.39 mmol, 1 equiv.) at 0 °C, the mixture was warmed to 25 °C, and stirred for 1.5 hours under N2 atmosphere. The reaction mixture was quenched by addition of saturated NH4Cl (150 mL) under 0°C. The mixture was extracted with DCM 300 mL (100 mL × 3). The combined organic layers were washed with brine 200 mL (100 mL × 2), dried over sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~13% ethyl acetate / petroleum ether) to give ethyl (E)-3-((1s,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)acrylat e (11.3 g, 30.7 mmol, 39.7% yield) as a colorless oil. Step D. Procedure for preparation of ethyl 3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propanoate A mixture of ethyl (E)-3-((1s,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)acrylat e (11 g, 29.95 mmol, 1 equiv.) and PtO2 (3.40 g, 14.98 mmol, 0.5 equiv.) in EtOH (200 mL) was degassed and purged with H 2 three times, and then the mixture was stirred at 20 °C for 23 hours under H2 atmosphere. The mixture was filtered by EtOH (50 mL) and concentrated under reduced pressure to give ethyl 3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propanoate (9.1 g, 20.1 mmol, 67.3% yield, 81.8% purity) as a gray oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.15 - 7.11 (m, 1H), 7.06 (t, J = 8.0 Hz, 1H), 7.02 - 6.99 (m, 1H), 4.26 - 4.17 (m, 1H), 4.05 (q, J = 7.2 Hz, 2H), 2.29 (t, J = 7.6 Hz, 2H), 2.20 (s, 3H), 2.03 (dd, J = 2.8, 12.4 Hz, 2H), 1.78 - 1.71 (m, 2H), 1.46 (q, J = 7.2 Hz, 2H), 1.38 - 1.29 (m, 2H), 1.27 - 1.20 (m, 1H), 1.17 (t, J = 7.2 Hz, 3H), 1.10 - 0.99 (m, 2H) Step E. Procedure for preparation of 3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propanoic acid A mixture of ethyl 3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propanoate (3 g, 8.12 mmol, 1 equiv.) and LiOH•H 2 O (1 M, 8.12 mL, 1 equiv.) in THF (30 mL) was stirred at 25 °C for 18 hours. The mixture was concentrated and diluted with H2O (20 mL), then the pH of the mixture was adjusted to 3 with 1 M HCl. Then the mixture was filtered and concentrated to give 3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propanoic acid (2.5 g, 6.8 mmol, 83.6% yield, 92.4% purity) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 12.27 - 11.77 (m, 1H), 7.15 - 7.11 (m, 1H), 7.06 (t, J = 8.0 Hz, 1H), 7.02 - 6.99 (m, 1H), 4.26 - 4.17 (m, 1H), 2.25 - 2.21 (m, 2H), 2.20 (s, 3H), 2.07 - 2.00 (m, 2H), 1.75 (d, J = 11.6 Hz, 2H), 1.43 (q, J = 7.2 Hz, 2H), 1.38 - 1.30 (m, 2H), 1.25 (td, J = 3.6, 7.2 Hz, 1H), 1.09 - 0.98 (m, 2H) Step F. Procedure for preparation of 3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-N-methoxy-N-methylpropanamide A mixture of 3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propanoic acid (2.4 g, 7.03 mmol, 1 equiv.) and N,O-dimethylhydroxylamine (1.29 g, 21.10 mmol, 3 equiv.) in THF (24 mL) was added HATU (8.02 g, 21.10 mmol, 3 equiv.) and TEA (2.14 g, 21.10 mmol, 2.94 mL, 3 equiv.) and stirred at 25 °C for 18 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~26% ethyl acetate / petroleum ether) to give 3-((1r,4r)-4-(3- bromo-2-methylphenoxy)cyclohexyl)-N-methoxy-N-methylpropanam ide (2.5 g, 6.4 mmol, 91.8% yield, 99.2% purity) as a colorless oil. MS (ESI) m/z: 384.0 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 7.15 - 7.12 (m, 1H), 7.06 (t, J = 8.0 Hz, 1H), 7.03 - 7.00 (m, 1H), 4.22 (t, J = 4.0 Hz, 1H), 3.65 (s, 3H), 3.07 (s, 3H), 2.38 (t, J = 7.6 Hz, 2H), 2.20 (s, 3H), 2.08 - 2.00 (m, 2H), 1.77 (d, J = 11.6 Hz, 2H), 1.46 - 1.39 (m, 2H), 1.39 - 1.30 (m, 2H), 1.30 - 1.25 (m, 1H), 1.11 - 0.99 (m, 2H) Step G. Procedure for preparation of 4-((1r,4r)-4-(3-bromophenoxy)cyclohexyl)butan-2- one To a solution of 3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-N-methoxy- N- methylpropanamide (2.3 g, 5.98 mmol, 1 equiv.) in THF (23 mL) was added bromo(methyl)magnesium (3 M, 3.99 mL, 2 equiv.) at -78 °C. The mixture was stirred at 25 °C for 18 hours. The reaction mixture was quenched by addition of saturated NH4Cl (25 mL) at 0°C. The mixture was extracted with DCM 90 mL (30 mL × 3). The combined organic layers were washed with brine 60 mL (30 mL × 2), dried over sodium sulfate, filtered, and concentrated under reduced pressure to give 4-((1r,4r)-4-(3-bromophenoxy)cyclohexyl)butan- 2-one (2.05 g, crude) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.16 - 7.12 (m, 1H), 7.06 (t, J = 8.0 Hz, 1H), 7.03 - 7.00 (m, 1H), 4.27 - 4.17 (m, 1H), 2.44 (t, J = 7.6 Hz, 2H), 2.20 (s, 3H), 2.08 (s, 3H), 2.03 (d, J = 9.6 Hz, 2H), 1.77 - 1.71 (m, 2H), 1.41 - 1.36 (m, 2H), 1.35 - 1.27 (m, 2H), 1.22 - 1.15 (m, 1H), 1.09 - 0.99 (m, 2H) Step H. Procedure for preparation of 4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butan-2-ol To a mixture of 4-((1r,4r)-4-(3-bromophenoxy)cyclohexyl)butan-2-one (1.97 g, 5.81 mmol, 1 equiv.) in THF (20 mL) was slowly added LAH (220.50 mg, 5.81 mmol, 1 equiv.) at 0 °C, and then the mixture was stirred at 25 °C for 2 hours. The mixture was quenched by 3 g Na2SO4.10 H2O. The solution was slowly poured into ice-water (50 mL) and the pH was acidized to pH 4-5 with 1 M HCl, extracted with EtOAc 150 mL (50 mL × 3), washed with brine 80 mL, dried by sodium sulfate, filtered and concentrated under reduced pressure to give 4-((1r,4s)-4-(3- bromo-2-methylphenoxy)cyclohexyl)butan-2-ol (1.7 g, crude) as a colorless oil. Step I. Procedure for preparation of (R)-4-((1r,4S)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butan-2-ol. 4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butan-2-ol was purified by prep-HPLC to give (S)-4-((1r,4R)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butan-2 -ol and (R)-4-((1r,4S)-4- (3-bromo-2-methylphenoxy)cyclohexyl)butan-2-ol. 1 H NMR (400 MHz, DMSO-d6) δ = 7.16 - 7.11 (m, 1H), 7.06 (t, J = 8.0 Hz, 1H), 7.03 - 6.99 (m, 1H), 4.33 (s, 1H), 4.26 - 4.18 (m, 1H), 3.56 - 3.50 (m, 1H), 2.20 (s, 3H), 2.03 (d, J = 10.8 Hz, 2H), 1.75 (d, J = 12.4 Hz, 2H), 1.41 - 1.25 (m, 6H), 1.21 - 1.16 (m, 1H), 1.08 - 0.98 (m, 5H) 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.14 - 7.11 (m, 1H), 7.06 (t, J = 8.0 Hz, 1H), 7.02 - 6.98 (m, 1H), 4.35 (d, J = 4.8 Hz, 1H), 4.26 - 4.16 (m, 1H), 3.53 (td, J = 5.5, 11.1 Hz, 1H), 2.20 (s, 3H), 2.03 (d, J = 10.0 Hz, 2H), 1.75 (d, J = 12.4 Hz, 2H), 1.39 - 1.23 (m, 6H), 1.19 - 1.15 (m, 1H), 1.06 - 0.98 (m, 5H) Step J. Procedure for preparation of (R)-4-((1r,4S)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butan-2-yl 4-methylbenzenesulfonate To a mixture of (R)-4-((1r,4S)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butan-2 -ol (360 mg, 1.05 mmol, 1 equiv.), TEA (320.22 mg, 3.16 mmol, 440.46 µL, 3 equiv.), and DMAP (19.33 mg, 158.23 µmol, 0.15 equiv.) in DCM (5 mL) was added TsCl (301.65 mg, 1.58 mmol, 1.5 equiv.) at 0°C, and then the mixture was stirred at 40 °C for 3 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~17% ethyl acetate/petroleum ether) to give (R)-4-((1r,4S)-4- (3-bromo-2-methylphenoxy)cyclohexyl)butan-2-yl 4-methylbenzenesulfonate (488 mg, 984.9 µmol, 93.3% yield) as a yellow solid. Step K. Procedure for preparation of 3-(6-(4-((S)-4-((1r,4R)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butan-2-yl)piperazin-1-yl)-1-methyl -1H-indazol-3- yl)piperidine-2,6-dione To a solution of (R)-4-((1r,4S)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butan-2 -yl 4- methylbenzenesulfonate (200 mg, 403.66 µmol, 1 equiv.) in ACN (4 mL) was added K2CO3 (167.36 mg, 1.21 mmol, 3 equiv.), KI (67.01 mg, 403.66 µmol, 1 equiv.), and 3-(1-methyl-6- piperazin-1-yl-indazol-3-yl)piperidine-2,6-dione (161.55 mg, 444.02 µmol, 1.1 equiv., HCl). The mixture was stirred at 60 °C for 10 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether : ethyl acetate = 10/1 to 0/1) to give 3-(6-(4-((S)-4-((1r,4R)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butan-2-yl)piperazin-1-yl)-1-methyl -1H-indazol-3-yl)piperidine- 2,6-dione (120 mg, 184.4 µmol, 45.6% yield) as a yellow oil. MS (ESI) m/z: 682.4 [M+H] + . Step L. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-((3S)-3-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinate A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)picolinat e (100 mg, 163.25 µmol, 1 eq), 3- (6-(4-((S)-4-((1r,4R)-4-(3-bromo-2-methylphenoxy)cyclohexyl) butan-2-yl)piperazin-1-yl)-1- methyl-1H-indazol-3-yl)piperidine-2,6-dione (106.22 mg, 163.25 µmol, 1 equiv.), [2-(2- aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-butyl-phos phane;methanesulfonate (11.89 mg, 16.33 µmol, 0.1 equiv.), and KF (28.46 mg, 489.76 µmol, 11.47 µL, 3 equiv.) in dioxane (4 mL) and H 2 O (1 mL) was degassed and purged with N 2 three times and then the mixture was stirred at 100 °C for 1 hour under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , dichloromethane : methanol = 100/1 to 30/1) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1R,4r)-4-((3S)-3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (110 mg, 104.1 µmol, 63.7% yield) as a yellow solid. MS (ESI) m/z: 1056.5 [M+H] + . Step M. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-((3S)-3-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3- (((1R,4r)-4-((3S*)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-6-yl)piperazin- 1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1R,4r)-4-((3S)-3-(4-(3-(2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-6- yl)piperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picol inate (100.09 mg, 94.76 µmol, 1 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 142.54 equiv.). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1R,4r)-4-((3S)-3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r) -4-((3S*)-3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin-1-yl )butyl)cyclohexyl)oxy)-2- methylphenyl)picolinic acid] (16.5 mg, 15.1 µmol, 16.0% yield, 95.6% purity) as a yellow solid. MS (ESI) m/z: 1000.4 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.85 (s, 1H), 8.15 (s, 1H), 8.04 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 7.2 Hz, 1H), 7.52 - 7.43 (m, 4H), 7.41 - 7.32 (m, 2H), 7.10 - 7.04 (m, 1H), 6.98 - 6.90 (m, 3H), 6.84 (s, 1H), 6.62 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.28 - 4.24 (m, 1H), 4.20 ( t, J = 10.8 Hz, 1H), 3.92 ( s, 2H), 3.89 (s, 3H), 3.21 (s, 4H), 3.03 (t, J = 5.2 Hz, 2H), 2.68 (s, 2H), 2.63 - 2.57 (m, 5H), 2.32 - 2.25 (m, 1H), 2.20 - 2.13 (m, 1H), 2.11 - 2.05 (m, 2H), 1.87 (s, 3H), 1.83 - 1.77 (m, 2H), 1.58 - 1.50 (m, 1H), 1.40 - 1.33 (m, 2H), 1.31 - 1.22 (m, 4H), 1.12 - 1.04 (m, 2H), 0.98 ( d, J = 6.4 Hz, 3H) Example 208. Preparation of Compound 281b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1S,4r)-4- ((3R)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 6-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1S,4r)-4- ((3R*)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of [(1S)-3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]- 1-methyl-propyl] 4-methylbenzenesulfonate To a solution of (2S)-4-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]butan-2-ol (300 mg, 879.04 μmol, 1 equiv.) in DCM (3 mL) was added 4-methylbenzenesulfonyl chloride (184.34 mg, 966.94 μmol, 1.1 equiv.), TEA (444.75 mg, 4.40 mmol, 611.76 μL, 5 equiv.), and DMAP (10.74 mg, 87.90 μmol, 0.1 equiv.) at 25 °C. The reaction mixture was stirred at 60 °C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0 ~ 30% ethyl acetate/petroleum ether). The compound [(1S)-3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]- 1-methyl-propyl] 4-methylbenzenesulfonate (167 mg, 322.2 μmol, 36.6% yield, 95.6% purity) was obtained as a yellow oil. MS (ESI) m/z: 517.1 [M + Na] + . 1 H NMR (400 MHz, CDCl 3 -d) δ = 7.81 (d, J = 8.4 Hz, 2H), 7.35 (d, J = 8.0 Hz, 2H), 7.14 (d, J = 8.0 Hz, 1H), 6.97 (t, J = 8.0 Hz, 1H), 6.79 (d, J = 8.0 Hz, 1H), 4.65 - 4.56 (m, 1H), 4.10 - 3.99 (m, 1H), 2.46 (s, 3H), 2.29 (s, 3H), 2.09 (dd, J = 2.8, 12.4 Hz, 2H), 1.78 - 1.67 (m, 2H), 1.66 - 1.60 (m, 1H), 1.55 - 1.49 (m, 1H), 1.45 - 1.33 (m, 2H), 1.26 (d, J = 6.3 Hz, 3H), 1.22 - 1.10 (m, 3H), 1.00 - 0.87 (m, 2H) Step B. Procedure for preparation of 3-(6-(4-((R)-4-((1r,4S)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butan-2-yl)piperazin-1-yl)-1-methyl -1H-indazol-3- yl)piperidine-2,6-dione To a solution of [(1S)-3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-1-methyl-pr opyl] 4- methylbenzenesulfonate (80 mg, 161.46 μmol, 1 equiv.) in CH 3 CN (1 mL) was added 3-(1- methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dione (63.43 mg, 193.76 μmol, 1.2 equiv.), K2CO3 (66.95 mg, 484.39 μmol, 3 equiv.), and KI (26.80 mg, 161.46 μmol, 1 equiv.) at 25 °C. The reaction was stirred at 60 °C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~10% ethyl acetate/petroleum ether). The compound 3-(6-(4- ((R)-4-((1r,4S)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butan- 2-yl)piperazin-1-yl)-1- methyl-1H-indazol-3-yl)piperidine-2,6-dione (36 mg, 51.8 μmol, 32.1% yield, 93.7% purity) was obtained as a yellow oil. Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-((4-((3R)-3-(4-(3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy)-2-methy lphenyl)picolinate tert-Butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-2-carb oxylate (37.28 mg, 60.86 μmol, 1.1 equiv.), 3-(6-(4-((R)-4-((1r,4S)-4-(3-bromo-2-methylphenoxy)cyclohexy l)butan-2- yl)piperazin-1-yl)-1-methyl-1H-indazol-3-yl)piperidine-2,6-d ione (36 mg, 55.33 μmol, 1 equiv.), KF (9.64 mg, 165.99 μmol, 3.89 μL, 3 equiv.), and [2-(2- aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-butyl-phos phane;methanesulfonate (8.06 mg, 11.07 μmol, 0.2 equiv.) were taken up into a microwave tube in dioxane (2 mL) and H2O (0.2 mL). The sealed tube was heated at 100 °C for 1 hour under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0 ~ 50% ethyl acetate/petroleum ether). The compound tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-((4-((3R)-3-(4-(3-(2,6-dio xopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylph enyl)picolinate (69 mg, 29.4 μmol, 53.1% yield, 45% purity) was obtained as a yellow solid. MS (ESI) m/z: 1156.4 [M+H] + . Step D. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-((3R)-3-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3- (((1S,4r)-4-((3R*)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-6-yl)piperazin- 1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-((4-((3R)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-6-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (69 mg, 29.42 μmol, 45% purity, 1 equiv.) in DCM (0.5 mL) and TFA (0.5 mL) was stirred at 25 °C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC. The compound 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-((3R)-3-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1S,4r)-4- ((3R*)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] (13.0 mg, 12.2 μmol, 41.5% yield, 93.6% purity) was obtained as a yellow solid. MS (ESI) m/z: 1000.4 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.95 - 12.39 (m, 2H), 10.85 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.4 Hz, 1H), 7.65 - 7.60 (m, 1H), 7.55 (d, J = 8.4 Hz, 1H), 7.49 - 7.42 (m, 3H), 7.40 - 7.32 (m, 2H), 7.08 (t, J = 8.0 Hz, 1H), 7.00 - 6.90 (m, 4H), 6.62 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.30 - 4.17 (m, 2H), 3.91 (s, 6H), 3.03 (t, J = 5.6 Hz, 2H), 2.69 - 2.56 (m, 5H), 2.36 - 2.26 (m, 2H), 2.22 - 2.04 (m, 4H), 1.87 (s, 3H), 1.85 - 1.78 (m, 2H), 1.59 - 1.00 (m, 14H) Example 209. Preparation of Compound 283a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1R,4r)-4- ((3S)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 7-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1R,4r)-4- ((3S*)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -7-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of 3-(7-(4-((S)-4-((1r,4R)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butan-2-yl)piperazin-1-yl)-1-methyl -1H-indazol-3- yl)piperidine-2,6-dione To a solution of [(1R)-3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-1-methyl-pr opyl] 4- methylbenzenesulfonate (200 mg, 403.66 µmol, 1 equiv.) in CH 3 CN (4 mL) was added K 2 CO 3 (167.36 mg, 1.21 mmol, 3 equiv.), KI (67.01 mg, 403.66 µmol, 1 equiv.), and 3-(1-methyl-7- piperazin-1-yl-indazol-3-yl)piperidine-2,6-dione (161.55 mg, 444.03 µmol, 1.1 equiv., HCl). The mixture was stirred at 60 °C for 10 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether : ethyl acetate = 10/1 to 0/1) to give 3-(7-(4-((S)-4-((1r,4R)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butan-2-yl)piperazin-1-yl)-1-methyl -1H-indazol-3-yl)piperidine- 2,6-dione (120 mg, 184.4 µmol, 45.6% yield) was obtained as a yellow oil. MS (ESI) m/z: 650.3 [M+H] + . Step B. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[(3S)-3-[4-[3-(2,6- dioxo-3-piperidyl)-1-methyl- indazol-7-yl]piperazin-1-yl]butyl]cyclohexoxy]-2-methyl-phen yl]pyridine-2-carboxylate A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)picolinat e (110 mg, 179.58 µmol, 1 equiv.), 3-(7-(4-((S)-4-((1r,4R)-4-(3-bromo-2-methylphenoxy)cyclohexy l)butan-2-yl)piperazin-1-yl)- 1-methyl-1H-indazol-3-yl)piperidine-2,6-dione (116.84 mg, 179.58 µmol, 1 equiv.), [2-(2- aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-butyl-phos phane;methanesulfonate (13.08 mg, 17.96 µmol, 0.1 equiv.), and KF (31.30 mg, 538.74 µmol, 12.62 µL, 3 equiv.) in dioxane (4 mL) and H 2 O (1 mL) was degassed and purged with N 2 three times and then the mixture was stirred at 100 °C for 1 hour under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, dichloromethane : methanol = 100/1 to 30/1) to give tert-butyl 6-[8- (1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin- 2-yl]-3-[3-[4-[(3S)-3-[4-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]piperazin-1-yl ]butyl]cyclohexoxy]-2-methyl- phenyl]pyridine-2-carboxylate (100 mg, 94.6 µmol, 52.7% yield) as a yellow solid. MS (ESI) m/z: 1056.5 [M+H] + . Step C. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-((3S)-3-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3- (((1R,4r)-4-((3S*)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-7-yl)piperazin- 1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[4-[(3S)-3-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl -indazol-7-yl]piperazin-1- yl]butyl]cyclohexoxy]-2-methyl-phenyl]pyridine-2-carboxylate (100 mg, 94.67 µmol, 1 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 142.67 equiv.) and stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-((3S)-3-(4 -(3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1R,4r)-4- ((3S*)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -7-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] (58.3 mg, 55.0 µmol, 58.1% yield, 98.7% purity) as a off-white solid. MS (ESI) m/z: 1000.4 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.05 - 12.61 (m, 1H), 10.89 (s, 1H), 8.14 (s, 1H), 8.04 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 7.2 Hz, 1H), 7.50 - 7.43 (m, 3H), 7.42 - 7.33 (m, 3H), 7.11 - 7.00 (m, 3H), 6.98 - 6.90 (m, 2H), 6.62 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.37 - 4.32 (m, 1H), 4.25 (s, 3H), 4.21 (s, 1H), 3.92 (t, J = 5.2 Hz, 2H), 3.27 - 3.23 (m, 4H), 3.03 (t, J = 5.2 Hz, 2H), 2.95 - 2.82 (m, 4H), 2.66 - 2.54 (m, 3H), 2.34 - 2.27 (m, 1H), 2.20 - 2.14 (m, 1H), 2.09 (d, J = 9.6 Hz, 2H), 1.88 (s, 3H), 1.85 - 1.79 (m, 2H), 1.68 - 1.58 (m, 1H), 1.36 (d, J = 2.4 Hz, 2H), 1.33 - 1.22 (m, 4H), 1.17 - 1.08 (m, 2H), 1.07 (s, 3H) Example 210. Preparation of Compound 284a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1S,4r)-4- ((2S)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 7-yl)piperazin-1-yl)-2- methylpropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1S,4r)-4- ((2S*)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -7-yl)piperazin-1-yl)-2- methylpropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of (S)-3-((1r,4S)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2-methylpropanal To a solution of oxalyl dichloride (74.38 mg, 586.03 µmol, 51.30 µL, 2 equiv.) in DCM (5 mL) was added dropwise to a solution of DMSO (91.58 mg, 1.17 mmol, 91.58 µL, 4 equiv.) in DCM (2 mL) at -70 °C under N2 atmosphere. The mixture was stirred at -70 °C for 1 hour. After which time (2S)-3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-2-methyl-pro pan-1-ol (100 mg, 293.01 µmol, 1 equiv.) in DCM (2 mL) was added dropwise at -70 °C. The solution was stirred for 1 hour at -70 °C. Then TEA (177.90 mg, 1.76 mmol, 244.70 µL, 6 equiv.) was added into the solution. The solution was stirred at -70 °C for 0.5 hour under N2 atmosphere. Water (30 mL) was then added and extracted with EtOAc 20 mL (10 mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated under vacuum to give (S)-3-((1r,4S)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2-methylpropanal (80 mg, crude) as a yellow oil. Step B. Procedure for preparation of 3-(7-(4-((S)-3-((1r,4S)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2-methylpropyl)piperazin-1-yl)-1-m ethyl-1H-indazol-3- yl)piperidine-2,6-dione To a solution of 3-(1-methyl-7-(piperazin-1-yl)-1H-indazol-3-yl)piperidine-2, 6-dione (102.95 mg, 282.96 µmol, 1.2 equiv., HCl) in DCM (1 mL) was added NaBH(OAc)3 (149.93 mg, 707.41 µmol, 3 equiv.), then (S)-3-((1r,4S)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2- methylpropanal (80 mg, 235.80 µmol, 1 equiv.) was added into the mixture and stirred at 0 °C for 1 hour. Then the mixture was stirred at 25 °C for 15 hours. The mixture was filtered and concentrated under vacuum to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate = 4/1 to DCM:MeOH = 10:1) to give 3- (7-(4-((S)-3-((1r,4S)-4-(3-bromo-2-methylphenoxy)cyclohexyl) -2-methylpropyl)piperazin-1- yl)-1-methyl-1H-indazol-3-yl)piperidine-2,6-dione (40 mg, 61.4 µmol, 26.0% yield) as a yellow oil. MS (ESI) m/z: 650.2 [M+H] + . Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-((2S)-3-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)-2-methylpropyl)cycloh exyl)oxy)-2- methylphenyl)picolinate A mixture of 3-(7-(4-((S)-3-((1r,4S)-4-(3-bromo-2-methylphenoxy)cyclohexy l)-2- methylpropyl)piperazin-1-yl)-1-methyl-1H-indazol-3-yl)piperi dine-2,6-dione (40 mg, 61.48 µmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pic olinate (45.19 mg, 73.77 µmol, 1.2 equiv.), KF (10.71 mg, 184.43 µmol, 4.32 µL, 3 equiv.), H 2 O (0.1 mL), and [2-(2- aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-butyl-phos phane;methanesulfonate (4.48 mg, 6.15 µmol, 0.1 equiv.) in dioxane (1 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 100 °C for 1 hour under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 3/1, DCM: MeOH = 10:1) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- (3-(((1S,4r)-4-((2S)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-met hyl-1H-indazol-7-yl)piperazin-1- yl)-2-methylpropyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (30 mg, 28.4 µmol, 46.2% yield) as a yellow solid. MS (ESI) m/z: 1056.4 [M+H] + . Step D. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-((2S)-3-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)-2-methylpropyl)cycloh exyl)oxy)-2- methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-((2S*)-3-(4-(3 -(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)-2-methylpropyl)cycloh exyl)oxy)-2- methylphenyl)picolinic acid] To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1S,4r)-4-((2S)-3-(4-(3-(2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-7- yl)piperazin-1-yl)-2-methylpropyl)cyclohexyl)oxy)-2-methylph enyl)picolinate (30 mg, 28.40 µmol, 1 equiv.) in DCM (1 mL) was added TFA (770.00 mg, 6.75 mmol, 0.5 mL, 237.78 equiv.). The mixture was stirred at 25 °C for 8 hours. The reaction mixture was concentrated under reduced pressure to remove solvent and to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)- 3-(3-(((1S,4r)-4-((2S)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-m ethyl-1H-indazol-7-yl)piperazin- 1-yl)-2-methylpropyl)cyclohexyl)oxy)-2-methylphenyl)picolini c acid [6-(8-(benzo[d]thiazol- 2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4 r)-4-((2S*)-3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin-1-yl )-2- methylpropyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] (15.4 mg, 14.4 µmol, 50.7% yield, 97.6% purity) as a yellow solid. MS (ESI) m/z: 1000.5 [M+H] + 1 H NMR (400 MHz, DMSO) δ = 10.87 (s, 1H), 8.22 (s, 1H), 8.01 (d, J = 8.0 Hz, 1H), 7.80 - 7.74 (m, 1H), 7.63 (d, J = 6.8 Hz, 1H), 7.48 - 7.32 (m, 6H), 7.07 - 6.98 (m, 3H), 6.94 - 6.85 (m, 2H), 6.64 (d, J = 7.6 Hz, 1H), 5.75 (s, 1H), 5.01 - 4.94 (m, 2H), 4.36 - 4.31 (m, 1H), 4.24 (s, 3H), 4.22 - 4.15 (m, 1H), 3.93 - 3.88 (m, 2H), 3.02 (t, J = 5.6 Hz, 2H), 2.64 - 2.54 (m, 8H), 2.21 - 2.04 (m, 6H), 1.88 (s, 3H), 1.84 - 1.72 (m, 3H), 1.49 - 1.24 (m, 5H), 1.14 - 1.07 (m, 1H), 1.03 - 0.96 (m, 2H), 0.91 - 0.85 (m, 3H) Example 211. Preparation of Compound 283b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1S,4r)-4- ((3R)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 7-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1S,4r)-4- ((3R*)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -7-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of 3-[7-[4-[(1R)-3-[4-(3-bromo-2-methyl-phenoxy) cyclohexyl]-1-methyl-propyl]piperazin-1-yl]-1-methyl-indazol -3-yl]piperidine-2, 6-dione To a solution of [(1S)-3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-1-methyl-pr opyl]4- methylbenzenesulfonate (80 mg, 161.46 μmol, 1 equiv.) in CH 3 CN (1 mL) was added 3-(1- methyl-7-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dione (63.43 mg, 193.75 μmol, 1.2 equiv.), K2CO3 (66.95 mg, 484.38 μmol, 3 equiv.), and KI (26.80 mg, 161.46 μmol, 1 equiv.) at 25 °C. The reaction was stirred at 60 °C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~10% ethyl acetate/petroleum ether). The compound 3-[7-[4- [(1R)-3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-1-methyl-pr opyl]piperazin-1-yl]-1- methyl-indazol-3-yl]piperidine-2,6-dione (68 mg, 101.1 μmol, 62.6% yield, 96.8% purity) was obtained as a yellow oil. MS (ESI) m/z: 650.2 [M+H] + . Step B. Procedure for preparation of tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[(3R)-3-[4-[3-(2,6-di oxo-3-piperidyl)-1-methyl- indazol-7-yl]piperazin-1-yl]butyl]cyclohexoxy]-2-methyl-phen yl]pyridine-2-carboxylate tert-Butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-2-carb oxylate (70.42 mg, 114.96 μmol, 1.1 equiv.), 3-[7-[4-[(1R)-3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-1-m ethyl- propyl]piperazin-1-yl]-1-methyl-indazol-3-yl]piperidine-2,6- dione (68 mg, 104.51 μmol, 1 equiv.), KF (18.22 mg, 313.53 μmol, 7.35 μL, 3 equiv.), and [2-(2- aminophenyl)phenyl]palladium (1+);bis(1-adamantyl)-butyl-phosphane;methanesulfonate (15.22 mg, 20.90 μmol, 0.2 equiv.) were taken up into a microwave tube in dioxane (2 mL) and H2O (0.2 mL). The sealed tube was heated at 100 °C for 60 min under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0 ~ 30% ethyl acetate/petroleum ether). The compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[4-[(3R)-3-[4-[3-(2,6-dioxo-3-piperid yl)-1-methyl-indazol-7- yl]piperazin-1-yl]butyl]cyclohexoxy]-2-methyl-phenyl]pyridin e-2-carboxylate (100 mg, 53.9 μmol, 51.6% yield, 57% purity) was obtained as a yellow solid. MS (ESI) m/z: 1056.4 [M+H] + . Step C. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-((3R)-3-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3- (((1S,4r)-4-((3R*)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-7-yl)piperazin- 1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[4-[(3R)-3-[4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-i ndazol-7-yl]piperazin-1- yl]butyl]cyclohexoxy]-2-methyl-phenyl]pyridine-2-carboxylate (100 mg, 53.96 μmol, 57% purity, 1 equiv.) in DCM (0.5 mL) and TFA (0.5 mL) was stirred at 25 °C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give compound 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-((3R)-3-(4 -(3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1S,4r)-4- ((3R*)-3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -7-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] (17.5 mg, 15.9 μmol, 29.5% yield, 90.7% purity) was obtained as an off-white solid. MS (ESI) m/z: 1000.4 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.89 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.50 - 7.43 (m, 4H), 7.40 - 7.32 (m, 2H), 7.10 - 7.02 (m, 3H), 6.94 (dd, J = 8.4, 15.6 Hz, 2H), 6.62 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.35 (dd, J = 5.2, 9.2 Hz, 1H), 4.30 - 4.19 (m, 4H), 3.91 (t, J = 5.2 Hz, 2H), 3.02 (t, J = 5.2 Hz, 3H), 2.75 - 2.52 (m, 5H), 2.44 - 2.23 (m, 2H), 2.23 - 2.04 (m, 4H), 1.87 (s, 3H), 1.83 (s, 2H), 1.65 - 0.86 (m, 15H) Example 212. Preparation of Compound 278a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1S,4r)-4-(3- ((3R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)-3-methylpiperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of methyl 4-(3-bromo-2-methyl- phenoxy)cyclohexanecarboxylate To a solution of methyl 4-hydroxycyclohexanecarboxylate (16 g, 101.14 mmol, 1.0 equiv.) and 3-bromo-2-methyl-phenol (20.81 g, 111.26 mmol, 1.1 equiv.) in toluene (500 mL) was added 2-(tributyl-λ5-phosphanylidene)acetonitrile (29.29 g, 121.37 mmol, 1.2 equiv.). The mixture was stirred at 120 °C for 12 hours. The reaction mixture was then concentrated under reduced pressure to give the compound methyl 4-(3-bromo-2-methyl-phenoxy)cyclohexanecarboxylate (125 g, crude) was obtained as a black oil, which was used in the next step without further purification. Step B. Procedure for preparation of [4-(3-bromo-2-methyl- phenoxy)cyclohexyl]methanol A mixture of methyl 4-(3-bromo-2-methyl-phenoxy)cyclohexanecarboxylate (28 g, 34.23 mmol, 40% purity, 1.0 equiv.) in THF (300 mL) was slowly added LAH (1.36 g, 35.94 mmol, 1.05 equiv.) at 0 °C. The mixture was then stirred at 0 ℃ for 1 hour. The mixture was quenched by 2.5 g Na 2 SO 4 .10 H 2 O. The solution was poured into ice-water (250 mL) slowly and the pH was adjusted to pH 4-5 with 1 M HCl, extracted with EtOAc 600 mL (200 mL×3), washed with brine 300 mL (150 mL×2), dried over by sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (eluent of 0~18% ethyl acetate/petroleum ether). The compound [4-(3-bromo-2-methyl- phenoxy)cyclohexyl]methanol (10 g, 32.3 mmol, 47.2% yield, 96.7% purity) was obtained as a brown oil. Step C. Procedure for preparation of 4-(3-bromo-2-methyl- phenoxy)cyclohexanecarbaldehyde To a solution of DMSO (5.22 g, 66.84 mmol, 5.22 mL, 4 equiv.) in DCM (200 mL) was added the mixture of (COCl) 2 (4.24 g, 33.42 mmol, 2.93 mL, 2 equiv.) and DCM at -78°C and stirred for 1 hour. Then [4-(3-bromo-2-methyl-phenoxy)cyclohexyl]methanol (5 g, 16.71 mmol, 1.0 equiv.) was added into the mixture and stirred for 1 hour. TEA (10.15 g, 100.27 mmol, 13.96 mL, 6.0 equiv.) was then added into the mixture and stirred at -70 °C for 1 hour. The reaction mixture was diluted with H2O (200 mL) and extracted with DCM 60 mL (30 mL × 2). The combined organic layers were dried over sodium sulfate and concentrated under reduced pressure to give 4-(3-bromo-2-methyl-phenoxy)cyclohexanecarbaldehyde (17 g, crude) as a yellow oil, which was used in the next step without further purification. Step D. Procedure for preparation of ethyl (E)-3-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]prop-2-enoate A mixture of 4-(3-bromo-2-methyl-phenoxy)cyclohexanecarbaldehyde (17 g, 57.20 mmol, 1.0 equiv.), ethyl 2-diethoxyphosphorylacetate (17.95 g, 80.08 mmol, 15.89 mL, 1.4 equiv.), and NaH (3.66 g, 91.52 mmol, 60% purity, 1.6 equiv.) in THF (200 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 0 °C for 2 hours under N 2 atmosphere. The reaction mixture was quenched by addition NH4Cl (50 mL) at 25 °C, and then diluted with NH4Cl (50 mL) and extracted with ethyl acetate 300 mL (100 mL × 3). The combined organic layers were then dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~5% ethyl acetate/petroleum ether). The compound ethyl (E)-3-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]prop-2-enoate (8 g, 21.78mmol, 38.0% yield) was obtained as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ = 7.17 - 7.13 (m, 1H), 7.11 - 7.05 (m, 1H), 7.05 - 7.00 (m, 1H), 6.85 (dd, J = 6.8, 15.8 Hz, 1H), 5.83 (dd, J = 1.2, 15.8 Hz, 1H), 4.31 - 4.20 (m, 1H), 4.11 (q, J = 7.2 Hz, 2H), 2.23 (m, 1H), 2.21 (s, 3H), 2.12 - 2.04 (m, 2H), 1.82 (d, J = 11.2 Hz, 2H), 1.49 - 1.39 (m, 2H), 1.37 - 1.26 (m, 2H), 1.21 (t, J = 7.2 Hz, 3H) Step E. Procedure for preparation of ethyl 3-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]propanoate A mixture of ethyl (E)-3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]prop-2-enoate (8 g, 21.78 mmol, 1 equiv.) and PtO 2 (4.95 g, 21.78 mmol, 1.0 equiv.) in EtOH (100 mL) was degassed and purged with H 2 three times, and then the mixture was stirred at 25 °C for 2 hours under H 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The compound ethyl 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]propanoate (8 g, crude) was obtained as a brown oil. Step F. Procedure for preparation of 3-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]propan-1-ol To a mixture of ethyl 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]propanoate (8 g, 21.66 mmol, 1.0 equiv.) in THF (100 mL) was slowly added LAH (986.65 mg, 26.00 mmol, 1.2 equiv.) at 0 °C and then the mixture was stirred at 0 ℃ for 1.5 hours. The mixture was quenched by 2.0 g Na 2 SO 4 .10 H 2 O. The solution was slowly poured into ice-water (80 mL) and the pH was acidized to pH 4~5 with 1 M HCl. The mixture was extracted with EtOAc 180 mL (60 mL×3), washed with brine 100 mL (50 mL×2), dried by sodium sulfate, filtered, and concentrated under reduced pressure to give compound 3-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]propan-1-ol (5 g, crude) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.15 - 7.11 (m, 1H), 7.06 (t, J = 8.0 Hz, 1H), 7.02 - 6.99 (m, 1H), 4.36 (t, J = 5.2 Hz, 1H), 4.21 (m, J = 4.4, 10.6 Hz, 1H), 3.39 - 3.37 (m, 2H), 2.20 (s, 3H), 2.07 - 2.01 (m, 2H), 1.78 - 1.72 (m, 2H), 1.41 (dd, J = 7.2, 14.7 Hz, 3H), 1.37 - 1.29 (m, 2H), 1.22 - 1.18 (m, 2H), 1.07 - 0.97 (m, 2H) Step G. Procedure for preparation of 3-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]propanal To a solution of DMSO (2.39 g, 30.56 mmol, 2.39 mL, 4.0 equiv.) in DCM (40 mL) was added (COCl)2 (1.94 g, 15.28 mmol, 1.34 mL, 2.0 equiv.) under -78°C and stirred for 1 hour. Then 3- [4-(3-bromo-2-methyl-phenoxy)cyclohexyl]propan-1-ol (2.5 g, 7.64 mmol, 1.0 equiv.) was added into the mixture and stirred for 1 hour. TEA (4.64 g, 45.84 mmol, 6.38 mL, 6.0 equiv.) was then added into the mixture and stirred at -70 °C for 1 hour. The reaction mixture was diluted with H 2 O (30 mL), extracted with DCM 30 mL (15 mL × 2), dried over sodium sulfate, and concentrated under reduced pressure to give the compound 3-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]propanal (2.5 g, crude) as a yellow oil, which was used in the next step without further purification. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 9.67 (t, J = 1.6 Hz, 1H), 7.16 - 7.11 (m, 1H), 7.06 (t, J = 8.0 Hz, 1H), 7.03 - 6.99 (m, 1H), 4.27 - 4.17 (m, 1H), 2.45 (m, J = 1.6, 7.6 Hz, 2H), 2.20 (s, 3H), 2.08 - 2.00 (m, 2H), 1.80 - 1.71 (m, 2H), 1.45 (q, J = 7.2 Hz, 2H), 1.40 - 1.30 (m, 2H), 1.30 - 1.23 (m, 1H), 1.10 - 0.97 (m, 2H) Step H. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[2-methyl-3-[4-(3- oxopropyl)cyclohexoxy]phenyl]pyridine-2-carboxylate A mixture of 3-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]propanal (500 mg, 1.54 mmol, 1.0 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-2-carb oxylate (847.51 mg, 1.38 mmol, 0.9 equiv.), K 2 CO 3 (318.70 mg, 2.31 mmol, 1.5 equiv.), Ad 2 nBuP Pd G 3 (cataCXium® A Pd G3) (75 mg, 102.98 µmol, 0.2 equiv.) in dioxane (5 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 100 °C for 1 hour under microwave. The reaction mixture was filtered and concentrated under reduced pressure to remove solvent. The residue was purified by flash silica gel chromatography (eluent of 0~35% ethyl acetate/petroleum ether). The compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[2-methyl-3-[4-(3-oxopropyl)cyclohexoxy] phenyl]pyridine-2-carboxylate (700 mg, 957.7 µmol, 31.1% yield) was obtained as a yellow solid. MS (ESI) m/z: 731.4 [M+H] + Step I. Procedure for preparation of tert-butyl (3R)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1- methyl-indazol-6-yl]-3-methyl-piperazine-1-carboxylate A mixture of 6-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (1.5 g, 3.00 mmol, 1.0 equiv.), tert-butyl (3R)-3-methylpiperazine-1-carboxylate (1.20 g, 6.00 mmol, 2.0 equiv.), 1,3- bis[2,6-bis(1-propylbutyl)phenyl]-4,5-dichloro-2H-imidazol-1 -ium-2-ide;3- chloropyridine;dichloropalladium (291.61 mg, 299.77 µmol, 0.1 equiv.), and Cs2CO3 (2.93 g, 8.99 mmol, 3 equiv.) in 2-methyl-2-butanol (20 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 100 °C for 2 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to remove solvent. The residue was purified by flash silica gel chromatography (eluent of 0~35% ethyl acetate/petroleum ether). The compound tert-butyl (3R)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6- yl]-3-methyl-piperazine-1-carboxylate (580 mg, 935.8 µmol, 31.2% yield) was obtained as a yellow oil. MS (ESI) m/z: 620.4 [M+H] + Step J. Procedure for preparation of tert-butyl (3R)-4-[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]-3-methyl-piperazine-1-carboxylate A mixture of tert-butyl (3R)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-yl] -3- methyl-piperazine-1-carboxylate (580 mg, 935.86 µmol, 1.0 equiv.), Pd/C (200 mg, 10% purity, 1.0 equiv.), Pd(OH)2 (200 mg, 1.42 mmol, 1.52 equiv.), and AcOH (168.60 mg, 2.81 mmol, 160.57 µL, 3.0 equiv.) in EtOH (5 mL) and THF (5 mL) was degassed and purged with H 2 three times, and then the mixture was stirred at 25 °C for 12 hours under H 2 atmosphere. The mixture was filtered, washed with THF (30 mL), and concentrated under reduced pressure to give the compound tert-butyl (3R)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]-3- methyl-piperazine-1-carboxylate (480 mg, crude) as a yellow solid. MS (ESI) m/z: 442.3 [M+H] + Step K. Procedure for preparation of 3-[1-methyl-6-[(3S)-3-methyl-4-piperidyl]indazol- 3-yl]piperidine-2,6-dione A mixture of tert-butyl (3R)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]-3-m ethyl- piperazine-1-carboxylate (480 mg, 858.85 µmol, 79% purity, 1.0 equiv.) in HCl/EtOAc (7 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 25 °C for 2 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to remove solvent. The compound 3-[1-methyl-6-[(3S)-3-methyl-4-piperidyl]indazol-3- yl]piperidine-2,6-dione (470 mg, crude) was obtained as a brown solid. MS (ESI) m/z: 342.2 [M+H] + Step L. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[(3R)-4-[3-(2,6- dioxo-3-piperidyl)-1-methyl- indazol-6-yl]-3-methyl-piperazin-1-yl]propyl]cyclohexoxy]-2- methyl-phenyl]pyridine-2- carboxylate To a solution of 3-[1-methyl-6-[(3S)-3-methyl-4-piperidyl]indazol-3-yl]piperi dine-2,6-dione (100 mg, 293.76 µmol, 1 equiv.) in DCM (3 mL) was added the mixture of tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[2-methyl-3-[4-(3- oxopropyl)cyclohexoxy]phenyl]pyridine-2-carboxylate (150.30 mg, 205.63 µmol, 0.7 equiv.) and acetic acid at 25 °C. The mixture was stirred for 11 hours. NaBH(OAc) 3 (186.78 mg, 881.27 µmol, 3.0 equiv.) was added into the mixture and stirred for 1 hour. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by flash silica gel chromatography (eluent of 0~87% ethyl acetate/petroleum ether). The compound tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4- [3-[(3R)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl] -3-methyl-piperazin-1- yl]propyl]cyclohexoxy]-2-methyl-phenyl]pyridine-2-carboxylat e (140 mg, 132.5 µmol, 45.1% yield) was obtained as a yellow solid. MS (ESI) m/z: 529.1 [M/2+H] + Step M. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-(3-((3R)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-3-methylpiperazin-1-yl)propyl)cycloh exyl)oxy)-2- methylphenyl)picolinic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[4-[3-[(3R)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-i ndazol-6-yl]-3-methyl-piperazin- 1-yl]propyl]cyclohexoxy]-2-methyl-phenyl]pyridine-2-carboxyl ate (140 mg, 132.54 µmol, 1.0 equiv.) in DCM (1.5 mL) and TFA (1.5 mL) was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by prep-HPLC to give compound 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-(3-((3R)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-3-methylpiperazin-1-yl)propyl)cycloh exyl)oxy)-2- methylphenyl)picolinic acid (20.2 mg, 19.0 µmol, 14.3% yield, 94.2% purity) as a brown solid. MS (ESI) m/z: 501.1 [M/2+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.86 (s, 1H), 10.90 - 10.84 (m, 1H), 9.16 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.56 (d, J = 8.8 Hz, 1H), 7.50 - 7.41 (m, 3H), 7.37 (m, 2H), 7.08 (t, J = 7.8 Hz, 1H), 6.99 - 6.89 (m, 4H), 6.62 (d, J = 8.0 Hz, 1H), 4.98 (s, 2H), 4.49 (d, J = 2.0 Hz, 1H), 4.25 (s, 2H), 3.97 (s, 1H), 3.94 - 3.89 (m, 4H), 3.73 - 3.49 (m, 3H), 3.19 - 3.12 (m, 3H), 3.03 (t, J = 5.2 Hz, 3H), 2.68 - 2.61 (m, 2H), 2.54 (s, 4H), 2.33 (s, 1H), 2.19 - 2.09 (m, 3H), 1.89 - 1.80 (m, 5H), 1.45 - 1.31 (m, 3H), 1.24 (d, J = 4.6 Hz, 2H), 1.09 (d, J = 7.0 Hz, 3H), 0.96 (d, J = 6.8 Hz, 1H) Example 213. Preparation of Compound 273a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1S,4r)-4-(3- ((2R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)-2-methylpiperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl (2R)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1- methyl-indazol-6-yl]-2-methyl-piperazine-1-carboxylate A mixture of 6-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (1.5 g, 3.00 mmol, 1 equiv.), tert-butyl (2R)-2-methylpiperazine-1-carboxylate (1.80 g, 8.99 mmol, 3 equiv.), RuPhos (279.77 mg, 599.54 µmol, 0.2 equiv.), Pd 2 (dba) 3 (274.50 mg, 299.77 µmol, 0.1 equiv.), and Cs 2 CO 3 (2.93 g, 8.99 mmol, 3 equiv.) in toluene (2 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 110 °C for 16 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~30% ethyl acetate/petroleum ether) to afford tert-butyl (2R)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl- indazol-6-yl]-2-methyl-piperazine-1-carboxylate (900 mg, 1.2 mmol, 42.6% yield, 88% purity) as a yellow oil. MS (ESI) m/z: 620.5 [M+H] + . Step B. Procedure for preparation of tert-butyl (2R)-4-[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]-2-methyl-piperazine-1-carboxylate To a solution of tert-butyl (2R)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-yl] -2- methyl-piperazine-1-carboxylate (1.05 g, 1.69 mmol, 1 equiv.) in EtOH (10 mL) and THF (10 mL) was added Pd/C (300 mg, 1.69 mmol, 10% purity, 1.00 equiv.), Pd(OH) 2 (300 mg, 213.62 µmol, 10% purity, 1.26e-1 equiv.), and AcOH (101.74 mg, 1.69 mmol, 96.90 µL, 1 equiv.) under N2 atmosphere. The suspension was degassed and purged with H2 three times. The mixture was then stirred under H 2 (15 Psi) at 25 °C for 16 hours. The reaction mixture was filtered and washed with THF (50 mL). The filtrate was concentrated to afford tert-butyl (2R)- 4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]-2-methyl -piperazine-1-carboxylate (630 mg, crude) as a black brown oil, and was used in the next step without further purification. Step C. Procedure for preparation of 3-[1-methyl-6-[(3R)-3-methylpiperazin-1- yl]indazol-3-yl]piperidine-2,6-dione To a solution of tert-butyl (2R)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]-2-m ethyl- piperazine-1-carboxylate (630 mg, 1.43 mmol, 1 equiv.) in dioxane (8 mL) was added HCl/dioxane (4 M, 8 mL, 22.43 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was filtered to afford 3-[1-methyl-6-[(3R)-3-methylpiperazin-1-yl]indazol-3- yl]piperidine-2,6-dione (400 mg, crude) as a black brown solid, which was used in the next step without further purification. Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[(2R)-4-[3-(2,6- dioxo-3-piperidyl)-1-methyl- indazol-6-yl]-2-methyl-piperazin-1-yl]propyl]cyclohexoxy]-2- methyl-phenyl]pyridine-2- carboxylate To a solution of 3-[1-methyl-6-[(3R)-3-methylpiperazin-1-yl]indazol-3-yl]pipe ridine-2,6- dione (84.08 mg, 246.27 µmol, 1.2 equiv.) in DCM (2 mL) was added tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[2-methyl-3-[4-(3- oxopropyl)cyclohexoxy]phenyl]pyridine-2-carboxylate (150 mg, 205.22 µmol, 1 equiv.). The mixture was stirred at 25 °C for 1 hour, and then NaBH(OAc)3 (130.49 mg, 615.67 µmol, 3 equiv.) was added and the mixture was stirred at 25 °C for 16 h. The reaction mixture was filtered and concentrated under vacuum to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~10% methanol/dichloromethane) to afford tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[3-[(2R)-4- [3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]-2-methyl-p iperazin-1- yl]propyl]cyclohexoxy]-2-methyl-phenyl]pyridine-2-carboxylat e (80 mg, 72.7 µmol, 35.4% yield, 96% purity) as a light yellow solid. MS (ESI) m/z: 1056.5 [M+H] + . Step E. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-(3-((2R)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-2-methylpiperazin-1-yl)propyl)cycloh exyl)oxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[4-[3-[(2R)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl -indazol-6-yl]-2-methyl- piperazin-1-yl]propyl]cyclohexoxy]-2-methyl-phenyl]pyridine- 2-carboxylate (70 mg, 66.27 µmol, 1 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 203.82 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to afford 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1S,4r)-4-(3-((2R)- 4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)-2-m ethylpiperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (27.8 mg, 27.6 µmol, 41.6% yield, 99.1% purity) as a white solid. MS (ESI) m/z: 500.9 [M/2+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.84 (s, 1H), 8.02 (d, J = 7.2 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.65 - 7.59 (m, 1H), 7.51 - 7.42 (m, 4H), 7.40 - 7.32 (m, 2H), 7.10 - 7.03 (m, 1H), 6.97 - 6.89 (m, 3H), 6.82 (s, 1H), 6.65 - 6.58 (m, 1H), 4.98 (s, 2H), 4.29 - 4.15 (m, 1H), 3.95 - 3.86 (m, 5H), 3.56 - 3.49 (m, 2H), 3.05 - 3.00 ( m, 2H), 2.93 - 2.84 (m, 2H), 2.67 (s, 1H), 2.62 - 2.59 (m, 2H), 2.33 - 2.31 (m, 3H), 2.10 - 2.07 (m, 2H), 1.87 (s, 3H), 1.81 - 1.78 (m, 2H), 1.46 - 1.20 (m, 10H), 1.13 - 1.05 (m, 5H) Example 214. Preparation of Compound 278b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1R,4r)-4-(3- ((3S)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)-3-methylpiperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl (3S)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1- methyl-indazol-6-yl]-3-methyl-piperazine-1-carboxylate A mixture of 6-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (1.2 g, 2.40 mmol, 1.0 equiv.) , tert-butyl (3S)-3-methylpiperazine-1-carboxylate (1.92 g, 9.59 mmol, 4.0 equiv.), Cs2CO3 (2.34 g, 7.19 mmol, 3.0 equiv.) and 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5- dichloro-2H-imidazol-1-ium-2-ide;3-chloropyridine;dichloropa lladium (200.00 mg, 205.60 µmol, 0.085 equiv.) in 2-methyl-2-butanol (30 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 100 °C for 16 hours under N2 atmosphere. The mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (eluent of 0~30% ethyl acetate/petroleum ether) to give tert-butyl (3S)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-yl] -3- methyl-piperazine-1-carboxylate (1.2 g, 1.8 mmol, 76.8% yield, 95.2% purity) as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.89 (d, J = 8.0 Hz, 1H), 7.52 (d, J = 9.2 Hz, 1H), 7.49 - 7.44 (m, 2H), 7.43 - 7.33 (m, 5H), 7.32 - 7.25 (m, 3H), 6.85 - 6.78 (m, 2H), 6.57 (d, J = 8.0 Hz, 1H), 5.44 (s, 2H), 5.41 (s, 2H), 4.11 - 4.05 (m, 1H), 3.96 (s, 3H), 3.84 - 3.65 (m, 2H), 3.30 (d, J = 12.0 Hz, 2H), 3.10 - 2.98 (m, 2H), 1.43 (s, 9H), 0.90 (d, J = 6.4 Hz, 3H) Step B. Procedure for preparation of tert-butyl (3S)-4-[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]-3-methyl-piperazine-1-carboxylate To a solution of tert-butyl (3S)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-yl] -3- methyl-piperazine-1-carboxylate (1.2 g, 1.94 mmol, 1.0 equiv.) in THF (5 mL) and EtOH (5 mL) was added Pd/C (300 mg, 10% purity), Pd(OH)2 (300 mg, 2.14 mmol, 1.10 equiv.) and AcOH (11.63 mg, 193.63 µmol, 11.07 µL, 0.1 equiv.) under N 2 atmosphere. The suspension was degassed and purged with H 2 three times. The mixture was then stirred under H 2 (15 Psi) at 25 °C for 16 hours. The reaction mixture was filtered and washed with THF (50 mL). The filtrate was concentrated to give tert-butyl (3S)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol- 6-yl]-3-methyl-piperazine-1-carboxylate (930 mg, 1.90 mmol, 97.9% yield, 90% purity) as a black brown solid. Step C. Procedure for preparation of 3-[1-methyl-6-[(2S)-2-methylpiperazin-1- yl]indazol-3-yl]piperidine-2,6-dione To a mixture of tert-butyl (3S)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]-3-m ethyl- piperazine-1-carboxylate (927.88 mg, 2.10 mmol, 1.0 equiv.) in DCM (5 mL) was added HCl/dioxane (4 M, 9.98 mL, 18.99 equiv.). The reaction was stirred at 25 °C for 2 hours. The reaction was then filtered to give 3-[1-methyl-6-[(2S)-2-methylpiperazin-1-yl]indazol-3- yl]piperidine-2,6-dione (600 mg, 1.4 mmol, 70.1% yield, 92.8% purity) as a white solid. Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[(3S)-4-[3-(2,6- dioxo-3-piperidyl)-1-methyl- indazol-6-yl]-3-methyl-piperazin-1-yl]propyl]cyclohexoxy]-2- methyl-phenyl]pyridine-2- carboxylate A mixture of 3-[1-methyl-6-[(2S)-2-methylpiperazin-1-yl]indazol-3-yl]pipe ridine-2,6-dione (270 mg, 714.54 µmol, 3.48 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[2-methyl-3-[4-(3-oxopropyl)c yclohexoxy]phenyl]pyridine- 2-carboxylate (150 mg, 205.22 µmol, 1.0 equiv.), and NaBH(OAc) 3 (260.97 mg, 1.23 mmol, 6.0 equiv.) in DCM (2 mL) was stirred at 25 °C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep- TLC (SiO2, DCM: MeOH = 15:1) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[3-[(3S)-4-[3-(2,6- dioxo-3-piperidyl)-1-methyl- indazol-6-yl]-3-methyl-piperazin-1-yl]propyl]cyclohexoxy]-2- methyl-phenyl]pyridine-2- carboxylate (60 mg, 55.2 µmol, 26.9% yield, 97.3% purity) as a brown oil. MS (ESI) m/z: 529.1 [M+H] + . Step E. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-(3-((3S)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-3-methylpiperazin-1-yl)propyl)cycloh exyl)oxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[4-[3-[(3S)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl -indazol-6-yl]-3-methyl- piperazin-1-yl]propyl]cyclohexoxy]-2-methyl-phenyl]pyridine- 2-carboxylate (60 mg, 56.80 µmol, 1 equiv.) in TFA (2 mL) and DCM (2 mL) .The mixture was stirred at 25 °C for 12 hours. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1R,4r)-4-(3-((3S)-4-(3-(2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-6- yl)-3-methylpiperazin-1-yl)propyl)cyclohexyl)oxy)-2-methylph enyl)picolinic acid (16.5 mg, 14.9 µmol, 26.2% yield, 89.0% purity) as a white solid. MS (ESI) m/z: 500.8 [M/2+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.84 (s, 1H), 8.17 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.51 - 7.41 (m, 4H), 7.39 - 7.31 (m, 2H), 7.11 - 7.03 (m, 1H), 6.96 - 6.86 (m, 3H), 6.77 (s, 1H), 6.62 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.29 - 4.18 (m, 2H), 4.14 - 4.07 (m, 1H), 3.94 - 3.86 (m, 5H), 3.02 (m, 3H), 2.87 (d, J = 10.4 Hz, 1H), 2.72 (d, J = 10.0 Hz, 1H), 2.64 - 2.59 (m, 2H), 2.34 - 2.23 (m, 4H), 2.19 - 2.06 (m, 4H), 1.87 (s, 3H), 1.84 - 1.75 (m, 2H), 1.52 - 1.44 (m, 2H), 1.41 - 1.22 (m, 6H), 1.15 - 1.05 (m, 2H), 1.01 (d, J = 6.4 Hz, 3H) Example 215. Preparation of Compound 266a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4-[(3R)-3-[1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-o xo-ethyl]-4-piperidyl]butoxy]- 2-methyl-phenyl]pyridine-2-carboxylic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(4-((3R)-3-(1- (2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)am ino)-2-oxoethyl)piperidin-4- yl)butoxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of tert-butyl 4-[3-(4-bromo-3-methyl-phenoxy)-1- methyl-propyl]piperidine-1-carboxylate To a solution of tert-butyl 4-(3-hydroxy-1-methyl-propyl)piperidine-1-carboxylate (2.2 g, 8.55 mmol, 1.0 equiv.) and 4-bromo-3-methyl-phenol (1.76 g, 9.40 mmol, 1.1 equiv.) in toluene (20 mL) was added 2-(tributyl-λ5-phosphanylidene)acetonitrile (2.48 g, 10.26 mmol, 1.2 equiv.). The mixture was stirred at 120 °C for 12 hours. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by reverse-phase HPLC to give tert-butyl 4-[3-(4-bromo-3-methyl-phenoxy)-1-methyl-propyl]piperidine-1 -carboxylate (2.7 g, 6.1 mmol, 72.1% yield, 97.4% purity) as a brown oil. MS (ESI) m/z: 448.2 [M+23] + 1 H NMR (400 MHz, DMSO-d6) δ = 7.42 (d, J = 8.8 Hz, 1H), 6.95 (d, J = 2.8 Hz, 1H), 6.71 (dd, J = 2.8, 8.8 Hz, 1H), 4.06 - 3.89 (m, 4H), 2.72 - 2.54 (m, 2H), 2.29 (s, 3H), 1.84 - 1.74 (m, 1H), 1.58 - 1.45 (m, 4H), 1.38 (s, 10H), 1.15 - 0.97 (m, 2H), 0.85 (d, J = 6.4 Hz, 3H) Step B. Procedure for preparation of tert-butyl 4-[(1S)-3-(4-bromo-3-methyl-phenoxy)-1- methyl-propyl]piperidine-1-carboxylate The compound tert-butyl 4-[3-(4-bromo-3-methyl-phenoxy)-1-methyl-propyl]piperidine-1 - carboxylate (1.4 g, 3.28 mmol, 1.0 equiv.) was purified by prep-HPLC to give tert-butyl 4- [(1R)-3-(4-bromo-3-methyl-phenoxy)-1-methyl-propyl]piperidin e-1-carboxylate (430 mg, 1.00 mmol, 30.51% yield, 99.32% purity) as a brown oil and tert-butyl 4-[(1S)-3-(4-bromo-3- methyl-phenoxy)-1-methyl-propyl]piperidine-1-carboxylate (380 mg, 865.7 µmol, 26.3% yield, 97.1% purity) as a brown oil MS (ESI) m/z: 448.2 [M+23] + MS (ESI) m/z: 448.2 [M+23] + Step C. Procedure for preparation of 4-[(1S)-3-(4-bromo-3-methyl-phenoxy)-1-methyl- propyl]piperidine A mixture of tert-butyl 4-[(1S)-3-(4-bromo-3-methyl-phenoxy)-1-methyl-propyl]piperid ine-1- carboxylate (430 mg, 1.01 mmol, 1.0 equiv.) and HCl/EtOAc (4 M, 5 mL, 19.83 equiv.) was stirred at 25 °C for 1 hour. The mixture was concentrated under reduced pressure to give 4- [(1R)-3-(4-bromo-3-methyl-phenoxy)-1-methyl-propyl]piperidin e (380 mg, crude) as a brown solid. MS (ESI) m/z: 328.2 [M+H] + ( 80 Br). Step D. Procedure for preparation of ethyl 2-[4-[(1S)-3-(4-bromo-3-methyl-phenoxy)-1- methyl-propyl]-1-piperidyl]acetate To a solution of 4-[(1R)-3-(4-bromo-3-methyl-phenoxy)-1-methyl-propyl]piperid ine (380 mg, 1.05 mmol, 1.0 equiv., HCl) and ethyl 2-bromoacetate (157.46 mg, 942.85 µmol, 104.28 µL, 0.9 equiv.) in CH3CN (4 mL) in DMF (0.5 mL) was added K2CO3 (434.36 mg, 3.14 mmol, 3.0 equiv.). The mixture was stirred at 60 °C for 1 hour. The mixture was then filtered to give a filtrate, which was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether / ethyl acetate= 5/1 to 4/1) to give ethyl 2-[4-[(1R)-3-(4-bromo-3-methyl-phenoxy)-1-methyl-propyl]-1-p iperidyl]acetate (370 mg, 892.1 µmol, 85.1% yield, 99.4% purity) as a light yellow oil. MS (ESI) m/z: 414.2 [M+2+H] + ( 80 Br). 1 H NMR (400 MHz, DMSO-d6) δ = 7.42 (d, J = 8.8 Hz, 1H), 6.95 (d, J = 2.8 Hz, 1H), 6.71 (dd, J = 2.8, 8.8 Hz, 1H), 4.07 (q, J = 7.2 Hz, 2H), 4.03 - 3.90 (m, 2H), 3.14 (s, 2H), 2.88 - 2.80 (m, 2H), 2.29 (s, 3H), 2.13 - 2.04 (m, 2H), 1.84 - 1.75 (m, 1H), 1.56 - 1.43 (m, 4H), 1.33 - 1.21 (m, 2H), 1.21 - 1.15 (m, 4H), 0.86 (d, J = 6.4 Hz, 3H) Step E. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[(3R)-3-[1-(2-ethoxy-2 -oxo-ethyl)-4- piperidyl]butoxy]-2-methyl-phenyl]pyridine-2-carboxylate Ethyl 2-[4-[(1R)-3-(4-bromo-3-methyl-phenoxy)-1-methyl-propyl]-1-p iperidyl]acetate (150 mg, 363.76 µmol, 1.0 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborol an-2-yl)pyridine-2-carboxylate (222.82 mg, 363.76 µmol, 1.0 equiv.), Ad 2 nBuP Pd G 3 (26.49 mg, 36.38 µmol, 0.1 equiv.), and KF (63.40 mg, 1.09 mmol, 25.56 µL, 3.0 equiv.) were taken up into a microwave tube in dioxane (2 mL) and H2O (0.2 mL). The sealed tube was heated at 100 °C for 1 hour under microwave. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=3/1 to 2/1) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[4-[(3R)-3-[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]butox y]-2-methyl-phenyl]pyridine-2- carboxylate (170 mg, 179.1 µmol, 49.2% yield, 86.1% purity) as a yellow oil. MS (ESI) m/z: 818.5 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 12.98 - 12.70 (m, 1H), 8.01 (d, J = 7.6 Hz, 1H), 7.77 (d, J = 8.0 Hz, 1H), 7.59 (d, J = 7.6 Hz, 1H), 7.50 - 7.28 (m, 5H), 6.92 (d, J = 8.8 Hz, 1H), 6.85 (d, J = 8.4 Hz, 1H), 6.83 - 6.80 (m, 1H), 6.72 (dd, J = 2.4, 8.4 Hz, 1H), 4.96 (s, 2H), 4.06 (q, J = 6.8 Hz, 2H), 4.03 - 3.91 (m, 2H), 3.86 (t, J = 6.0 Hz, 2H), 3.14 (s, 2H), 3.03 (t, J = 6.0 Hz, 2H), 2.87 - 2.80 (m, 2H), 2.12 - 2.03 (m, 2H), 2.02 - 1.97 (m, 3H), 1.82 - 1.75 (m, 1H), 1.56 - 1.45 (m, 4H), 1.26 - 1.22 (m, 2H), 1.20 - 1.15 (m, 4H), 1.07 - 0.99 (m, 9H), 0.86 (d, J = 6.4 Hz, 3H) Step F. Procedure for preparation of 2-[4-[(1R)-3-[4-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-3- methyl-phenoxy]-1-methyl-propyl]-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[4-[(3R)-3-[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]but oxy]-2-methyl-phenyl]pyridine- 2-carboxylate (170 mg, 207.82 µmol, 1.0 equiv.) in THF (1.5 mL) and H2O (0.5 mL) was added LiOH•H2O (26.16 mg, 623.45 µmol, 3.0 equiv.).The mixture was stirred at 25 °C for 16 hours. The mixture was concentrated under reduced pressure to remove THF, diluted with H 2 O (5 mL), added HCl (1 M) to pH= 2, filtered, and concentrated under reduced pressure to give 2- [4-[(1R)-3-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dih ydro-1H-isoquinolin-2-yl]-2- tert-butoxycarbonyl-3-pyridyl]-3-methyl-phenoxy]-1-methyl-pr opyl]-1-piperidyl]acetic acid (160 mg, crude) as a yellow solid. MS (ESI) m/z: 790.5 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 13.15 - 12.50 (m, 1H), 8.08 - 7.96 (m, 1H), 7.86 - 7.70 (m, 1H), 7.66 - 7.54 (m, 1H), 7.53 - 7.24 (m, 5H), 6.95 - 6.89 (m, 1H), 6.86 (d, J = 8.0 Hz, 1H), 6.84 - 6.79 (m, 1H), 6.76 - 6.69 (m, 1H), 4.96 (s, 2H), 4.06 - 3.93 (m, 3H), 3.88 - 3.84 (m, 2H), 3.39 - 3.34 (m, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.80 - 2.70 (m, 2H), 2.02 - 1.98 (m, 3H), 1.84 - 1.77 (m, 1H), 1.72 - 1.67 (m, 2H), 1.58 - 1.47 (m, 4H), 1.23 (s, 2H), 1.03 (s, 9H), 0.87 (d, J = 6.0 Hz, 3H) Step G. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[(3R)-3-[1-[2-[[3-(2,6 -dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-4-piperidyl]butoxy]-2-methy l-phenyl]pyridine-2- carboxylate To a solution of 2-[4-[(1R)-3-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-d ihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-3-methyl- phenoxy]-1-methyl-propyl]-1- piperidyl]acetic acid (160 mg, 202.54 µmol, 1.0 equiv.) and HATU (92.41 mg, 243.04 µmol, 1.2 eq.) in DMF (1.5 mL) was added DIEA (78.53 mg, 607.61 µmol, 105.83 µL, 3.0 equiv.) and 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (62.77 mg, 243.04 µmol, 1.2 equiv.). The mixture was stirred at 25 °C for 16 hours. The mixture was added into H2O (10 mL), filtered, and concentrated under reduced pressure to give tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[4-[(3R)-3-[1-[2-[[3-(2,6- dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl] -4-piperidyl]butoxy]-2-methyl- phenyl]pyridine-2-carboxylate (140 mg, crude) as a pink solid. MS (ESI) m/z: 1030.6 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.13 - 12.25 (m, 1H), 10.88 (s, 1H), 9.82 (s, 1H), 8.09 - 8.04 (m, 1H), 7.99 (d, J = 8.0 Hz, 1H), 7.75 (d, J = 8.0 Hz, 1H), 7.66 - 7.56 (m, 2H), 7.48 - 7.40 (m, 3H), 7.38 - 7.29 (m, 2H), 7.25 - 7.18 (m, 1H), 6.92 (d, J = 8.8 Hz, 1H), 6.86 (d, J = 8.4 Hz, 1H), 6.84 - 6.80 (m, 1H), 6.76 - 6.70 (m, 1H), 4.97 (s, 2H), 4.36 - 4.28 (m, 1H), 4.08 - 3.89 (m, 5H), 3.86 (t, J = 5.6 Hz, 2H), 3.12 (s, 2H), 3.04 - 3.01 (m, 2H), 2.97 - 2.89 (m, 2H), 2.17 - 2.08 (m, 2H), 2.01 (s, 3H), 1.92 - 1.80 (m, 2H), 1.65 - 1.44 (m, 6H), 1.41 - 1.33 (m, 2H), 1.17 (s, 2H), 1.03 (s, 9H), 0.90 (d, J = 6.8 Hz, 3H) Step H. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[4-[(3R)-3-[1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]butoxy]-2-methyl-phenyl]p yridine-2-carboxylic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(4-((3R)-3- (1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl )amino)-2- oxoethyl)piperidin-4-yl)butoxy)-2-methylphenyl)picolinic acid] To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[4-[(3R)-3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methy l-indazol-6-yl]amino]-2-oxo- ethyl]-4-piperidyl]butoxy]-2-methyl-phenyl]pyridine-2-carbox ylate (140 mg, 135.89 µmol, 1.0 equiv.) in DCM (0.7 mL) was added TFA (1.08 g, 9.45 mmol, 0.7 mL, 69.57 equiv.). The mixture was stirred at 25 °C for 12 hours. The mixture was concentrated under reduced pressure to remove DCM. The mixture was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[(3R)-3-[ 1-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]-4-piper idyl]butoxy]-2-methyl- phenyl]pyridine-2-carboxylic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-((3R)-3-(1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)butoxy)-2-meth ylphenyl)picolinic acid] (26.1 mg, 24.8 µmol, 18.3% yield, 92.5% purity) as a yellow solid. MS (ESI) m/z: 974.5 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 12.99 - 12.74 (m, 1H), 10.89 (s, 1H), 8.10 - 7.94 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.69 - 7.59 (m, 2H), 7.50 - 7.41 (m, 3H), 7.40 - 7.32 (m, 2H), 7.20 - 7.14 (m, 1H), 6.99 - 6.88 (m, 2H), 6.82 - 6.78 (m, 1H), 6.75 - 6.68 (m, 1H), 4.97 (s, 2H), 4.37 - 4.29 (m, 1H), 4.02 - 3.95 (m, 2H), 3.95 - 3.86 (m, 5H), 3.02 (t, J = 5.6 Hz, 2H), 2.66 - 2.62 (m, 2H), 2.61 - 2.55 (m, 4H), 2.20 - 2.15 (m, 1H), 2.03 (s, 3H), 1.91 - 1.77 (m, 2H), 1.74 - 1.41 (m, 7H), 1.31 - 1.11 (m, 2H), 0.91 (d, J = 6.4 Hz, 3H) Example 216. Preparation of Compound 126 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(4-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2- oxoethyl)piperazin-1- yl)ethoxy)-2-(trifluoromethyl)phenyl)picolinic acid Step A. Procedure for preparation of 1-bromo-3-(2-bromoethoxy)-2- (trifluoromethyl)benzene 3-Bromo-2-(trifluoromethyl)phenol (601 mg, 1 equiv.2.49 mmol), 1,2-dibromoethane (4.68 g, 2.15 mL, 10 equiv., 24.9 mmol), and potassium carbonate (431 mg, 1.25 equiv., 3.12 mmol) were combined in DMF (10 mL) and heated overnight at 50 °C. The reaction mixture was diluted with ethyl acetate (100 mL) and brine (25 mL). The layers were separated, and the organic layers were dried over anhydrous magnesium sulfate, filtered, and concentrated to an oil, which was purified by silica gel chromatography (heptane/ethyl acetate gradient). The combined and concentrated product containing fractions afforded 1-bromo-3-(2- bromoethoxy)-2-(trifluoromethyl)benzene (602 mg, 1.73 mmol, 69.4% yield). MS (ESI) m/z: 429.0 [M+H 2 Br]+. Step B. Procedure for preparation of methyl 2-(piperazin-1-yl)acetate hydrochloride tert-Butyl 4-(2-methoxy-2-oxoethyl)piperazine-1-carboxylate (1.0 g, 1 equiv., 3.9 mmol) and HCl (0.71 g, 4.8 mL, 4 molar, 5 equiv., 19 mmol) (as a solution in dioxane) were combined in DCM (20 mL). The reaction was stirred overnight at ambient temperature. The reaction mixture was then concentrated to dryness, the solids were triturated in a mixture of diethyl ether and hexanes, and reconcentrated to provide methyl 2-(piperazin-1-yl)acetate hydrochloride (0.733 g, 3.77 mmol, 97% yield). MS (ESI) m/z: 159.1 [M+H]+. Step C. Procedure for preparation of methyl 2-(4-(2-(3-bromo-2- (trifluoromethyl)phenoxy)ethyl)piperazin-1-yl)acetate Methyl 2-(piperazin-1-yl)acetate hydrochloride (281 mg, 1 equiv., 1.44 mmol), 1-bromo-3-(2- bromoethoxy)-2-(trifluoromethyl)benzene (602 mg, 1.2 equiv., 1.73 mmol), and potassium carbonate (598 mg, 3 equiv., 4.33 mmol) were combined in DMF (8 mL) and heated overnight at 50 °C. The reaction mixture was diluted with ethyl acetate (100 mL) and brine (25 mL). The layers were separated, and the organic layer was dried over anhydrous magnesium sulfate, filtered, and concentrated to an oil, that was purified by silica gel chromatography (dichloromethane/methanol gradient). The combined and concentrated product containing fractions afforded methyl 2-(4-(2-(3-bromo-2-(trifluoromethyl)phenoxy)ethyl)piperazin- 1- yl)acetate (398 mg, 936 µmol, 64.9% yield). MS (ESI) m/z: 426.9 [M+H]+. Step D. Procedure for preparation of methyl 2-(4-(2-(3-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-2-(trifluoromethyl)phenoxy)ethyl)piperazi n-1-yl)acetate Methyl 2-(4-(2-(3-bromo-2-(trifluoromethyl)phenoxy)ethyl)piperazin- 1-yl)acetate (398 mg, 1 equiv., 936 µmol), bis(pinacolato)diboron (404 mg, 1.7 equiv., 1.59 mmol), and PdCl2(dppf) (76.4 mg, 0.1 equiv., 93.6 µmol) were combined in 1,4-dioxane (5 mL). Potassium acetate (276 mg, 3 equiv., 2.81 mmol) was added and the reaction mixture stirred at 90 °C overnight. The reaction mixture was then diluted with 125 mL ethyl acetate, 25 mL water, and 25 mL brine. The layers were separated and the organic layer was washed with an additional 50 mL brine, dried over anhydrous magnesium sulfate, filtered, and concentrated to dryness under reduced pressure. The residue was purified on silica gel with hexanes/ethyl acetate. The product containing fractions were combined, concentrated, and dried under vacuum to afford methyl 2-(4-(2-(3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2- (trifluoromethyl)phenoxy)ethyl)piperazin-1-yl)acetate (328 mg, 694 µmol, 74.2% yield). MS (ESI) m/z: 473.2 [M+H] + . Step E. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-(2-methoxy-2-oxoethy l)piperazin-1-yl)ethoxy)-2- (trifluoromethyl)phenyl)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- bromopicolinate (327 mg, 1.0 equiv., 579 µmol) and methyl 2-(4-(2-(3-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)-2-(trifluoromethyl)phenoxy)ethyl)pi perazin-1-yl)acetate (328 mg, 1.2 equiv., 694 µmol) were combined in 1,4-dioxane (4.0 mL). An aqueous solution of potassium phosphate tribasic (369 mg, 1.16 mL, 1.5 molar, 3 equiv., 1.74 mmol) and cataCXium ® Pd G3 (21.1 mg, 0.05 equiv., 28.9 µmol) was added. The reaction was stirred at 100 °C for 45 minutes using microwave heating and then cooled to ambient temperature. The reaction mixture was diluted with 125 mL ethyl acetate, 25 mL water, and 25 mL brine. The layers were separated and the organic layer was washed with an additional 50 mL brine, dried over anhydrous magnesium sulfate, filtered, and concentrated to dryness under reduced pressure. The residue was purified by silica gel chromatography (hexanes/ethyl acetate). The product containing fractions were combined and concentrated to afford tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(2-(4-(2-methoxy-2- oxoethyl)piperazin-1-yl)ethoxy)-2-(trifluoromethyl)phenyl)pi colinate (82 mg, 99 µmol, 17% yield). MS (ESI) m/z: 831.3 [M+H]+. Step F. Procedure for preparation of 2-(4-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyri din-3-yl)-2- (trifluoromethyl)phenoxy)ethyl)piperazin-1-yl)acetic acid tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(4- (2-methoxy-2-oxoethyl)piperazin-1-yl)ethoxy)-2-(trifluoromet hyl)phenyl)picolinate was dissolved in a mixture of THF (12 mL), methanol (3 mL), and water (3 mL). Lithium hydroxide (24 mg, 10 equiv., 0.99 mmol) was added and the reaction was stirred at ambient temperature overnight. The pH of the reaction was adjusted to ~ 9-10 with a 1N aqueous solution of HCl and the volatiles were removed under reduced pressure. The residue was purified by reverse- phase HPLC (ACN/Water with 0.1% formic acid). The combined product containing fractions were lyophilized overnight to afford 2-(4-(2-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- (trifluoromethyl)phenoxy)ethyl)piperazin-1-yl)acetic acid (50 mg, 61 µmol, 62% yield). MS (ESI) m/z: 817.1 [M+H]+. Step G. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)piperazin-1-yl)ethoxy)-2- (trifluoromethyl)phenyl)picolinate 2-(4-(2-(3-(6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin-2(1H)-yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-2-(trifluoromethyl)phenoxy)ethy l)piperazin-1-yl)acetic acid (50 mg, 1 equiv., 61 µmol) was dissolved in a mixture of DMF (5 mL) and DIEA (40 mg, 53 µL, 5 equiv., 0.31 mmol). HATU (28 mg, 1.2. equiv., 73 µmol) was added and the reaction was stirred at ambient temperature for a few minutes. 3-(6-amino-1-methyl-1H-indazol-3- yl)piperidine-2,6-dione (16 mg, 1 equiv., 61 µmol) was then added and the reaction was stirred overnight at ambient temperature. The reaction mixture was diluted with ethyl acetate (125 mL) and brine (30 mL). The layers were separated, and the organic layer dried over anhydrous magnesium sulfate, filtered, and concentrated to afford tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-(2- ((3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)piperazin-1-yl)etho xy)-2- (trifluoromethyl)phenyl)picolinate (65 mg, 61 µmol, quantitative yield assumed). MS (ESI) m/z: 1057.0 [M+H] + . Step H. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)piperazin-1-yl)ethoxy)-2- (trifluoromethyl)phenyl)picolinic acid tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(4- (2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)am ino)-2-oxoethyl)piperazin-1- yl)ethoxy)-2-(trifluoromethyl)phenyl)picolinate (65 mg, 1 equiv., 61 µmol) were dissolved in a mixture of DCM (10 mL) and trifluoroacetic acid (3.5 g, 2.4 mL, 500 equiv., 31 mmol). The reaction was stirred for about 60 hours at ambient temperature. The volatiles were removed under reduced pressure and the material purified by reverse-phase HPLC. The product containing fractions were lyophilized overnight to afford 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-(2- ((3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)piperazin-1-yl)etho xy)-2- (trifluoromethyl)phenyl)picolinic acid (40 mg, 40 µmol, 65% yield for two steps). NMR was consistent with structure. MS (ESI) m/z: 501.2 [M+2H/2] + Example 217. Preparation of Compound 136 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)oxy)ethyl )piperazin-1-yl)propoxy)-2- methylphenyl)picolinic acid Step A. Procedure for preparation of (3-(3-bromo-2-methylphenoxy)propoxy)(tert- butyl)dimethylsilane 3-Bromo-2-methylphenol (2.00 g, 1 Eq, 10.7 mmol), (3-bromopropoxy)(tert- butyl)dimethylsilane (3.39 g, 1.25 equiv., 13.4 mmol), and potassium carbonate (2.22 g, 1.50 equiv., 16.0 mmol) were combined in DMF and heated overnight at 50 °C. The reaction mixture was diluted with ethyl acetate (100 mL) and brine (25 mL). The layers were separated and the organic layers were dried over anhydrous magnesium sulfate, filtered, and concentrated to an oil that was purified by silica gel chromatography (heptane/ethyl acetate gradient). The combined and concentrated product containing fractions afforded (3-(3-bromo-2- methylphenoxy)propoxy)(tert-butyl)dimethylsilane (3.49 g, 9.71 mmol, 90.8% yield). NMR was consistent with structure. Step B. Procedure for preparation of tert-butyldimethyl(3-(2-methyl-3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)propoxy)silane Combined (3-(3-bromo-2-methylphenoxy)propoxy)(tert-butyl)dimethylsila ne (1.00 g, 1 equiv., 2.78 mmol), bis(pinacolato)diboron (1.20 g, 1.7 equiv., 4.73 mmol), and PdCl 2 (dppf) (227 mg, 0.1 equiv., 278 µmol) in 1,4-dioxane (15 mL). Potassium acetate (819 mg, 3 equiv., 8.35 mmol) was added and the mixture stirred at 90 °C overnight. The reaction mixture was diluted with 125 mL ethyl acetate, 25 mL water, and 25 mL brine. The layers were separated and the organic layer was washed with an additional 50 mL brine, dried over anhydrous magnesium sulfate, filtered, and concentrated to dryness under reduced pressure. The residue was purified by silica gel chromatography (hexanes/ethyl acetate). The product containing fractions were combined, concentrated, and dried under reduced pressure to afford tert- butyldimethyl(3-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxa borolan-2- yl)phenoxy)propoxy)silane (0.862 g, 2.12 mmol, 76.2% yield). was consistent with structure. MS (ESI) m/z: 407.4 [M+H]+. Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((tert-butyldimethylsil yl)oxy)propoxy)-2- methylphenyl)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- bromopicolinate (662 mg, 1 equiv., 1.17 mmol) and tert-butyldimethyl(3-(2-methyl-3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)propoxy)silane (571 mg, 1.2 equiv., 1.40 mmol) were combined in 1,4-dioxane (8.0 mL). Potassium phosphate (745 mg, 2.34 mL, 1.5 molar, 3 equiv., 3.51 mmol) and cataCXium ® Pd G3 (42.6 mg, 0.05 equiv., 58.5 µmol) were added to the mixture. The reaction was stirred at 100 °C for 40 minutes in the microwave and then cooled to ambient temperature overnight. The reaction mixture was then diluted with 125 mL ethyl acetate, 25 mL water, and 25 mL brine. The layers were separated and the organic layer was washed with an additional 50 mL brine, dried over anhydrous magnesium sulfate, filtered, and concentrated to dryness under reduced pressure. The residue was purified by silica gel chromatography (hexanes/ethyl acetate). The product containing fractions were combined, concentrated, and dried under reduced pressure to afford tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((tert - butyldimethylsilyl)oxy)propoxy)-2-methylphenyl)picolinate (530 mg, 693 µmol, 59.2% yield). MS (ESI) m/z: 765.3 [M+H]+. Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-hydroxypropoxy)-2-methy lphenyl)picolinate TBAF (217 mg, 831 µL, 1.0 molar, 1.2 equiv., 831 µmol) as a 1N solution was combined in THF with tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)- 3-(3-(3-((tert-butyldimethylsilyl)oxy)propoxy)-2-methylpheny l)picolinate (530 mg, 1 equiv., 693 µmol) dissolved in THF (10.0 mL). The reaction mixture was stirred at ambient temperature overnight. An additional portion of TBAF (217 mg, 831 µL, 1.0 molar, 1.2 equiv., 831 µmol) was added and the reaction was stirred again overnight at ambient temperature. The reaction mixture was then diluted with 125 mL ethyl acetate, 25 mL water, and 25 mL brine. The layers were separated and the organic layer was washed with an additional 50 mL brine, dried over anhydrous magnesium sulfate, filtered, and concentrated to dryness under reduced pressure. The residue was purified by silica gel (hexanes/ethyl acetate). The product containing fractions were combined, concentrated, triturated with a mixture of diethyl ether and hexanes, and re-concentrated to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-hydroxypropoxy)-2-m ethylphenyl)picolinate (322 mg, 495 µmol, 71.4% yield). MS (ESI) m/z: 651.2 [M+H]+. Step E. Procedure for preparation of tert-butyl 4-(2-((3-(2,6-bis(benzyloxy)pyridin-3-yl)- 1-methyl-1H-indazol-6-yl)oxy)ethyl)piperazine-1-carboxylate tert-Butyl 4-(2-hydroxyethyl)piperazine-1-carboxylate (920.5 mg, 2 equiv., 3.997 mmol) and 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-bromo-1-methyl-1H-indaz ole (1000 mg, 1 equiv., 1.998 mmol) were dissolved in dioxane (15 mL). To this solution tBuBrettPhos Pd G3 (85.38 mg, 0.05 equiv., 99.92 µmol) was added followed by potassium tert-butoxide (269.1 mg, 2.398 mL, 1.0 molar in THF, 1.2 equiv., 2.398 mmol). The reaction was then stirred at ambient temperature for about 60 hours. The reaction mixture was diluted with 125 mL ethyl acetate, 25 mL water, and 25 mL brine. The layers were separated and the organic layer was washed with an additional 50 mL brine, dried over anhydrous magnesium sulfate, filtered, and concentrated to dryness under reduced pressure. The residue was purified by silica gel chromatography (heptane/ethyl acetate containing 10% of a 7N solution of NH3 in MeOH). The concentrated product containing fractions afforded tert-butyl 4-(2-((3-(2,6-bis(benzyloxy)pyridin-3-yl)-1- methyl-1H-indazol-6-yl)oxy)ethyl)piperazine-1-carboxylate (821 mg, 1.26 mmol, 63.2% yield). MS (ESI) m/z: 650.3 [M+H]+. Step F. Procedure for preparation of tert-butyl 4-(2-((3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)oxy)ethyl)piperazine-1-carboxylate tert-Butyl 4-(2-((3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazo l-6- yl)oxy)ethyl)piperazine-1-carboxylate (821 mg, 1 equiv., 1.26 mmol) was dissolved in ethyl acetate (10 mL). Palladium on carbon (672 mg, 10% Wt, 0.25 equiv., 316 µmol) was added and the reaction was degassed under vacuum and then stirred at ambient temperature under an H 2 atmosphere overnight. The reaction was filtered through a pre-wetted pad of celite, concentrated to an oil under reduced pressure, and redissolved in ethyl acetate. Palladium on carbon (672 mg, 10% Wt, 0.25 eq, 316 µmol) was then added and the reaction was again degassed and stirred under a hydrogen atmosphere at ambient temperature for about 60 hours. The reaction was filtered through a pre-wetted pad of celite, and concentrated to an oil under reduced pressure to afford tert-butyl 4-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 6-yl)oxy)ethyl)piperazine-1-carboxylate (596 mg, 1.26 mmol, quantitative yield assumed). MS (ESI) m/z: 472.3 [M+H]+. Step G. Procedure for preparation of 3-(1-methyl-6-(2-(piperazin-1-yl)ethoxy)-1H- indazol-3-yl)piperidine-2,6-dione hydrochloride Tert-butyl 4-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)oxy)ethyl)piperazine-1-carboxylate (594 mg, 1 equiv., 914 µmol) was dissolved in DCM (50 mL), and 4N HCl in dioxane (667 mg, 4.57 mL, 4.0 molar, 20 equiv., 18.3 mmol) was added. The reaction mixture was stirred at ambient temperature overnight. The reaction was then concentrated under reduced pressure, and triturated in a mixture of diethyl ether and hexanes. The reconcentrated material afforded 3-(1-methyl-6-(2-(piperazin-1-yl)ethoxy)-1H- indazol-3-yl)piperidine-2,6-dione hydrochloride (306 mg, 750 µmol, 82.1% yield). MS (ESI) m/z: 372.3 [M+H]+. Step H. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-chloropropoxy)-2-methyl phenyl)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3- hydroxypropoxy)-2-methylphenyl)picolinate (322 mg, 1 equiv., 495 µmol) was dissolved in DCM (10 mL). Thionyl chloride (118 mg, 72.2 µL, 2 equiv., 990 µmol) was added and the reaction mixture was stirred overnight at ambient temperature. The reaction mixture was diluted with additional DCM and a saturated aqueous solution of sodium bicarbonate. The layers were separated, and the organic layer dried over anhydrous magnesium sulfate, filtered, and concentrated to dryness under vacuum. The residue was purified by silica gel chromatography with hexanes/ethyl acetate. The product containing fractions were combined, concentrated, triturated with a mixture of diethyl ether and hexanes, and reconcentrated to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- (3-(3-chloropropoxy)-2-methylphenyl)picolinate (163 mg, 244 µmol, 49.2% yield). MS (ESI) m/z: 669.1 [M+H]+. Step I. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(4-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)oxy)ethyl)piperazin-1-yl)propoxy)-2-methylpheny l)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3- chloropropoxy)-2-methylphenyl)picolinate (75 mg, 1 equiv., 0.11 mmol) and 3-(1-methyl-6- (2-(piperazin-1-yl)ethoxy)-1H-indazol-3-yl)piperidine-2,6-di one hydrochloride (62 mg, 1.5 equiv., 0.17 mmol) were dissolved in DMF (0.5 mL). DIEA (72 mg, 98 µL, 5 equiv., 0.56 mmol) was added and the reaction was stirred at 60°C overnight. An additional amount of DIEA (72 mg, 98 µL, 5 equiv., 0.56 mmol) was added and the reaction was stirred again overnight at 60 °C. The reaction was purified by reverse-phase HPLC (ACN/water, with 0.1% formic acid). The concentrated product containing fractions afforded tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(4-(2- ((3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)oxy)ethyl)piperazin-1-yl)propoxy)-2-m ethylphenyl)picolinate (110 mg, 0.11 mmol, quantitative yield assumed). MS (ESI) m/z: 1004.2 [M+H]+. Step J. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(4-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)oxy)ethyl)piperazin-1-yl)propoxy)-2-methylpheny l)picolinic acid tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4- (2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)ox y)ethyl)piperazin-1- yl)propoxy)-2-methylphenyl)picolinate (110 mg, 1 equiv., 110 µmol) was dissolved in a mixture of DCM (30 mL) and HCl in dioxane (2.00 g, 13.7 mL, 4.0 molar, 500 equiv., 54.8 mmol). The reaction was concentrated to dryness and the residue redissolved in DMSO. The material was purified by reverse-phase HPLC (ACN/water, with 0.1% formic acid). The product containing fractions were combined and lyophilized overnight to afford 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-(4-(2-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)oxy)ethyl)pipe razin-1-yl)propoxy)-2- methylphenyl)picolinic acid (10.2 mg, 10.4 µmol, 9.46 % yield over two steps). 1 H NMR was consistent with structure. MS (ESI) m/z: 948.2 [M+H]+. Example 218. Preparation of Compound 165

6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4-(3-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)oxy)propy l)-3- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linic acid Step A. Procedure for preparation of tert-butyl 4-allyl-3-(trifluoromethyl)piperazine-1- carboxylate tert-Butyl 3-(trifluoromethyl)piperazine-1-carboxylate (500 mg, 1 equiv., 1.97 mmol), allyl bromide (1.19 g, 851 µL, 5 equiv., 9.83 mmol), and DIEA (2.54 g, 3.43 mL, 10 equiv., 19.7 mmol) were combined in acetonitrile (8 mL) and heated overnight at 60 °C. Added 1/2 the amounts of bromide and DIEA as originally used and continued heating at 60 °C for an additional night. The reaction mixture was diluted with ethyl acetate (100 mL) and brine (25 mL). The layers were separated, and the organic layers were over anhydrous magnesium sulfate, filtered, and concentrated to an oil that was purified by silica gel chromatography (heptane/ethyl acetate gradient). The combined and concentrated product containing fractions afforded tert-butyl 4-allyl-3-(trifluoromethyl)piperazine-1-carboxylate (533 mg, 1.81 mmol, 92.1% yield). MS (ESI) m/z: 948.2 [M+H]+. Step B. Procedure for preparation of tert-butyl 4-(3-hydroxypropyl)-3- (trifluoromethyl)piperazine-1-carboxylate tert-Butyl 4-allyl-3-(trifluoromethyl)piperazine-1-carboxylate (400 mg, 1 equiv., 1.36 mmol) and 9-borabicyclo[3.3.1]nonane (199 mg, 3.26 mL, 0.5 molar, 1.2 equiv., 1.63 mmol) were combined in THF (10 mL) and stirred overnight at ambient temperature. Water (10 mL) was added to the reaction followed by sodium perborate tetrahydrate (627 mg, 3 equiv., 4.08 mmol) and the biphasic mixture was stirred for a few hours at ambient temperature. The reaction mixture was then diluted with ethyl acetate (125 mL), water (30 mL), and brine (30 mL). The layers were separated and the organic layer was dried over anhydrous magnesium sulfate, filtered and purified by silica gel chromatography (ethyl acetate / hexanes). The combined and concentrated product containing fractions afforded tert-butyl 4-(3-hydroxypropyl)-3- (trifluoromethyl)piperazine-1-carboxylate (371 mg, 1.19 mmol, 87.4% yield). 1 H NMR (400 MHz, DMSO) δ 4.37 (t, J = 5.2 Hz, 1H), 3.97 (s, 1H), 3.82 – 3.52 (m, 2H), 3.49 – 3.36 (m, 2H), 3.24 (s, 1H), 3.05 (d, J = 65.8 Hz, 1H), 2.85 – 2.60 (m, 4H), 1.57 (p, J = 6.6 Hz, 2H), 1.38 (s, 9H). Step C. Procedure for preparation of tert-butyl 4-(3-((3-(2,6-bis(benzyloxy)pyridin-3-yl)- 1-methyl-1H-indazol-6-yl)oxy)propyl)-3-(trifluoromethyl)pipe razine-1-carboxylate tert-Butyl 4-(3-hydroxypropyl)-3-(trifluoromethyl)piperazine-1-carboxyl ate (370 mg, 2 equiv., 1.43 mmol) and 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-bromo-1-methyl-1H-indaz ole (358 mg, 1 equiv., 716 µmol) were dissolved in dioxane (10 mL). To this mixture tBuBrettPhos Pd G3 (30.6 mg, 0.05 equiv., 35.8 µmol) was added followed by potassium tert-butoxide (96.4 mg, 859 µL, 1.0 molar, 1.2 equiv., 859 µmol) and the reaction was stirred at ambient temperature overnight. The reaction was then diluted with ethyl acetate and brine. The layers were separated and the aqueous layers extracted with additional ethyl acetate. The combined organic layers were dried over anhydrous magnesium sulfate, filtered, and purified by silica gel chromatography (ethyl acetate/heptane). The combined product containing fractions were dried under vacuum over the weekend to afford tert-butyl 4-(3-((3-(2,6-bis(benzyloxy)pyridin-3-yl)- 1-methyl-1H-indazol-6-yl)oxy)propyl)-3-(trifluoromethyl)pipe razine-1-carboxylate (524 mg, quantitative yield assumed). MS (ESI) m/z: 338.8 [M+2H/2]+. Step D. Procedure for preparation of tert-butyl 4-(3-((3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)oxy)propyl)-3-(trifluoromethyl)pipera zine-1-carboxylate tert-Butyl 4-(3-((3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazo l-6-yl)oxy)propyl)- 3-(trifluoromethyl)piperazine-1-carboxylate (433 mg, 1 equiv., 639 µmol) was dissolved in EtOAc (15 mL). To this solution Pd/C (340 mg, 10% Wt, 0.5 equiv., 319 µmol) was added. The resulting mixture was evacuated and backfilled with a balloon of hydrogen (3X). The mixture was stirred under a balloon of hydrogen at ambient temperature overnight. The reaction mixture was filtered through celite, and the filter cake was rinsed with ethyl acetate. The material was concentrated to dryness under reduced pressure and the residue purified by silica gel chromatography (ethyl acetate/hexanes) to afford tert-butyl 4-(3-((3-(2,6-dioxopiperidin-3-yl)- 1-methyl-1H-indazol-6-yl)oxy)propyl)-3-(trifluoromethyl)pipe razine-1-carboxylate (120 mg, 217 µmol, 33.9% yield). MS (ESI) m/z: 554.3 [M+H]+. Step E. Procedure for preparation of 3-(1-methyl-6-(3-(2-(trifluoromethyl)piperazin-1- yl)propoxy)-1H-indazol-3-yl)piperidine-2,6-dione hydrochloride tert-Butyl 4-(3-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl) oxy)propyl)-3- (trifluoromethyl)piperazine-1-carboxylate (120 mg, 1 equiv., 217 µmol) was dissolved in DCM (10 mL). To this solution, HCl in dioxane (158 mg, 1.08 mL, 4.0 molar, 20 equiv., 4.34 mmol) was added and the reaction was stirred overnight at ambient temperature. The reaction was concentrated under reduced pressure to solids that were triturated with diethyl ether/hexanes, re-concentrated, and dried overnight under vacuum to afford 3-(1-methyl-6-(3- (2-(trifluoromethyl)piperazin-1-yl)propoxy)-1H-indazol-3-yl) piperidine-2,6-dione hydrochloride (136 mg, quantitative yield assumed). MS (ESI) m/z: 454.1 [M+H]+. Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(4-(3-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)oxy)propyl)-3-(trifluoromethyl)piperazin-1-yl)p ropoxy)-2- methylphenyl)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3- chloropropoxy)-2-methylphenyl)picolinate (100 mg, 1.0 equiv., 149 µmol) and 3-(1-methyl-6- (3-(2-(trifluoromethyl)piperazin-1-yl)propoxy)-1H-indazol-3- yl)piperidine-2,6-dione hydrochloride (68 mg, 1.0 equiv., 0.15 mmol) were dissolved in NMP (1.0 mL). DIEA (97 mg, 0.13 mL, 5 equiv., 0.75 mmol) and sodium iodide (22 mg, 1 equiv., 0.15 mmol) were added and the reaction was stirred at 60°C overnight. The reaction was purified by reverse-phase HPLC (ACN/water, with 0.1% formic acid). Product containing fractions were lyophilized to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4- (3-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)ox y)propyl)-3- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linate (16 mg, 15 µmol, 9.8% yield). MS (ESI) m/z: 544.0 [M+2H/2]+. Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(4-(3-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)oxy)propyl)-3-(trifluoromethyl)piperazin-1-yl)p ropoxy)-2- methylphenyl)picolinic acid tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4- (3-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)ox y)propyl)-3- (trifluoromethyl)piperazin-1-yl)propoxy)-2-methylphenyl)pico linate (16 mg, 1 equiv., 15 µmol) was dissolved in DCM (5.0 mL). Trifluoroacetic acid (1.7 g, 1.1 mL, 1000 equiv., 15 mmol) was added and the reaction was stirred at ambient temperature overnight. The reaction mixture was concentrated to dryness under reduced pressure and the resulting residue was purified by reverse-phase HPLC (ACN/water, with 0.1% formic acid). The product containing fractions were combined and lyophilized to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(4-(3-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)oxy)propyl)-3-(trifluoromethyl)piperazin-1-yl)p ropoxy)-2- methylphenyl)picolinic acid (16.8 mg, 14.7 µmol, 100% yield). 1 H NMR was consistent with structure. MS (ESI) m/z: 1030.4 [M+H]+. Example 219. Preparation of Compound 172 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(3-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)oxy)propy l)piperidin-4-yl)propoxy)-2- methylphenyl)picolinic acid Step A. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-(3-((tert- butyldimethylsilyl)oxy)propoxy)-1-methyl-1H-indazole Dissolved 3-((tert-butyldimethylsilyl)oxy)propan-1-ol (100 mg, 2 equiv., 525 µmol) and 3- (2,6-bis(benzyloxy)pyridin-3-yl)-6-bromo-1-methyl-1H-indazol e (131 mg, 1 equiv., 263 µmol) in dioxane (4 mL). To this mixture was added tBuBrettPhos Pd G3 (11.2 mg, 0.05 equiv., 13.1 µmol) followed by potassium tert-butoxide (35.4 mg, 315 µL, 1.0 M in THF, 1.2 equiv., 315 µmol) and the reaction was allowed to mix at ambient temperature overnight. The reaction mixture was diluted with ethyl acetate and brine. The layers were separated and the extracted aqueous layers were washed with additional ethyl acetate. The combined organic layers were dried over anhydrous magnesium sulfate, filtered, and purified by silica gel chromatography (ethyl acetate/heptane). Product containing fractions were combined to afford 3-(2,6- bis(benzyloxy)pyridin-3-yl)-6-(3-((tert-butyldimethylsilyl)o xy)propoxy)-1-methyl-1H- indazole (160 mg, quantitative yield assumed). MS (ESI) m/z: 610.4 [M+H]+. Step B. Procedure for preparation of 3-(6-(3-((tert-butyldimethylsilyl)oxy)propoxy)-1- methyl-1H-indazol-3-yl)piperidine-2,6-dione 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-(3-((tert-butyldimethyl silyl)oxy)propoxy)-1-methyl- 1H-indazole (30 mg, 1 equiv., 49 µmol) was dissolved in ethyl acetate (10 mL). Palladium on carbon (52 mg, 10% Wt, 1 equiv., 49 µmol) was added and the reaction mixture was evacuated and back-filled with hydrogen (balloon). This process was repeated twice the mixture was stirred overnight at ambient temperature under a hydrogen atmosphere. The reaction mixture was then filtered through a bed of celite and concentrated to afford 3-(6-(3-((tert- butyldimethylsilyl)oxy)propoxy)-1-methyl-1H-indazol-3-yl)pip eridine-2,6-dione (21 mg, quantitative yield assumed) that was used without further purification. MS (ESI) m/z: 432.3 [M+H]+. Step C. Procedure for preparation of 3-(6-(3-hydroxypropoxy)-1-methyl-1H-indazol-3- yl)piperidine-2,6-dione Dissolved 3-(6-(3-((tert-butyldimethylsilyl)oxy)propoxy)-1-methyl-1H-i ndazol-3- yl)piperidine-2,6-dione (363 mg, 1 equiv., 841 µmol) in THF (10 mL). Added TBAF (264 mg, 1.01 mL, 1.0 M in THF, 1.2 equiv., 1.01 mmol) and reaction was stirred overnight at ambient temperature. The reaction mixture was then concentrated to dryness under reduce pressure and purified by reverse-phase HPLC (ACN/water, with 0.1% formic acid). Pure product containing fractions were combined and concentrated under reduced pressure to afford 3-(6-(3- hydroxypropoxy)-1-methyl-1H-indazol-3-yl)piperidine-2,6-dion e (115 mg, 0.36 mmol, 43.1 % yield). MS (ESI) m/z: 318.0 [M+H]+. Step D. Procedure for preparation of 3-(6-(3-chloropropoxy)-1-methyl-1H-indazol-3- yl)piperidine-2,6-dione Dissolved 3-(6-(3-hydroxypropoxy)-1-methyl-1H-indazol-3-yl)piperidine- 2,6-dione (143 mg, 1 equiv., 451 µmol) in DCM (10 mL). Added thionyl chloride (107 mg, 65.8 µL, 2 equiv., 901 µmol) and stirred reaction overnight at ambient temperature. An additional equivalent of thionyl chloride was added and the reaction was stirred overnight at ambient temperature. The reaction mixture was diluted with additional DCM and a saturated aqueous solution of sodium bicarbonate. The layers were separated , and the organic layer was dried over anhydrous magnesium sulfate, filtered, and concentrated to dryness under reduced pressure to afford 3- (6-(3-chloropropoxy)-1-methyl-1H-indazol-3-yl)piperidine-2,6 -dione (143 mg, 451 µmol, 39.7 % yield). MS (ESI) m/z: 336.1 [M+H]+. Step E. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(3-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)oxy)propyl)piperidin-4-yl)propoxy)-2-methylphen yl)picolinate Dissolved 3-(6-(3-chloropropoxy)-1-methyl-1H-indazol-3-yl)piperidine-2 ,6-dione (30 mg, 1 equiv., 89 µmol) and tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2-methyl-3-(3-(piperidin-4-yl)propoxy)phenyl)pi colinate (64 mg, 1 equiv., 89 µmol) in NMP (1 mL). Added DIEA (58 mg, 78 µL, 5 equiv., 0.45 mmol) and sodium iodide (13 mg, 1 equiv., 89 µmol) to the reaction mixture and reaction was stirred overnight at 60 °C. The reaction mixture was purified by reversed-phase HPLC (ACN/water, with 0.1% formic acid. The combined and concentrated product containing fractions afforded tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-(1-(3-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)oxy)propyl)pip eridin-4-yl)propoxy)-2- methylphenyl)picolinate (91 mg, quantitative yield assumed). MS (ESI) m/z: 1017.5 [M+H]+. Step F. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(3-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)oxy)propyl)piperidin-4-yl)propoxy)-2-methylphen yl)picolinic acid Dissolved tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)- 3-(3-(3-(1-(3-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-inda zol-6-yl)oxy)propyl)piperidin- 4-yl)propoxy)-2-methylphenyl)picolinate (90 mg, 1 equiv., 88 µmol) in DCM (20 mL). Added a 4.0 M solution of hydrochloric acid in dioxane (0.16 g, 1.1 mL, 4.0 molar, 50 equiv., 4.4 mmol) and stirred reaction overnight at ambient temperature. The reaction was concentrated to dryness under reduced pressure, redissolved in a mixture of ACN and water, and lyophilized material to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- (3-(3-(1-(3-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazo l-6-yl)oxy)propyl)piperidin-4- yl)propoxy)-2-methylphenyl)picolinic acid (10.6 mg, 10.7 µmol, 12 % yield). 1 H NMR (400 MHz, DMSO-d6) δ 12.87 (s, 1H), 10.87 (s, 1H), 9.49 (s, 1H), 8.04 (d, J = 7.8 Hz, 1H), 7.80 (d, J = 7.9 Hz, 1H), 7.61 (dd, J = 17.5, 8.1 Hz, 2H), 7.52 – 7.42 (m, 4H), 7.41 – 7.32 (m, 2H), 7.19 – 7.05 (m, 2H), 6.98 (d, J = 8.8 Hz, 1H), 6.89 (d, J = 8.2 Hz, 1H), 6.82 – 6.70 (m, 1H), 6.64 (d, J = 7.6 Hz, 1H), 4.99 (s, 2H), 4.31 (dd, J = 9.3, 5.0 Hz, 1H), 4.23 – 4.08 (m, 2H), 4.03 – 3.86 (m, 7H), 3.22 (s, 2H), 2.94 (t, J = 11.6 Hz, 2H), 2.77 – 2.58 (m, 2H), 2.26 (d, J = 55.2 Hz, 4H), 1.83 (d, J = 58.1 Hz, 10H), 1.57 (s, 1H), 1.54 – 1.29 (m, 5H), 1.24 (d, J = 4.6 Hz, 3H). MS (ESI) m/z: 961.4 [M+H]+. Example 220. Preparation of Compound 189 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4-((1-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperidin -4-yl)methyl)piperazin-1- yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(4-((1-(3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)piperidin-4-yl)methyl)piperazin-1-yl)propoxy)-2 -methylphenyl)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3- chloropropoxy)-2-methylphenyl)picolinate (83 mg, 1 equiv., 0.12 mmol) was dissolved in NMP (1 mL). To the mixture was added 3-(1-methyl-6-(4-(piperazin-1-ylmethyl)piperidin-1- yl)-1H-indazol-3-yl)piperidine-2,6-dione hydrochloride (57 mg, 1 equiv., 0.12 mmol) and sodium iodide (19 mg, 1 equiv., 0.12 mmol), followed by DIEA (80 mg, 0.11 mL, 5 equiv., 0.62 mmol). The mixture was then heated at 60 °C overnight. The reaction mixture was purified by reverse-phase HPLC (ACN/water, with 0.1% formic acid). Product containing fractions were combined and lyophilized to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(4-((1-(3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)piperidin-4-yl)methyl)piperazin-1-yl)propoxy)-2 -methylphenyl)picolinate (40 mg, 0.038 mmol, 31% yield). MS (ESI) m/z: 529.5 [M+2H/2]+. Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(4-((1-(3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)piperidin-4-yl)methyl)piperazin-1-yl)propoxy)-2 -methylphenyl)picolinic acid tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4- ((1-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pi peridin-4-yl)methyl)piperazin- 1-yl)propoxy)-2-methylphenyl)picolinate (40 mg, 1 equiv., 38 µmol) was dissolved in DCM (2.4 mL). To the mixture was added 0.6 mL TFA and the reaction mixture was then stirred at ambient temperature for two nights. The reaction mixture was concentrated under reduced pressure and the residue was resuspended in ACN/water and lyophilized to afford 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-(4-((1-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperidin-4-yl )methyl)piperazin-1- yl)propoxy)-2-methylphenyl)picolinic acid ditrifluoroacetate (51 mg, 41 µmol, quantitative yield). 1 H NMR was consistent with structure. MS (ESI) m/z: 1000.7 [M+H]+. Example 221. Preparation of Compound 192 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-(1-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperidin -4-yl)ethyl)piperidin-4- yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of 6-(4-(2-(benzyloxy)ethyl)piperidin-1-yl)-3-(2,6- bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazole 3-(2,6-Bis(benzyloxy)pyridin-3-yl)-6-bromo-1-methyl-1H-indaz ole (1.0 g, 1 equiv., 2.0 mmol) was suspended in 1,4-dioxane (15 mL). To the mixture was added 4-(2- (benzyloxy)ethyl)piperidine hydrochloride (0.61 g, 1.2 equiv., 2.4 mmol), followed by (2- dicyclohexylphosphino-2',6'-diisopropoxy-1,1'-biphenyl)[2-(2 '-amino-1,1'- biphenyl)]palladium(II) methanesulfonate (RuPhos Pd G3, 0.17 g, 0.1 eq, 0.20 mmol), and sodium 2-methylpropan-2-olate (0.58 g, 3.0 mL, 2.0 M in THF, 3 equiv., 6.0 mmol). The mixture was then purged with N 2 and heated at 100 °C for 2 h. The mixture was cooled and then diluted with ethyl acetate and water. The organic layer was separated, washed with brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to dryness. The residue was purified by RP-HPLC. The desired fractions were combined and concentrated under reduced pressure to afford 6-(4-(2-(benzyloxy)ethyl)piperidin-1-yl)-3-(2,6- bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazole (0.65 g, 1.0 mmol, 51 % yield). MS (ESI) m/z: 661.5 [M+Na]+. Step B. Procedure for preparation of 3-(6-(4-(2-hydroxyethyl)piperidin-1-yl)-1-methyl- 1H-indazol-3-yl)piperidine-2,6-dione 6-(4-(2-(Benzyloxy)ethyl)piperidin-1-yl)-3-(2,6-bis(benzylox y)pyridin-3-yl)-1-methyl-1H- indazole (650 mg, 1 equiv., 1.02 mmol) was dissolved in ethyl acetate (15 mL). To the mixture was added Pd/C (541 mg, 10% Wt, 0.5 equiv., 509 µmol). The mixture was evacuated and backfilled with H2 (3X). The mixture was then stirred at ambient temperature under a H2 atmosphere (balloon pressure). The reaction mixture was filtered and the filter cake was washed with ethyl acetate. The filtrate was concentrated under reduced pressure and the resulting crude material was then purified by reverse-phase HPLC (ACN/water, with 0.1% formic acid) to afford 3-(6-(4-(2-hydroxyethyl)piperidin-1-yl)-1-methyl-1H-indazol- 3-yl)piperidine-2,6- dione (101 mg, 273 µmol, 26.8 %). NMR was consistent with structure. MS (ESI) m/z: 371.3 [M+H]+. Step C. Procedure for preparation of 2-(1-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)piperidin-4-yl)acetaldehyde 3-(6-(4-(2-Hydroxyethyl)piperidin-1-yl)-1-methyl-1H-indazol- 3-yl)piperidine-2,6-dione (40 mg, 1 equiv., 0.11 mmol) was dissolved in DCM (4 mL). To the mixture was added Dess-Martin periodinane (50 mg, 37 µL, 1.1 equiv., 0.12 mmol). The mixture was then stirred at ambient temperature for ~1 h. A few drops methanol was added to the reaction mixture and it was then concentrated under reduced pressure to afford 2-(1-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)piperidin-4-yl)acetaldehyde (40 mg, quantitative yield assumed) as a crude material that was used without further purification. MS (ESI) m/z: 369.3 [M+H]+. Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-(1-(3-(2,6-dioxop iperidin-3-yl)-1-methyl-1H- indazol-6-yl)piperidin-4-yl)ethyl)piperidin-4-yl)propoxy)-2- methylphenyl)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(3-(piperidin-4-yl)propoxy)phenyl)picolinate (60 mg, 1 equiv., 84 µmol) and 2-(1- (3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperi din-4-yl)acetaldehyde (40 mg, 1.3 equiv., 0.11 mmol) were suspended in ethanol (8 mL). To the mixture was added sodium cyanoborohydride (26 mg, 5 equiv., 0.42 mmol), followed by acetic acid (15 mg, 14 µL, 3 equiv., 0.25 mmol). The mixture was then stirred at ambient temperature overnight. The mixture was evaporated under reduced pressure and the residue was then purified by reverse- phase HPLC (ACN/water, with 0.1% formic acid) to afford tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2- (1-(3-(2,6-dioxopiperidin-3-yl)- 1-methyl-1H-indazol-6-yl)piperidin-4-yl)ethyl)piperidin-4-yl )propoxy)-2- methylphenyl)picolinate (51 mg, 48 µmol, 57 % yield). MS (ESI) m/z: 536.0 [M+2H/2]+. Step E. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-(1-(3-(2,6-dioxop iperidin-3-yl)-1-methyl-1H- indazol-6-yl)piperidin-4-yl)ethyl)piperidin-4-yl)propoxy)-2- methylphenyl)picolinic acid tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1- (2-(1-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl) piperidin-4-yl)ethyl)piperidin- 4-yl)propoxy)-2-methylphenyl)picolinate (30 mg, 1 equiv., 28 µmol) was dissolved in DCM (2.4 mL). To the mixture was added 0.6 mL TFA. The mixture was then stirred at ambient temperature for two nights. The solvents were removed under reduced pressure and the residue was purified by reverse-phase HPLC (ACN/water, with 0.1% formic acid). The desired fractions were combined and concentrated under reduced pressure to afford 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-(1-(2-(1-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperidin-4-yl )ethyl)piperidin-4-yl)propoxy)- 2-methylphenyl)picolinic acid (24 mg, 24 µmol, 84 % yield). 1 H NMR was consistent with structure. MS (ESI) m/z: 1014.8 [M+H]+. Example 222. Preparation of Compound 198 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(4-((1-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperidin -4-yl)methyl)piperazin-1- yl)ethoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-((1-(3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)piperidin-4-yl)methyl)piperazin-1-yl)ethoxy)-2- methylphenyl)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2- chloroethoxy)-2-methylphenyl)picolinate (100 mg, 1 equiv., 153 µmol), 3-(1-methyl-6-(4- (piperazin-1-ylmethyl)piperidin-1-yl)-1H-indazol-3-yl)piperi dine-2,6-dione hydrochloride (70.4 mg, 1 equiv., 153 µmol), and sodium iodide (27.5 mg, 1.2 equiv., 183 µmol) were combined. To this mixture was added DIEA (98.6 mg, 133 µL, 5 equiv., 763 µmol). The resulting mixture was heated at 60 °C overnight, cooled, and then purified directly by reverse- phase HPLC (ACN/water, with 0.1% formic acid) to afford tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-((1 -(3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperidin-4-yl)methyl)piperazin-1-yl) ethoxy)-2- methylphenyl)picolinate (33 mg, 32 µmol, 21% yield). MS (ESI) m/z: 522.5 [M+2H/2]+. Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-((1-(3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)piperidin-4-yl)methyl)piperazin-1-yl)ethoxy)-2- methylphenyl)picolinic acid tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(4- ((1-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pi peridin-4-yl)methyl)piperazin- 1-yl)ethoxy)-2-methylphenyl)picolinate (33 mg, 1 equiv., 32 µmol)) was dissolved in DCM (2.4 mL). To the mixture was added 0.6 mL TFA. The mixture was then stirred at ambient temperature for two nights. The reaction was concentrated under reduced pressure, resuspended in ACN/water, and lyophilized overnight to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-((1-(3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)piperidin-4-yl)methyl)piperazin-1-yl)ethoxy)-2- methylphenyl)picolinic acid difluoroacetate (40 mg, 33 µmol, quantitative yield). NMR was consistent with structure. MS (ESI) m/z: 987.6 [M+H]+. Example 223. Preparation of Compound 203 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(4-(1-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2- oxoethyl)piperidin-4- yl)butoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(1-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)butoxy)-2-meth ylphenyl)picolinate To a solution of 2-(4-(4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)butyl)piperidin-1-yl)acetic acid (60 mg, 1 equiv., 76 µmol) in DCM (5 mL) was added HATU (32 mg, 1.1 equiv., 84 µmol), followed by 3-(6-amino-1-methyl-1H-indazol-3-yl)piperidine-2,6-dione (22 mg, 1.1 equiv., 84 µmol) and diisopropylethylamine (29 mg, 40 µL, 3 equiv., 0.23 mmol). The reaction solution was then stirred at room temperature. After 16 h, the reaction solution was filtered and concentrated to provide crude tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(1-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)butoxy)-2-meth ylphenyl)picolinate, which was carried forward without further purification. MS (ESI) m/z: 1029.6 [M+H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(1-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)butoxy)-2-meth ylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(4-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-meth yl-1H-indazol-6-yl)amino)-2- oxoethyl)piperidin-4-yl)butoxy)-2-methylphenyl)picolinate (78 mg, 76 µmol, 1 equiv.) in DCM (4 mL) was added trifluoroacetic acid (1 mL), and the reaction solution was stirred at room temperature. After 16 h, the reaction solution was concentrated, dissolved in DMSO, and purified by reverse-phase HPLC to provide 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(1-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)butoxy)-2-meth ylphenyl)picolinic acid (30 mg, 31 µmol, 41% yield) as a white solid. MS (ESI) m/z: 974 [M+H] + . 1 H NMR is consistent with structure. Example 224. Preparation of Compound 236 6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(3-(1-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino)-2- oxoethyl)piperidin-4- yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(3-(1-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-7-yl)amino)-2-oxoethyl)piperidin-4-yl)propoxy)-2-met hylphenyl)picolinate A mixture of 2-(4-(3-(4-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-3-methylpheno xy)propyl)piperidin-1- yl)acetic acid (37 mg, 1 equiv, 48 µmol), 3-(7-amino-1-methyl-1H-indazol-3-yl)piperidine- 2,6-dione (15 mg, 1.2 equiv, 57 µmol), N,N,N’,N’-tetramethylchloroformamidinium- hexafluorophosphate (16 mg, 1.2 equiv, 57 µmol) and 1-methylimidazole (14 mg, 13 µL, 3.5 equiv., 0.17 mmol) was suspended in MeCN (5 mL) and the reaction solution was stirred at room temperature. After 16 h, the reaction solution was concentrated, re-suspended in ethyl acetate and water, and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated to provide tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(4-(3-(1-(2-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino)-2-oxoet hyl)piperidin-4-yl)propoxy)- 2-methylphenyl)picolinate, which was carried forward without further purification. MS (ESI) m/z: 1016.6 [M+H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(3-(1-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-7-yl)amino)-2-oxoethyl)piperidin-4-yl)propoxy)-2-met hylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(3-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-meth yl-1H-indazol-7-yl)amino)-2- oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinate (49 mg, 48 µmol, 1 equiv.) in DCM (4 mL) was added 1 mL trifluoroacetic acid and the reaction solution was stirred at room temperature. After 12 h, the reaction solution was concentrated, the material re-suspended in DMSO, and the material purified by reverse-phase HPLC to provide 6-(8-(benzo[d]thiazol- 2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(4-(3-(1-( 2-((3-(2,6-dioxopiperidin-3-yl)- 1-methyl-1H-indazol-7-yl)amino)-2-oxoethyl)piperidin-4-yl)pr opoxy)-2- methylphenyl)picolinic acid. (21 mg, 22 µmol, 46 %) as a white solid. MS (ESI) m/z: 960.5 [M+H] + . consistent with structure. Example 225. Preparation of Compound 223 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(1-(2-(1-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperidin -4-yl)ethyl)piperidin-4- yl)ethoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(1-(2-(1-(3-(2,6-dioxop iperidin-3-yl)-1-methyl-1H- indazol-6-yl)piperidin-4-yl)ethyl)piperidin-4-yl)ethoxy)-2-m ethylphenyl)picolinate To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2-methyl-3-(2-(piperidin-4-yl)ethoxy)phenyl)pic olinate (77 mg, 1 equiv., 0.11 mmol) in DCM (5 mL) was added 2-(1-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)piperidin-4-yl)acetaldehyde (41 mg, 1 equiv., 0.11 mmol), followed by sodium cyanoborohydride (35 mg, 32 µL, 5 equiv., 0.55 mmol) and acetic acid (20 mg, 19 µL, 3 equiv., 0.33 mmol). The reaction solution was stirred at room temperature. After 14 h, the reaction solution was concentrated, re-suspended in DMSO, and purified by reverse-phase HPLC to provide tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- (3-(2-(1-(2-(1-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-inda zol-6-yl)piperidin-4- yl)ethyl)piperidin-4-yl)ethoxy)-2-methylphenyl)picolinate (40 mg, 38 µmol, 34% yield) as an off-white solid. MS (ESI) m/z: 529.0 [M+H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(1-(2-(1-(3-(2,6-dioxop iperidin-3-yl)-1-methyl-1H- indazol-6-yl)piperidin-4-yl)ethyl)piperidin-4-yl)ethoxy)-2-m ethylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(1-(2-(1-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)piperidin- 4-yl)ethyl)piperidin-4-yl)ethoxy)-2-methylphenyl)picolinate (10 mg, 1 equiv., 9.5 µmol) in DCM (4 mL) was added trifluoroacetic acid (1.1 mg, 0.73 µL, 1 equiv., 9.5 µmol) and the reaction solution was stirred at room temperature. After 16 h, the reaction solution was concentrated, re-suspended in DMSO, and purified by reverse-phase HPLC to provide 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(2-(1-(2-(1-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperidin-4-yl )ethyl)piperidin-4-yl)ethoxy)-2- methylphenyl)picolinic acid (6 mg, 6 µmol, 60% yield). MS (ESI) m/z: 501.3 [2M+H] + ; 1 H NMR (400 MHz, DMSO) δ 10.76 (s, 1H), 7.94 (d, J = 8.1 Hz, 1H), 7.70 (d, J = 8.1 Hz, 1H), 7.56 (d, J = 7.4 Hz, 1H), 7.33 (ddd, J = 35.0, 14.7, 7.8 Hz, 7H), 7.00 (t, J = 7.8 Hz, 1H), 6.82 (d, J = 7.8 Hz, 3H), 6.74 (s, 1H), 6.58 (d, J = 7.6 Hz, 1H), 6.47 (s, 1H), 4.91 (s, 2H), 4.17 (dd, J = 9.2, 5.1 Hz, 2H), 3.92 (s, 2H), 3.82 (d, J = 12.4 Hz, 5H), 3.68 (d, J = 12.2 Hz, 2H), 2.95 (s, 2H), 2.81 (d, J = 10.8 Hz, 3H), 2.64 (d, J = 11.9 Hz, 3H), 2.09 (s, 1H), 1.83 (d, J = 5.1 Hz, 5H), 1.69 (d, J = 12.8 Hz, 3H), 1.61 (s, 3H), 1.41 (s, 1H), 1.33 (d, J = 16.1 Hz, 3H), 1.17 (s, 3H). Example 226. Preparation of Compound 221 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4-((1-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperidin -4-yl)methyl)piperazin-1- yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(4-((1-(3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-7-yl)piperidin-4-yl)methyl)piperazin-1-yl)propoxy)-2 -methylphenyl)picolinate To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-chloropropoxy)-2-methylphenyl)picolinate (100 mg, 1 equiv., 149 µmol) in NMP (1 mL) was added 3-(1-methyl-7-(4-(piperazin-1-ylmethyl)piperidin-1-yl)-1H- indazol-3-yl)piperidine-2,6-dione (63.4 mg, 1 equiv., 149 µmol), followed by sodium iodide (26.9 mg, 1.2 equiv., 179 µmol) and diisopropylethylamine (96.6 mg, 130 µL, 5 equiv., 747 µmol) and the reaction solution was stirred at 60 ºC. After 48 h, the reaction solution was concentrated, re-suspended in DMSO, and purified by reverse-phase HPLC to provide tert- butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4-((1- (3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperi din-4-yl)methyl)piperazin-1- yl)propoxy)-2-methylphenyl)picolinate (45 mg, 43 µmol, 28% yield) as a white solid. MS (ESI) m/z: 529.5 [2M+H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(4-((1-(3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-7-yl)piperidin-4-yl)methyl)piperazin-1-yl)propoxy)-2 -methylphenyl)picolinic acid. To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4-((1-(3-(2,6-dioxopiperidin-3-yl)-1-meth yl-1H-indazol-7-yl)piperidin-4- yl)methyl)piperazin-1-yl)propoxy)-2-methylphenyl)picolinate (45 mg, 1 equiv, 43 µmol) in DCM (2.4 mL) was added 0.6 mL TFA and the reaction solution was stirred at room temperature. After 40 h, the reaction solution was concentrated, re-suspended in water/acetonitrile, and lyophilized to provide 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(4-((1-(3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-7-yl)piperidin-4-yl)methyl)piperazin-1-yl)propoxy)-2 -methylphenyl)picolinic acid trifluoroacetate (55 mg, 45 µmol, 110 %) as an off-white solid. MS (ESI) m/z: 501.5 [M+H] + ; Example 227. Preparation of Compound 228 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(4-((1-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperidin -4-yl)methyl)piperazin-1- yl)ethoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-((1-(3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-7-yl)piperidin-4-yl)methyl)piperazin-1-yl)ethoxy)-2- methylphenyl)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2- chloroethoxy)-2-methylphenyl)picolinate (100 mg, 1 equiv., 153 µmol) was suspended in NMP (1 mL). To the mixture was added 3-(1-methyl-7-(4-(piperazin-1-ylmethyl)piperidin- 1-yl)-1H-indazol-3-yl)piperidine-2,6-dione (64.8 mg, 1 equiv., 153 µmol), followed by sodium iodide (27.5 mg, 1.2 equiv, 183 µmol) and DIEA (98.6 mg, 133 µL, 5 equiv., 763 µmol). The mixture was then purged with nitrogen and heated at 60 ºC. After 48 h, the reaction solution was concentrated, re-suspended in DMSO, and purified by reverse-phase HPLC to provide tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(4- ((1-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pi peridin-4-yl)methyl)piperazin- 1-yl)ethoxy)-2-methylphenyl)picolinate (41 mg, 39 µmol, 26% yield). MS (ESI) m/z: 522.5 [2M+H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(2-(4-((1-(3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-7-yl)piperidin-4-yl)methyl)piperazin-1-yl)ethoxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(2-(4-((1-(3-(2,6-dioxopiperidin-3-yl)-1-meth yl-1H-indazol-7-yl)piperidin-4- yl)methyl)piperazin-1-yl)ethoxy)-2-methylphenyl)picolinate (41 mg, 1 equiv., 39 µmol) in DCM (2.4 mL) was added 0.6 mL TFA and the reaction solution was stirred at room temperature. After 40 h, the reaction solution was concentrated to provide 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(2-(4-((1-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperidin-4-yl )methyl)piperazin-1-yl)ethoxy)- 2-methylphenyl)picolinic acid, trifluoroacetate (46 mg, 38 µmol, 96% yield) as a light brown solid. MS (ESI) m/z: 987.6 [M+H] + . 1 H NMR is consistent with structure. Example 228. Preparation of Compound 260 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4-((2-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)-2-azaspi ro[3.5]nonan-7- yl)methyl)piperazin-1-yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of (2-Azaspiro[3.5]nonan-7-yl)methanol To a stirred solution of tert-butyl 7-(hydroxymethyl)-2-azaspiro[3.5]nonane-2-carboxylate (8.70 g, 34.07 mmol) in DCM (87.0 mL) was added 4M hydrochloric acid in dioxane (43.5 mL) at 0 °C and the resulting reaction mixture was stirred at room temperature. After 2 h, the reaction mixture was concentrated, and the crude solids were triturated with petroleum ether to afford (2-azaspiro[3.5]nonan-7-yl)methanol (6.07 g, 34.07 mmol), which was carried forward without further purification. 1 H NMR (DMSO-D6) δ 3.62-3.54 (m, 4H), 3.18 (d, J = 6.00 Hz, 2H), 2.00-1.97 (m, 2H), 1.62-1.58 (m, 2H), 1.39-1.29 (m, 3H), 0.89-0.75 (m, 2H). Step B. Procedure for preparation of (2-(3-(2,6-Bis(benzyloxy)pyridin-3-yl)-1-methyl-1H- indazol-7-yl)-2-azaspiro[3.5]nonan-7-yl)methanol To a stirred solution of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-7-bromo-1-methyl-1H-indaz ole (5.0 g, 9.99 mmol) and (2-azaspiro[3.5]nonan-7-yl)methanol (3.10 g, 19.98 mmol) in 1,4- dioxane (100 mL), was added cesium carbonate (9.76 g, 29.97 mmol) and the solution was degassed with argon for 20 minutes. Pd-PEPPSI-IHeptCl (0.49 g, 0.49 mmol) was added and the resulting reaction mixture was stirred at 100 °C. After 16 h, the reaction mixture was cooled to room temperature, filtered through celite and concentrated. The resulting crude material was purified by silica gel column chromatography (40% ethyl acetate in petroleum ether) to provide (2-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-7 -yl)-2-azaspiro[3.5]nonan-7- yl)methanol (4.05 g, 71 mmol, 71%) as a pale yellow solid. MS (ESI) m/z: 575.70 [M+H] + ; 1 H NMR; 400 MHz, DMSO-d6: δ 7.84 (d, J = 8.00 Hz, 1H), 7.48-7.26 (m, 10H), 7.15 (d, J = 8.00 Hz, 1H), 6.90 (t, J = 7.60 Hz, 1H), 6.72 (d, J = 7.20 Hz, 1H), 6.57 (d, J = 8.00 Hz, 1H), 5.42 (d, J = 6.00 Hz, 4H), 4.39 (t, J = 5.20 Hz, 1H), 4.26 (s, 3H), 3.59 (d, J = 20.00 Hz, 4H), 3.23 (t, J = 5.60 Hz, 2H), 1.98 (d, J = 13.20 Hz, 2H), 1.67 (d, J = 10.80 Hz, 2H), 1.52-1.35 (m, 3H), 1.00 (q, J = 11.60 Hz, 2H). Step C. Procedure for preparation of (2-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H- indazol-7-yl)-2-azaspiro[3.5]nonan-7-yl)methyl methanesulfonate To a stirred solution of (2-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-7 -yl)-2- azaspiro[3.5]nonan-7-yl)methanol (4.0 g, 6.95 mmol) in DCM (40 mL) was added triethylamine (2.9 mL, 20.87 mmol) followed by methanesulfonyl chloride (0.64 mL, 8.35 mmol) and the resulting reaction mixture was stirred at room temperature. After 1 h, the reaction solution was diluted with saturated aqueous sodium bicarbonate (20 mL) and extracted with dichloromethane (2 x 30 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated to provide (2-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1- methyl-1H-indazol-7-yl)-2-azaspiro[3.5]nonan-7-yl)methyl methanesulfonate (4.31 g, 95% yield) as a pale yellow solid, which was carried forward without further purification. MS (ESI) m/z: 653.6 [M+H] + . Step D. Procedure for preparation of tert-Butyl 4-((2-(3-(2,6-bis(benzyloxy)pyridin-3-yl)- 1-methyl-1H-indazol-7-yl)-2-azaspiro[3.5]nonan-7-yl)methyl)p iperazine-1-carboxylate To a stirred solution of (2-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-7 -yl)-2- azaspiro[3.5]nonan-7-yl)methyl methanesulfonate (4.30 g, 6.58 mmol) and tert-butyl piperazine-1-carboxylate (4.90 g, 26.34 mmol) in DMSO (43.0 mL) was added N,N- diisopropylethylamine (3.44 mL, 19.76 mmol) and the reaction mixture was stirred at 100 °C. After 16 h, the reaction solution was cooled to room temperature and diluted with cold water to precipitate the product out of solution. The resulting solids were collected by filtration and purified by silica gel column chromatography (30-40% EtOAc in petroleum ether) to provide tert-butyl 4-((2-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazo l-7-yl)-2- azaspiro[3.5]nonan-7-yl)methyl)piperazine-1-carboxylate (2.84 g, 3.81 mmol, 58%) yield as an off-white solid. MS (ESI) m/z: 743.8 [M+H] + ; 1 H NMR; 400 MHz, DMSO-d6: δ 7.84 (d, J = 8.00 Hz, 1H), 7.47 (d, J = 7.20 Hz, 2H), 7.41-7.26 (m, 8H), 7.15 (d, J = 8.00 Hz, 1H), 6.90 (t, J = 7.60 Hz, 1H), 6.71 (d, J = 7.20 Hz, 1H), 6.57 (d, J = 8.00 Hz, 1H), 5.42 (d, J = 6.00 Hz, 4H), 4.25 (s, 3H), 3.58 (d, J = 19.60 Hz, 4H), 3.32-3.30 (m, 3H), 2.27-2.26 (m, 4H), 2.09 (d, J = 7.20 Hz, 2H), 1.96 (d, J = 12.80 Hz, 2H), 1.70 (d, J = 11.20 Hz, 2H), 1.49 (t, J = 2.80 Hz, 3H), 1.39 (s, 9H), 1.00-0.21 (m, 2H). Step E. Procedure for preparation of tert-butyl 4-((2-(3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)-2-azaspiro[3.5]nonan-7-yl)methyl)pip erazine-1-carboxylate To a solution of tert-butyl 4-((2-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazo l-7- yl)-2-azaspiro[3.5]nonan-7-yl)methyl)piperazine-1-carboxylat e (2.50 g, 3.36 mmol) in THF (75.0 mL) was added palladium hydroxide on carbon (2.50 g, 100% w/w) and the reaction mixture was stirred under hydrogen atmosphere (60 psi) at room temperature. After 4 h, the reaction mixture was diluted with THF (60 mL) and filtered through celite and washed with 200 mL of THF: DCM (1:1). The combined organic solutions were concentrated to provide tert-butyl 4-((2-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl) -2- azaspiro[3.5]nonan-7-yl)methyl)piperazine-1-carboxylate (1.85 g, 3.26 mmol, 97% yield) as a pale yellow solid. MS (ESI) m/z: 565.4 [M+H] + . Step F. Procedure for preparation of 3-(1-methyl-7-(7-(piperazin-1-ylmethyl)-2- azaspiro[3.5]nonan-2-yl)-1H-indazol-3-yl)piperidine-2,6-dion e trifluoroacetate To a stirred solution of tert-butyl 4-((2-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7- yl)-2-azaspiro[3.5]nonan-7-yl)methyl)piperazine-1-carboxylat e (1.80 g, 3.18 mmol) in DCM (18 mL) was added trifluoroacetic acid (18.0 mL) at 0 °C, and the reaction mixture was stirred at room temperature. After 3 h, the reaction solution was concentrated, and the resulting crude material was triturated with diethyl ether to provide 3-(1-methyl-7-(7-(piperazin-1-ylmethyl)- 2-azaspiro[3.5]nonan-2-yl)-1H-indazol-3-yl)piperidine-2,6-di one trifluoroacetate (2.75 g) as an off-white solid which was carried forward without further purification. MS (ESI) m/z: 465.5 [M+H] + ; 1 H NMR; 400 MHz, DMSO-d6: δ 10.85 (s, 1H), 7.21 (d, J = 7.60 Hz, 1H), 6.99 (t, J = 7.60 Hz, 1H), 6.73 (d, J = 7.20 Hz, 1H), 4.32-4.30 (m, 1H), 4.17 (s, 3H), 3.71-3.56 (br, 4H), 3.41-2.81 (br, 10H), 2.67-2.60 (m, 2H), 2.33-2.17 (m, 2H), 1.99 (d, J = 12.80 Hz, 2H), 1.74- 1.71 (m, 3H), 1.51 (t, J = 12.80 Hz, 2H), 1.36-1.06 (m, 2H). Step G. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(4-((2-(3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-7-yl)-2-azaspiro[3.5]nonan-7-yl)methyl)piperazin-1-y l)propoxy)-2- methylphenyl)picolinate tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3- chloropropoxy)-2-methylphenyl)picolinate (60 mg, 90 µmol, 1 equiv.), 3-(1-methyl-7-(7- (piperazin-1-ylmethyl)-2-azaspiro[3.5]nonan-2-yl)-1H-indazol -3-yl)piperidine-2,6-dione (46 mg, 99 µmol, 1.1 equiv.) and sodium iodide (16 mg, 0.11 mmol, 1.2 equiv.) were suspended in NMP (2 mL). To the mixture was added diisopropylethylamine (58 mg, 78 µL, 0.45 mmol, 5 equiv.). The mixture was then purged with nitrogen and heated at 60 ºC. After 72 h, additional diisopropylethylamine (58 mg, 78 µL, 0.45 mmol, 5 equiv.) was added. The mixture was stirred at 60 ºC and the crude material was then purified by reverse-phase HPLC to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- (3-(3-(4-((2-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazo l-7-yl)-2-azaspiro[3.5]nonan-7- yl)methyl)piperazin-1-yl)propoxy)-2-methylphenyl)picolinate (19 mg, 17 µmol, 19% yield) as an off-white solid. MS (ESI) m/z: 549.6 [M+2H/2] + . Step H. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(4-((2-(3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-7-yl)-2-azaspiro[3.5]nonan-7-yl)methyl)piperazin-1-y l)propoxy)-2- methylphenyl)picolinic acid tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(4- ((2-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)-2 -azaspiro[3.5]nonan-7- yl)methyl)piperazin-1-yl)propoxy)-2-methylphenyl)picolinate (19 mg, 1 equiv., 17 µmol) was dissolved in 1,2-dichloromethane (2.4 mL). To the mixture was added 0.6 mL trifluoroacetic acid and the mixture was stirred at room temperature. After 48 h, the reaction solution was concentrated, the material was resuspended in water/acetonitrile, and lyophilized to afford 6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2( 1H)-yl)-3-(3-(3-(4-((2-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)-2-azaspiro[3. 5]nonan-7-yl)methyl)piperazin- 1-yl)propoxy)-2-methylphenyl)picolinic acid (22 mg, 17 µmol) as an off-white solid. MS (ESI) m/z: 1041.8 [M+H] + . 1 H NMR is consistent with structure. Example 229. Preparation of Compound 292. 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-4-piperidyl]butyl]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 4-(3-oxopropyl)piperidine-1-carboxylate To a solution of tert-butyl 4-(3-hydroxypropyl)piperidine-1-carboxylate (5.0 g, 20.55 mmol, 1.0 equiv.) in DCM (50 mL) was added DMP (10.46 g, 24.66 mmol, 7.64 mL, 1.2 equiv.) under 0 °C. Then the mixture was stirred at 25°C for 16 hours. The reaction mixture was then filtered and washed with ethyl acetate (30 mL × 2). The filtrate was concentrated under reduced pressure to give a residue which was purified by flash silica gel chromatography (17~35% ethyl acetate in petroleum ether) to give tert-butyl 4-(3-oxopropyl)piperidine-1-carboxylate (5.5 g, 19.67 mmol, 95.7% yield) as a light yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 9.67 (t, J = 1.6 Hz, 1H), 3.91 (m, 2H), 2.75 - 2.54 (m, 2H), 2.45 (m, 2H), 1.60 (d, J = 12.8 Hz, 2H), 1.49 - 1.40 (m, 2H), 1.38 (s, 9H), 1.36 - 1.30 (m, 1H), 1.00 - 0.88 (m, 2H). Step B. Procedure for preparation of tert-butyl 4-but-3-ynylpiperidine-1-carboxylate A mixture of tert-butyl 4-(3-oxopropyl)piperidine-1-carboxylate (5.0 g, 20.72 mmol, 1.0 equiv.), 1-diazo-1-dimethoxyphosphoryl-propan-2-one (4.78 g, 24.86 mmol, 1.2 equiv.) and K 2 CO 3 (7.16 g, 51.80 mmol, 2.5 equiv.) in MeOH (100 mL) was degassed and purged with N 2 three times. The reaction mixture was stirred at 40 °C for 16 hours under N2 atmosphere. The reaction mixture was then concentrated under reduced pressure to give a residue which was purified by column chromatography (SiO 2 , petroleum ether : ethyl acetate = 20:1) to give tert- butyl 4-but-3-ynylpiperidine-1-carboxylate (4.38 g, 18.4 mmol, 89.0% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 3.91 (d, J = 11.6 Hz, 2H), 2.75 (t, J = 2.4 Hz, 1H), 2.72 - 2.58 (m, 2H), 2.18 (m, 2H), 1.62 (d, J = 12.4 Hz, 2H), 1.50 (m, 1H), 1.43 - 1.34 (m, 11H), 0.94 (m, 2H). Step C. Procedure for preparation of tert-butyl 4-[4-(3-bromo-2-methyl-phenyl)but-3- ynyl]piperidine-1-carboxylate A mixture of tert-butyl 4-but-3-ynylpiperidine-1-carboxylate (600 mg, 2.53 mmol, 1.0 equiv.), 1-bromo-3-iodo-2-methyl-benzene (900.79 mg, 3.03 mmol, 1.2 equiv.), TEA (1.28 g, 12.64 mmol, 1.76 mL, 5.0 equiv.), CuI (96.29 mg, 505.61 μmol, 0.2 equiv.), and Pd(PPh 3 ) 2 Cl 2 (177.44 mg, 252.81 μmol, 0.1 equiv.) in DMF (20 mL) was degassed and purged with N 2 . The reaction mixture was stirred at 60 °C for 16 hours under N2 atmosphere. The reaction mixture was then diluted with water (50 mL) and extracted with ethyl acetate (40 mL × 3). The combined organic layers were washed with brine (50 mL × 2), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue which was purified by column chromatography (SiO2, petroleum ether : ethyl acetate = 20:1) to give tert-butyl 4-[4-(3-bromo-2-methyl-phenyl)but- 3-ynyl]piperidine-1-carboxylate (950 mg, 2.34 mmol, 92.4% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.56 (d, J = 8.0 Hz, 1H), 7.37 (d, J = 7.6 Hz, 1H), 7.10 (t, J = 8.0 Hz, 1H), 3.93 (d, J = 12.8 Hz, 2H), 2.70 - 2.65 (m, 1H), 2.46 (s, 4H), 1.68 (d, J = 13.2 Hz, 2H), 1.55 - 1.49 (m, 2H), 1.39 (s, 9H), 1.06 - 0.95 (m, 2H), -0.09 (s, 3H). Step D. Procedure for preparation of tert-butyl 4-[4-(3-bromo-2-methyl- phenyl)butyl]piperidine-1-carboxylate A mixture of tert-butyl 4-[4-(3-bromo-2-methyl-phenyl)but-3-ynyl]piperidine-1-carbox ylate (950 mg, 2.34 mmol, 1 equiv.) and Rh/Al2O3 (300 mg, 2.92 mmol, 1.25 equiv.) in EtOAc (10 mL) was degassed and purged with N2 three times and with H2 three times. The reaction mixture was stirred at 25 °C for 16 hours under H 2 (15 Psi) atmosphere. The mixture was filtered, and the filter cake was washed with ethyl acetate (40 mL). The filtrate was concentrated under reduced pressure to give tert-butyl 4-[4-(3-bromo-2-methyl- phenyl)butyl]piperidine-1-carboxylate (700 mg, 1.71 mmol, 72.6% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.56 (d, J = 8.0 Hz, 1H), 7.37 (d, J = 7.6 Hz, 1H), 7.10 (t, J = 7.6 Hz, 1H), 3.93 (m, 2H), 2.75 - 2.62 (m, 2H), 2.52 - 2.51 (m, 3H), 2.46 (s, 4H), 1.68 (m, 2H), 1.57 (m, 1H), 1.51 (q, J = 7.2 Hz, 2H), 1.38 (s, 11H), 1.10 - 0.92 (m, 2H). Step E. Procedure for preparation of 4-[4-(3-bromo-2-methyl-phenyl)butyl]piperidine To a solution of tert-butyl 4-[4-(3-bromo-2-methyl-phenyl)butyl]piperidine-1-carboxylate (700 mg, 1.71 mmol, 1.0 equiv.) in DCM (2 mL) was added HCl/dioxane (10 mL). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to remove HCl/dioxane and DCM and then concentrated under reduced pressure to give 4-[4-(3-bromo-2-methyl-phenyl)butyl]piperidine (590 mg, 1.70 mmol, 99.7% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ = 7.42 (d, J = 7.6 Hz, 1H), 7.15 (d, J = 7.6 Hz, 1H), 7.06 - 6.98 (m, 1H), 3.19 (m, 2H), 2.78 (m, 2H), 2.67 - 2.60 (m, 2H), 2.33 (s, 3H), 1.74 (m, 2H), 1.51 - 1.44 (m, 3H), 1.35 - 1.21 (m, 7H). Step F. Procedure for preparation of ethyl 2-[4-[4-(3-bromo-2-methyl-phenyl)butyl]-1- piperidyl]acetate A mixture of 4-[4-(3-bromo-2-methyl-phenyl)butyl]piperidine (300 mg, 865.22 μmol, 1.0 equiv.), ethyl 2-bromoacetate (130.04 mg, 778.70 μmol, 86.18 μL, 0.90 equiv.), TEA (262.65 mg, 2.60 mmol, 361.28 μL, 3.0 equiv.), and KI (28.73 mg, 173.04 μmol, 0.2 equiv.) in MeCN (5 mL) was stirred at 25 °C for 1 hour. The reaction mixture was diluted with water (40 mL) and extracted with ethyl acetate (40 mL × 3). The combined organic layers were washed with brine (40 mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give ethyl 2-[4-[4-(3-bromo-2-methyl-phenyl)butyl]-1-piperidyl]acetate (280 mg, 706.4 μmol, 81.6% yield) as a yellow oil. MS (ESI) m/z: 396.2 [M+H] + . Step G. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[1-(2-ethoxy-2-oxo- ethyl)-4-piperidyl]butyl]-2- methyl-phenyl]pyridine-2-carboxylate A mixture of ethyl 2-[4-[4-(3-bromo-2-methyl-phenyl)butyl]-1-piperidyl]acetate (280 mg, 706.43 μmol, 1.0 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (519.26 mg, 847.71 μmol, 1.2 equiv.), Ad2nBuP Pd G3 (102.89 mg, 141.29 μmol, 0.2 equiv.), and K2CO3 (146.45 mg, 1.06 mmol, 1.5 equiv.) in dioxane (5 mL) and H2O (1 mL) was degassed and purged with N 2 three times. The reaction mixture was stirred at 100 °C for 1 hour under microwave. The reaction mixture was diluted with water (40 mL) and extracted with ethyl acetate (40 mL × 3). The combined organic layers were washed with brine (40 mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue which was purified by flash silica gel chromatography (30-40% ethyl acetate in petroleum ether) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3- [4-[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]butyl]-2-methyl-phe nyl]pyridine-2-carboxylate (270 mg, 267.63 μmol, 37.9% yield) as a yellow solid. MS (ESI) m/z: 802.6 [M+H] + Step H. Procedure for preparation of 2-[4-[4-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyr idyl]-2-methyl- phenyl]butyl]-1-piperidyl]acetic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[4-[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]butyl]-2-m ethyl-phenyl]pyridine-2- carboxylate (270 mg, 336.64 μmol, 1.0 equiv.) and LiOH•H2O (42.38 mg, 1.01 mmol, 3.0 equiv.) in THF (2 mL) and H2O (2 mL) was stirred at 25 °C for 12 hours under N2 atmosphere. Afterwards, H 2 O (5mL) was added, and the pH of the reaction mixture was adjusted to ~ 6 by progressively adding diluted HCl. The mixture was filtered to give 2-[4-[4-[3-[6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -2-tert-butoxycarbonyl-3- pyridyl]-2-methyl-phenyl]butyl]-1-piperidyl]acetic acid (240 mg, 216.44 μmol, 64.2% yield) as a yellow solid MS (ESI) m/z: 775.0 [M+H] + Step I. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[1-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-4-piperidyl]butyl]-2-methyl-phenyl] pyridine-2-carboxylate A mixture of 2-[4-[4-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H-isoquinolin- 2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl-phenyl]butyl ]-1-piperidyl]acetic acid (100 mg, 129.20 μmol, 1.0 equiv.), 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (45.70 mg, 155.04 μmol, 1.2 equiv., HCl), and EDCI (37.15 mg, 193.80 μmol, 1.5 equiv.) in pyridine (2 mL), was stirred at 25 °C for 16 hours. The reaction mixture was then concentrated under reduced pressure, diluted with water, and extracted with DCM (40 mL × 3). The combined organic layers were washed with brine (40 mL × 2), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue which was purified by prep-TLC (SiO 2 , petroleum ether / ethyl acetate = 0:1) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[4-[1-[2-[[3-(2,6-diox o-3-piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-4-piperidyl]butyl]-2-methyl-phenyl] pyridine-2-carboxylate (70 mg, 65.68 μmol, 50.8% yield) as a red oil. MS (ESI) m/z: 1014.6 [M+H] + . Step J. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[4-[1-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]butyl]-2-methyl-phenyl]py ridine-2-carboxylic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[4-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indaz ol-6-yl]amino]-2-oxo-ethyl]-4- piperidyl]butyl]-2-methyl-phenyl]pyridine-2-carboxylate (70 mg, 69.02 μmol, 1.0 equiv.) and DCM (1.5 mL) and TFA (0.5 mL) was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue which was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[4-[1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-o xo-ethyl]-4-piperidyl]butyl]-2- methyl-phenyl]pyridine-2-carboxylic acid (28.14 mg, 28.91 μmol, 41.9% yield) as a yellow solid. MS (ESI) m/z: 958.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.01 - 12.75 (m, 1H), 10.89 (s, 1H), 8.13 (s, 1H), 8.07 - 7.98 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.64 (m, 2H), 7.50 - 7.41 (m, 3H), 7.36 (m, 2H), 7.16 (m, 1H), 7.07 (q, J = 7.6 Hz, 2H), 6.97 (d, J = 8.8 Hz, 1H), 6.86 (m, 1H), 4.98 (m, 2H), 4.33 (m, 1H), 4.04 - 3.81 (m, 6H), 3.03 (m, 2H), 2.70 - 2.56 (m, 6H), 2.54 (s, 2H), 2.39 - 2.30 (m, 2H), 2.21 - 2.14 (m, 1H), 1.99 (s, 3H), 1.75 (m, 2H), 1.52 (m, 2H), 1.45 - 1.24 (m, 7H). Example 230: Preparation of Compound 293a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- (4-(3-(2,6-dioxopiperidin-3-yl)-7-fluoro-1-methyl-1H-indazol -6-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of (E)-4-bromo-2,3-difluorobenzaldehyde O-methyl oxime To a solution of O-methylhydroxylamine (6.24 g, 74.66 mmol, 1.1 equiv., HCl) in DME (150 mL) was added K 2 CO 3 (28.14 g, 203.62 mmol, 3.0 equiv.) and 4-bromo-2,3-difluoro- benzaldehyde (15.0 g, 67.87 mmol, 1.0 equiv.). The mixture was stirred at 40 °C for 3 hours. The reaction mixture was filtered and concentrated under reduced pressure to give (E)-4- bromo-2,3-difluorobenzaldehyde O-methyl oxime (52 g, crude) as a white solid. Step B. Procedure for preparation of 6-bromo-7-fluoro-1H-indazole To a solution of (E)-4-bromo-2,3-difluorobenzaldehyde O-methyl oxime (26 g, 103.98 mmol, 1.0 equiv.) in DME (260 mL) was added N 2 H 4 •H 2 O (19.42 g, 380.17 mmol, 18.82 mL, 98% purity, 3.66 equiv.). The mixture was stirred at 90 °C for 16 hours. The reaction mixture was cooled to 20 °C, concentrated under reduced pressure, added into H2O, filtered, and concentrated under reduced pressure to give a residue. The residue was triturated with petroleum ether at 20 °C for 10 min to give 6-bromo-7-fluoro-1H-indazole (44.0 g, 193.70 mmol, 93.1% yield) as a white solid. MS (ESI) m/z: 215.2 [M + H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 8.22 (d, J = 3.4 Hz, 1H), 7.56 (d, J = 8.5 Hz, 1H), 7.29 (dd, J = 5.8, 8.5 Hz, 1H) 19 F NMR (377 MHz, DMSO-d6) δ = -125.15 (s, 1F) Step C. Procedure for preparation of 6-bromo-7-fluoro-3-iodo-1H-indazole To a solution of 6-bromo-7-fluoro-1H-indazole (44 g, 204.63 mmol, 1.0 equiv.) in DMF (400 mL) was added KOH (22.96 g, 409.26 mmol, 2.0 equiv.) and I2 (83.10 g, 327.41 mmol, 65.95 mL, 1.6 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was quenched by addition of saturated Na2SO3 solution (20 mL) at 20 °C, diluted with H2O, filtered, and concentrated under reduced pressure to give a residue. The residue was triturated with H2O at 20 °C for 10 min to give 6-bromo-7-fluoro-3-iodo-1H-indazole (49.0 g, 134.96 mmol, 65.9% yield) as a light brown solid. MS (ESI) m/z: 341.0 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 7.40 - 7.34 (m, 1H), 7.25 - 7.20 (m, 1H) 19 F NMR (377 MHz, DMSO-d 6 ) δ = -124.27 (s, 1F) Step D. Procedure for preparation of 6-bromo-7-fluoro-3-iodo-1-methyl-indazole To a solution of 6-bromo-7-fluoro-3-iodo-1H-indazole (49.0 g, 143.73 mmol, 1.0 equiv.) in DMF (500 mL) was added K 2 CO 3 (59.59 g, 431.19 mmol, 3.0 equiv.) and MeI (40.80 g, 287.46 mmol, 17.90 mL, 2.0 equiv.). The mixture was stirred at 40 °C for 16 hours. The reaction mixture was quenched by the addition of saturated NH 4 Cl solution (100 mL) at 0 °C, diluted with H 2 O, and extracted with EtOAc (1000 mL × 3). The combined organic layers were washed with H2O, dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether / ethyl acetate = 200/1 to 100/1) to give 6-bromo-7-fluoro-3-iodo-1-methyl-indazole (29.3 g, 81.75 mmol, 56.8% yield) as a white solid. MS (ESI) m/z: 357.0 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.42 - 7.34 (m, 1H), 7.21 (d, J = 8.8 Hz, 1H), 4.21 - 4.13 (m, 3H) 19 F NMR (377 MHz, DMSO-d6) δ = -126.90 (s, 1F) Step E. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-bromo-7- fluoro-1-methyl-1H-indazole A mixture of 6-bromo-7-fluoro-3-iodo-1-methyl-indazole (29.3 g, 82.55 mmol, 1.0 equiv.), 2,6-dibenzyloxy-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)pyridine (37.89 g, 90.80 mmol, 1.1 equiv.), Cs2CO3 (80.69 g, 247.64 mmol, 3.0 equiv.), and Pd(dppf)Cl2•CH2Cl2 (3.37 g, 4.13 mmol, 0.05 equiv.) in THF (300 mL) and H2O (30 mL) was degassed and purged with N 2 three times. The mixture was stirred at 60 °C for 16 hours under N 2 atmosphere. The mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether / ethyl acetate = 200/1 to 50/1) to give 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-bromo-7-fluoro-1-methyl -1H-indazole (30.35 g, 53.95 mmol, 65.3% yield) as a light yellow solid. MS (ESI) m/z: 520.2 [M+H] + 1 H NMR (400 MHz, CDCl 3 ) δ = 7.82 (d, J = 8.0 Hz, 1H), 7.42 - 7.38 (m, 2H), 7.36 - 7.29 (m, 3H), 7.27 - 7.19 (m, 6H), 6.99 (dd, J = 5.6, 8.4 Hz, 1H), 6.48 (d, J = 8.0 Hz, 1H), 5.40 (s, 2H), 5.37 (s, 2H), 4.23 - 4.18 (m, 3H) 19 F NMR (377 MHz, CDCl3) δ = -128.97 (s, 1F) Step F. Procedure for preparation of tert-butyl 4-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-7- fluoro-1-methyl-1H-indazol-6-yl)piperazine-1-carboxylate A mixture of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-bromo-7-fluoro-1-methyl -1H-indazole (500 mg, 964.55 μmol, 1 equiv.), tert-butyl piperazine-1-carboxylate (359.30 mg, 1.93 mmol, 2 equiv.), RuPhos (90.02 mg, 192.91 μmol, 0.2 equiv.), Pd2(dba)3 (88.33 mg, 96.46 μmol, 0.1 equiv.) ,and Cs 2 CO 3 (942.81 mg, 2.89 mmol, 3 equiv.) in toluene (6 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 110 °C for 12 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~20% ethyl acetate / petroleum ether) to give tert-butyl 4-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-7-fluoro-1-methyl- 1H-indazol-6-yl)piperazine-1-carboxylate (420 mg, 637.49 μmol, 66.0% yield) as a yellow oil. MS (ESI) m/z: 624.3 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 , EC11342-39-P1B1) δ = 7.87 (d, J = 8.0 Hz, 1H), 7.51 - 7.42 (m, 2H), 7.41 - 7.25 (m, 9H), 6.84 (dd, J = 7.6, 8.4 Hz, 1H), 6.58 (d, J = 8.0 Hz, 1H), 5.43 (d, J = 12.8 Hz, 4H), 4.14 (s, 3H), 3.49 (d, J = 4.4 Hz, 4H), 3.08 - 2.97 (m, 4H), 1.43 (s, 9H). Step G. Procedure for preparation of tert-butyl 4-(3-(2,6-dioxopiperidin-3-yl)-7-fluoro-1- methyl-1H-indazol-6-yl)piperazine-1-carboxylate To a solution of tert-butyl 4-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-7-fluoro-1-methyl-1H- indazol-6-yl)piperazine-1-carboxylate (400 mg, 641.32 μmol, 1 equiv.) in EtOH (2 mL) and THF (2 mL) was added Pd/C (90.06 mg, 641.32 μmol, 1 equiv.) and Pd(OH)2 (90.06 mg, 641.32 μmol, 1 equiv.) under N 2 atmosphere. The suspension was degassed under vacuum and purged with H2 several times. The mixture was then stirred at 25 °C for 2 hours under H2 (15 psi) atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~50% ethyl acetate / petroleum ether) to give tert-butyl 4-(3-(2,6-dioxopiperidin-3-yl)-7-fluoro-1-methyl- 1H-indazol-6-yl)piperazine-1-carboxylate (180 mg, 402.92 μmol, 62.8% yield) as a white solid. MS (ESI) m/z: 446.2 [M+H] + Step H. Procedure for preparation of 3-(7-fluoro-1-methyl-6-(piperazin-1-yl)-1H-indazol- 3-yl)piperidine-2,6-dione A solution of tert-butyl 4-(3-(2,6-dioxopiperidin-3-yl)-7-fluoro-1-methyl-1H-indazol- 6- yl)piperazine-1-carboxylate (190 mg, 426.50 μmol, 1 equiv.) in HCl/dioxane (2 mL) was stirred at 25 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give 3-(7-fluoro-1-methyl-6-(piperazin-1-yl)-1H-indazol-3-yl)pipe ridine-2,6-dione (150 mg, crude) as a white solid and used into the next step without further purification. MS (ESI) m/z: 346.0 [M+H] + . Step I. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-7-fluoro- 1-methyl-1H-indazol-6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy )-2- methylphenyl)picolinate A solution tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)- 3-(2-methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)oxy)phenyl) picolinate (90 mg, 120.82 μmol, 1 equiv.) and 3-(7-fluoro-1-methyl-6-(piperazin-1-yl)-1H-indazol-3-yl)pipe ridine-2,6- dione (55.36 mg, 144.98 μmol, 1.2 equiv., HCl) in DCM (1 mL) was stirred at 0 °C for 2 hours. Then NaBH(OAc) 3 (76.82 mg, 362.45 μmol, 3 equiv.) was added into the solution. The solution was stirred at 25 °C for 4 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC to give tert-butyl 6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2( 1H)-yl)-3-(3-(((1s,4r)-4-(4-(4- (3-(2,6-dioxopiperidin-3-yl)-7-fluoro-1-methyl-1H-indazol-6- yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (100 mg, 87.50 μmol, 72.4% yield) as a yellow solid. MS (ESI) m/z: 1075.5 [M+H] + . Step J. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-7-fluoro- 1-methyl-1H-indazol-6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy )-2-methylphenyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-7-fl uoro-1-methyl-1H-indazol-6- yl)piperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picol inate (90 mg, 83.77 μmol, 1 equiv.) in DCM (1 mL) and TFA (1 mL) was stirred at 25 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-7-fluoro-1- methyl-1H-indazol-6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinic acid (46.43 mg, 44.11 μmol, 52.6% yield) as a white solid. MS (ESI) m/z: 1018.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.89 (s, 1H), 8.14 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.41 (s, 7H), 7.10 - 7.03 (m, 1H), 6.93 (dd, J = 9.2, 14.4 Hz, 3H), 6.61 (d, J = 7.2 Hz, 1H), 4.97 (s, 2H), 4.32 (dd, J = 5.2, 10.0 Hz, 1H), 4.22 - 4.13 (m, 1H), 4.06 (s, 3H), 3.91 (t, J = 5.8 Hz, 2H), 3.09 (s, 4H), 3.02 (t, J = 5.6 Hz, 2H), 2.68 - 2.59 (m, 2H), 2.54 (s, 2H), 2.37 - 2.31 (m, 4H), 2.19 - 2.09 (m, 1H), 2.07 (d, J = 9.2 Hz, 2H), 1.87 (s, 3H), 1.81 - 1.74 (m, 2H), 1.48 - 1.40 (m, 2H), 1.37 - 1.28 (m, 4H), 1.23 (s, 3H), 1.11 - 0.99 (m, 2H). Example 231. Preparation of Compound 294 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4-[4-[1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-4-piperidyl]butyl]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 4-[4-(4-bromo-3-methyl-phenyl)but-3- ynyl]piperidine-1-carboxylate A mixture of 1-bromo-4-iodo-2-methyl-benzene (1.05 g, 3.54 mmol, 1.2 equiv.), tert-butyl 4- but-3-ynylpiperidine-1-carboxylate (700 mg, 2.95 mmol, 1.0 equiv.), TEA (1.49 g, 14.75 mmol, 2.05 mL, 5.0 equiv.), CuI (112.34 mg, 589.88 μmol, 0.2 equiv.), and dichloropalladiumtriphenylphosphane (207.02 mg, 294.94 μmol, 0.1 equiv.) in DMF (20 mL) was degassed and purged with N2 three times. The mixture was stirred at 60 °C for 12 hours under N2 atmosphere. The reaction mixture was partitioned between ethyl acetate (80 mL) and water (50 mL). The organic phase was separated, washed with brine (30 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~7% ethyl acetate/petroleum ether) to give tert- butyl 4-[4-(4-bromo-3-methyl-phenyl)but-3-ynyl]piperidine-1-carbox ylate (880 mg, 2.01 mmol, 68.2% yield) as a yellow oil. MS (ESI) m/z: 350.0 [M+H] + . Step B. Procedure for preparation of tert-butyl 4-[4-(4-bromo-3-methyl- phenyl)butyl]piperidine-1-carboxylate A mixture of tert-butyl 4-[4-(4-bromo-3-methyl-phenyl)but-3-ynyl]piperidine-1-carbox ylate (880 mg, 2.17 mmol, 1.0 equiv.) and Rh/Al2O3 (300.85 mg, 2.92 mmol, 1.35 equiv.) in EtOAc (10 mL) was degassed and purged with H 2 three times, and then the mixture was stirred at 25 °C for 16 hours under H2 atmosphere. The mixture was filtered, and the filter cake was washed with ethyl acetate (40 mL). The filtrate was concentrated under reduced pressure to give tert- butyl 4-[4-(4-bromo-3-methyl-phenyl)butyl]piperidine-1-carboxylate (800 mg, crude) as a yellow oil. MS (ESI) m/z: 354.2 [M-56+H] + . Step C. Procedure for preparation of 4-[4-(4-bromo-3-methyl-phenyl)butyl]piperidine A solution of tert-butyl 4-[4-(4-bromo-3-methyl-phenyl)butyl]piperidine-1-carboxylate (800 mg, 1.95 mmol, 1.0 equiv.) in HCl/dioxane (10 mL) was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give 4-[4-(4-bromo-3-methyl- phenyl)butyl]piperidine (660 mg, crude, HCl) as a white solid. MS (ESI) m/z: 310.2 [M+H] + 1 H NMR (400 MHz, CDCl 3 ) δ = 7.34 (d, J = 8.0 Hz, 1H), 6.95 (s, 1H), 6.77 (d, J = 8.0 Hz, 1H), 3.39 (d, J = 12.0 Hz, 2H), 2.77 - 2.68 (m, 2H), 2.45 (t, J = 8.0 Hz, 2H), 2.29 (s, 3H), 2.03 (s, 1H), 1.79 (d, J = 13.6 Hz, 3H), 1.56 (d, J = 2.8 Hz, 1H), 1.40 (dd, J = 6.0, 7.2 Hz, 2H), 1.25 (d, J = 3.6 Hz, 5H) Step D. Procedure for preparation of ethyl 2-[4-[4-(4-bromo-3-methyl-phenyl)butyl]-1- piperidyl]acetate To a solution of 4-[4-(4-bromo-3-methyl-phenyl)butyl]piperidine (300 mg, 865.22 μmol, 1.0 equiv., HCl) and ethyl 2-bromoacetate (130.04 mg, 778.70 μmol, 86.18 μL, 0.9 equiv.) in MeCN (5 mL) was added TEA (262.65 mg, 2.60 mmol, 361.28 μL, 3.0 equiv.) and KI (143.63 mg, 865.22 μmol, 1.0 equiv.). The mixture was stirred at 40 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give ethyl 2-[4-[4-(4-bromo-3-methyl- phenyl)butyl]-1-piperidyl]acetate (280 mg, crude) as a yellow solid. MS (ESI) m/z: 398.2 [M+H] + Step E. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[4-[1-(2-ethoxy-2-oxo- ethyl)-4-piperidyl]butyl]-2- methyl-phenyl]pyridine-2-carboxylate To a solution of ethyl 2-[4-[4-(4-bromo-3-methyl-phenyl)butyl]-1-piperidyl]acetate (280 mg, 706.43 μmol, 1.0 equiv.) and tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborol an-2-yl)pyridine-2-carboxylate (519.26 mg, 847.71 μmol, 1.2 equiv.) in dioxane (5 mL) and H2O (0.5 mL) was added K2CO3 (292.90 mg, 2.12 mmol, 3.0 equiv.) and Ad2nBuP Pd G3 (102.89 mg, 141.29 μmol, 0.2 equiv.). The mixture was stirred at 100 °C for 2 hours. The reaction mixture was partitioned between DCM (20 mL) and water (10 mL). The organic phase was separated, washed with brine (10 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~39% ethyl acetate/petroleum ether) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[4-[4-[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]butyl]-2-m ethyl-phenyl]pyridine-2- carboxylate (320 mg, 279.2 μmol, 39.5% yield) as a white solid. MS (ESI) m/z: 802.6 [M+H] + Step F. Procedure for preparation of 2-[4-[4-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyr idyl]-3-methyl- phenyl]butyl]-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[4-[4-[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]butyl]-2 -methyl-phenyl]pyridine-2- carboxylate (320 mg, 398.99 μmol, 1.0 equiv.) in THF (5 mL) and H2O (1 mL) was added LiOH•H2O (83.71 mg, 1.99 mmol, 5.0 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was acidified to pH = 3-4 with citric acid, filtered, and concentrated under reduced pressure to give 2-[4-[4-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-3-methyl- phenyl]butyl]-1-piperidyl]acetic acid (300 mg, crude) as a white solid. MS (ESI) m/z: 774.6 [M+H] + Step G. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[4-[1-[2-[[3-(2,6-diox o-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-4-piperidyl]butyl]-2-methyl -phenyl]pyridine-2- carboxylate To a solution of 2-[4-[4-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-3-methyl- phenyl]butyl]-1-piperidyl]acetic acid (100 mg, 129.20 μmol, 1.0 equiv.) and 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6- dione (33.37 mg, 129.20 μmol, 1.0 equiv.) in pyridine (1 mL) was added EDCI (37.15 mg, 193.80 μmol, 1.5 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was partitioned between ethyl acetate and water. The organic phase was separated, washed with brine, dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , petroleum ether: ethyl acetate = 0:1) to give tert- butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4-[4-[1-[2- [[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-o xo-ethyl]-4-piperidyl]butyl]-2- methyl-phenyl]pyridine-2-carboxylate (70 mg, 66.9 μmol, 51.8% yield) as a white solid. MS (ESI) m/z: 1014.4 [M+H] + Step H. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[4-[4-[1-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]butyl]-2-methyl-phenyl]py ridine-2-carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[4-[4-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-ind azol-6-yl]amino]-2-oxo-ethyl]-4- piperidyl]butyl]-2-methyl-phenyl]pyridine-2-carboxylate (70 mg, 69.02 μmol, 1.0 equiv.) in DCM (1 mL) was added TFA (7.87 mg, 69.02 μmol, 5.13 μL, 1.0 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by reverse-phase HPLC to give 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[4-[4-[1-[2-[[3-(2,6-dioxo- 3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]-4-pip eridyl]butyl]-2-methyl- phenyl]pyridine-2-carboxylic acid (25.45 mg, 26.3 μmol, 38.1% yield) as a yellow solid. MS (ESI) m/z: 958.6 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 12.87 (s, 1H), 10.88 (s, 1H), 10.03 - 9.65 (m, 1H), 8.13 (s, 1H), 8.07 - 8.00 (m, 2H), 7.79 (d, J = 7.6 Hz, 1H), 7.68 - 7.60 (m, 2H), 7.51 - 7.42 (m, 3H), 7.41 - 7.31 (m, 2H), 7.20 (s, 1H), 7.04 (s, 1H), 7.00 - 6.89 (m, 3H), 4.97 (s, 2H), 4.33 (dd, J = 5.2, 9.6 Hz, 1H), 3.96 - 3.87 (m, 5H), 3.02 (t, J = 5.6 Hz, 2H), 2.65 - 2.60 (m, 2H), 2.59 - 2.54 (m, 5H), 2.22 - 2.15 (m, 2H), 2.03 (s, 3H), 1.91 - 1.62 (m, 3H), 1.61 - 1.44 (m, 3H), 1.40 - 1.16 (m, 8H) Example 232. Preparation of Compound 295 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[1-[2-[[3- (2,6-dioxo-3-piperidyl)-5-fluoro-1-methyl-indazol-6-yl]amino ]-2-oxo-ethyl]-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 4-[3-(3-bromo-2-methyl- phenoxy)propyl]piperidine-1-carboxylate A mixture of 3-bromo-2-methyl-phenol (600 mg, 3.21 mmol, 1 equiv.), tert-butyl 4-(3- hydroxypropyl)piperidine-1-carboxylate (936.76 mg, 3.85 mmol, 1.2 equiv.), and 2-(tributyl- phosphanylidene)acetonitrile (1.55 g, 6.42 mmol, 2 equiv.) in toluene (10 mL) was degassed and purged with N2 three times. The mixture was stirred at 120 °C for 4 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 11% ethyl acetate / petroleum ether) to give tert-butyl 4-[3-(3-bromo-2-methyl-phenoxy)propyl]piperidine-1- carboxylate (1.2 g, 2.91 mmol, 90.7% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.17 - 7.13 (m, 1H), 7.12 - 7.07 (m, 1H), 6.96 (d, J = 7.6 Hz, 1H), 4.01 - 3.86 (m, 4H), 2.75 - 2.58 (m, 2H), 2.23 (s, 3H), 1.80 - 1.71 (m, 2H), 1.67 - 1.64 (m, 2H), 1.49 - 1.42 (m, 1H), 1.39 (s, 9H), 1.37 - 1.33 (m, 2H), 1.04 - 0.89 (m, 2H). Step B. Procedure for preparation of 4-[3-(3-bromo-2-methyl-phenoxy)propyl]piperidine A solution of tert-butyl 4-[3-(3-bromo-2-methyl-phenoxy)propyl]piperidine-1-carboxyla te (1.2 g, 2.91 mmol, 1 equiv.) in DCM (9 mL) and TFA (1 mL) was stirred at 25 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to 4-[3-(3-bromo- 2-methyl-phenoxy)propyl]piperidine (1.45 g, crude) as a white solid. Step C. Procedure for preparation of ethyl 2-[4-[3-(3-bromo-2-methyl-phenoxy)propyl]- 1-piperidyl]acetate A mixture of 4-[3-(3-bromo-2-methyl-phenoxy)propyl]piperidine (1.3 g, 2.44 mmol, 1 equiv., TFA) , ethyl 2-bromoacetate (366.70 mg, 2.20 mmol, 243.01 μL, 0.9 equiv.), and K 2 CO 3 (1.01 g, 7.32 mmol, 3 equiv.) in MeCN (10 mL) was degassed and purged with N2 three times. The mixture was stirred at 60 °C for 2 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~25% ethyl acetate/petroleum ether) to give ethyl 2-[4-[3-(3- bromo-2-methyl-phenoxy)propyl]-1-piperidyl]acetate (700 mg, 1.76 mmol, 72.0% yield) as a white solid. MS (ESI) m/z: 400.0 [M+H] + Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-(2-ethoxy-2-oxo- ethyl)-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate A mixture of ethyl 2-[4-[3-(3-bromo-2-methyl-phenoxy)propyl]-1-piperidyl]acetat e (180 mg, 451.88 μmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (332.16 mg, 542.26 μmol, 1.2 equiv.), Ad 2 nBuP Pd G 3 (98.73 mg, 135.56 μmol, 0.3 equiv.), and KF (39.38 mg, 677.82 μmol, 15.88 μL, 1.5 equiv.) in dioxane (4 mL) and H2O (0.4 mL) was degassed and purged with N2 three times. The mixture was stirred at 80 °C for 1 hour under microwave. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 24% ethyl acetate / petroleum ether) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[3-[1-(2-ethoxy-2-oxo-ethyl)-4-piperi dyl]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (200 mg, 248.7 μmol, 55.0% yield) as a white solid. MS (ESI) m/z: 804.7 [M+H] + Step E. Procedure for preparation of 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyr idyl]-2-methyl- phenoxy]propyl]-1-piperidyl]acetic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]propoxy]-2 -methyl-phenyl]pyridine-2- carboxylate (200 mg, 248.75 μmol, 1 equiv.) and lithium hydroxide hydrate (31.32 mg, 746.26 μmol, 3 equiv.) in THF (2 mL) and H2O (0.4 mL) was degassed and purged with N2 three times. The mixture was stirred at 40 °C for 3 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to remove THF. The resulting residue was diluted with H2O and extracted with DCM / MeOH (10:1) (3 mL × 3). The combined organic layers were washed with H 2 O (3 mL × 3), filtered, and concentrated under reduced pressure to give 2-[4- [3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-2-tert- butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]propyl]-1-piperid yl]acetic acid (150 mg, crude) as a white solid. MS (ESI) m/z: 776.6 [M+H] + Step F. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3- piperidyl)-5-fluoro-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-4-piperidyl]propoxy]-2-meth yl-phenyl]pyridine-2- carboxylate A mixture of 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H-isoquinolin- 2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]prop yl]-1-piperidyl]acetic acid (80 mg, 103.10 μmol, 1 equiv.), 3-(6-amino-5-fluoro-1-methyl-indazol-3-yl)piperidine-2,6-dio ne (28.48 mg, 103.10 μmol, 1 equiv.), and EDCI (59.29 mg, 309.30 μmol, 3 equiv.) in pyridine (3 mL) was degassed and purged with N2 three times. The mixture was stirred at 40 °C for 2 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~38% ethyl acetate / petroleum ether) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3- piperidyl)-5-fluoro-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-4-piperidyl]propoxy]-2-meth yl-phenyl]pyridine-2- carboxylate (100 mg, 96.69 μmol, 93.7% yield) as a white solid. MS (ESI) m/z: 1034.4 [M+H] + Step G. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3-piperidy l)-5-fluoro-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-4-piperidyl]propoxy]-2-methyl-pheny l]pyridine-2-carboxylic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-5-fluoro-1-met hyl-indazol-6-yl]amino]-2-oxo- ethyl]-4-piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carbo xylate (100 mg, 96.69 μmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL) was stirred at 40 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)- 5-fluoro-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]propoxy]-2-methyl-phenyl] pyridine-2-carboxylic acid (17.10 mg, 16.78 μmol, 17.3% yield) as an off-white solid. MS (ESI) m/z: 978.8 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 12.86 (d, J = 8.0 Hz, 1H), 12.69 - 12.41 (m, 1H), 10.89 (s, 1H), 8.32 - 8.22 (m, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 7.6 Hz, 1H), 7.70 - 7.59 (m, 2H), 7.52 - 7.28 (m, 6H), 7.13 - 7.06 (m, 1H), 6.97 (d, J = 8.4 Hz, 1H), 6.88 (d, J = 8.4 Hz, 1H), 6.63 (d, J = 8.0 Hz, 1H), 5.08 - 4.86 (m, 2H), 4.38 - 4.29 (m, 1H), 3.96 (s, 3H), 3.95 - 3.90 (m, 2H), 3.22 - 3.15 (m, 2H), 3.04 - 3.01 (m, 2H), 2.94 - 2.85 (m, 1H), 2.68 - 2.60 (m, 4H), 2.34 - 2.30 (m, 4H), 2.20 - 2.13 (m, 2H), 1.90 (s, 3H), 1.82 - 1.73 (m, 4H), 1.43 - 1.40 (m, 2H), 1.28 - 1.19 (m, 2H). Example 233. Preparation of Compound 297a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- ((2R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)-2-methylpiperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of methyl (1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexane-1-carboxylate A mixture of methyl (1s,4s)-4-hydroxycyclohexane-1-carboxylate (10.0 g, 63.21 mmol, 1.0 equiv.), 3-bromo-2-methyl-phenol (14.19 g, 75.86 mmol, 1.2 equiv.), and 2-(tributyl-λ5- phosphanylidene)acetonitrile (21.36 g, 88.50 mmol, 1.4 equiv.) in toluene (100 mL) was degassed and purged with N 2 three times, and then the mixture was stirred at 125 °C for 16 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~7 % ethyl acetate/petroleum ether) to give methyl (1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexane-1- carboxylate (76 g, 139.36 mmol, crude) as a light yellow oil. Step B. Procedure for preparation of ((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)methanol To mixture of methyl (1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexane-1-carboxylate (25.0 g, 45.84 mmol, 60% purity, 1.0 equiv.) in THF (200 mL) was added LiAlH4 (1.91 g, 50.43 mmol, 1.1 equiv.) at 0 °C for 10 min under N 2 , and then the mixture was stirred at 0 °C for 2 hours under N2 atmosphere. The mixture solution was quenched by Na2SO4•10H2O (50 g), filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate = 20/1 to 5/1) to give ((1r,4r)- 4-(3-bromo-2-methylphenoxy)cyclohexyl)methanol (34.0 g, 113.20 mmol, 82.3% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.13 (d, J = 7.6 Hz, 1H), 7.06 (t, J = 8.0 Hz, 1H), 7.01 (d, J = 7.6 Hz, 1H), 4.44 (t, J = 5.2 Hz, 1H), 4.25 - 4.18 (m, 1H), 3.23 (t, J = 5.6 Hz, 2H), 2.21 (s, 3H), 2.06 ( dd, J = 2.8, 12.0 Hz, 2H), 1.78 (d, J = 12.0 Hz, 2H), 1.39 - 1.30 (m, 3H), 1.09 - 1.02 (m, 2H) Step C. Procedure for preparation of (1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexane- 1-carbaldehyde To a solution of oxalyl dichloride (9.33 g, 73.52 mmol, 6.44 mL, 2.0 equiv.) in DCM (80 mL) was added DMSO (11.49 g, 147.04 mmol, 11.49 mL, 4.0 equiv.) in DCM (40 mL) dropwise at -70 °C under N2 for 30 min. The reaction mixture was stirred at -70 °C for 1 hour. ((1r,4r)-4- (3-bromo-2-methylphenoxy)cyclohexyl)methanol (11 g, 36.76 mmol, 1.0 equiv.) in DCM (40 mL) was added into the mixture at -70 °C under N 2 for 30 min. The resulting mixture was stirred at -70 °C for 1 hour under N2. TEA (22.32 g, 220.56 mmol, 30.70 mL, 6.0 equiv.) in DCM (40 mL) was added into the mixture, and the reaction mixture was stirred at -70 °C for 1 hour. The reaction mixture was quenched by addition of H 2 O, diluted, and extracted with DCM. The combined organic layers were washed with brine (400 mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give (1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexane-1-carbaldehyde (33.0 g, crude) as a yellow solid. 1 H NMR (400 MHz, DMSO-d6) δ = 9.60 (s, 1H), 7.14 (d, J = 8.0 Hz, 1H), 7.07 (t, J = 8.0 Hz, 1H), 7.02 (d, J = 8.0 Hz, 1H), 4.32 - 4.27 (m, 1H), 2.38 - 2.33 (m, 1H), 2.22 (s, 3H), 2.02 - 1.93 (m, 4H), 1.54 - 1.38 (m, 4H) Step D. Procedure for preparation of 1-(((1r,4r)-4-((E)-4-(benzyloxy)but-1-en-1- yl)cyclohexyl)oxy)-3-bromo-2-methylbenzene To a mixture of 3-benzyloxypropyl(triphenyl)phosphonium;bromide (24.80 g, 50.47 mmol, 1.2 equiv.) in THF (150 mL) was added LiHMDS (1 M, 63.09 mL, 1.5 equiv.) dropwise at -70 °C under N2 in THF (150 mL). The mixture was stirred at -50 °C for 3 hours, and then treated with (1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexane-1-carbaldehyd e (12.5 g, 42.06 mmol, 1.0 equiv.). The mixture was stirred at 25 °C for 16 hours under N 2 atmosphere. The mixture was quenched by aqueous NH4Cl (1000 mL) and extracted with EtOAc (1000 mL ×2 ). The combined organic layers were washed with brine (500 mL ×2 ) and then concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~5% ethyl acetate/petroleum ether) to give 1-(((1r,4r)-4-((E)-4-(benzyloxy)but-1-en-1- yl)cyclohexyl)oxy)-3-bromo-2-methylbenzene (24.0 g, 50.11 mmol, 59.5% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.40 - 7.27 (m, 5H), 7.14 (d, J = 8.0 Hz, 1H), 7.07 (t, J = 8.0 Hz, 1H), 7.02 (d, J = 8.0 Hz, 1H), 5.37 - 5.23 (m, 2H), 4.46 (s, 2H), 4.27 - 4.20 (m, 1H), 3.43 (t, J = 6.8 Hz, 2H), 2.34 - 2.27 (m, 3H), 2.21 (s, 3H), 2.04 - 2.00 (m, 2H), 1.64-1.60 (m, 2H), 1.46 - 1.36 (m, 2H), 1.26 - 1.16 (m, 2H). Step E. Procedure for preparation of 4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butan-1-ol To a solution of 1-(((1r,4r)-4-((E)-4-(benzyloxy)but-1-en-1-yl)cyclohexyl)oxy )-3-bromo-2- methylbenzene (16.0 g, 37.26 mmol, 1.0 equiv.) in ethyl acetate (50 mL) was added PtO2 (5.08 g, 22.36 mmol, 0.6 equiv.) under N 2 . The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H 2 (50 psi) at 45 °C for 16 hours. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~30% ethyl acetate/petroleum ether) to give 4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butan-1- ol (3.6 g, 9.15 mmol, 24.5% yield) as a light brown oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.13 (d, J = 8.0 Hz, 1H), 7.07 (t, J = 8.0 Hz, 1H), 7.01 (d, J = 8.0 Hz, 1H), 4.33 - 4.331 (m, 1H), 4.25 - 4.19 (m, 1H), 3.40 - 3.36 (m, 2H), 2.21 (s, 3H), 2.05 - 2.02 (m, 2H), 1.78 - 1.74 (m, 2H), 1.41 - 1.16 (m, 9H), 1.07 - 0.97 (m, 2H) Step F. Procedure for preparation of 4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butanal To a solution of oxalyl dichloride (2.60 g, 20.51 mmol, 1.80 mL, 2.0 equiv.) in DCM (80 mL) was added DMSO (3.21 g, 41.02 mmol, 3.21 mL, 4.0 equiv.) in DCM (40 mL) dropwise at - 70 °C under N 2 for 30 min. The reaction mixture was stirred at -70 °C for 1 hour.4-((1r,4s)-4- (3-bromo-2-methylphenoxy)cyclohexyl)butan-1-ol (3.5 g, 10.26 mmol, 1.0 equiv.) in DCM (40 mL) was added into the mixture at -70 °C under N2 for 30 min, and the resulting mixture was stirred at -70 °C for 1 hour under N 2 . TEA (6.23 g, 61.53 mmol, 8.56 mL, 6.0 equiv.) in DCM (40 mL) was added into the mixture and stirred at -70 °C for 1 hour. The reaction mixture was quenched by addition of H2O (400 mL), and extracted with DCM (300 mL × 1). The combined organic layers were washed with brine (400 mL × 2), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give 4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butanal (3.4 g, 8.58 mmol, 83.7% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 9.66 (s, 1H), 7.13 (d, J = 8.0 Hz, 1H), 7.06 (t, J = 8.0 Hz, 1H), 7.01 (d, J = 8.0 Hz, 1H), 4.26 - 4.16 (m, 1H), 2.43 - 2.39 (m, 2H), 2.20 (s, 3H), 2.03 (d, J = 10.0 Hz, 2H), 1.76 (d, J = 12.0 Hz, 2H), 1.57 - 1.50 (m, 2H), 1.39 -1.29 (m, 2H), 1.26 - 1.26 (m, 3H), 1.07 - 0.98 (m, 2H) Step G. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(2-methyl-3-(((1s,4r)-4-(4- oxobutyl)cyclohexyl)oxy)phenyl)picolinate A mixture of 4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butanal (3.3 g, 8.33 mmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]- 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-2-ca rboxylate (5.10 g, 8.33 mmol, 1.0 equiv.), Ad2nBuP Pd G3 (910.09 mg, 1.25 mmol, 0.15 equiv.), and K2CO3 (3.45 g, 24.99 mmol, 3.0 equiv.) in dioxane (10 mL) and H 2 O (1 mL) was degassed and purged with N 2 three times. The mixture was stirred at 100 °C for 3 hours under N 2 atmosphere. The reaction mixture was diluted by addition of H2O (300 mL) and extracted with ethyl acetate (500 mL × 2). The combined organic layers were washed with brine (250 mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~30% ethyl acetate/petroleum ether) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(2-methyl-3-(((1s,4r)-4- (4-oxobutyl)cyclohexyl)oxy)phenyl)picolinate (2.1 g, 2.62 mmol, 31.4% yield) as a yellow solid. MS (ESI) m/z: 745.1 [M+H] + . Step H. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-((2R)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-2-methylpiperazin-1-yl)butyl)cyclohe xyl)oxy)-2- methylphenyl)picolinate To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2-methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)o xy)phenyl)picolinate (150 mg, 201.36 μmol, 1 equiv.) in DCM (2 mL) was added 3-[1-methyl-6-[(3R)-3-methylpiperazin-1- yl]indazol-3-yl]piperidine-2,6-dione (75.62 mg, 221.49 μmol, 1.1 equiv.). The mixture was stirred at 25 °C for 1 hour, and then NaBH(OAc) 3 (128.03 mg, 604.08 μmol, 3 equiv.) was added. The mixture was stirred at 25 °C for 16 hours. The reaction mixture was diluted with H2O (20 mL) and extracted with DCM (20 mL × 3). The combined organic layers were washed with brine (30 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- (3-(((1s,4r)-4-(4-((2R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-met hyl-1H-indazol-6-yl)-2- methylpiperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)pi colinate (180 mg, crude) as a yellow oil. Step I. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-((2R)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-2-methylpiperazin-1-yl)butyl)cyclohe xyl)oxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4-((2R)-4-(3-(2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-6- yl)-2-methylpiperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphe nyl)picolinate (180 mg, 75.68 μmol, 1 equiv.) in DCM (2 mL) was added TFA (3.07 g, 26.92 mmol, 2 mL, 355.78 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to afford 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1s,4r)-4-(4-((2R)- 4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)-2-m ethylpiperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (27.57 mg, 26.64 μmol, 35.2% yield) as a yellow solid. MS (ESI) m/z: 1014.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.85 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.50 - 7.42 (m, 4H), 7.39 - 7.33 (m, 2H), 7.08 - 7.04 (m, 1H), 6.95 - 6.90 (m, 3H), 6.82 (s, 1H), 6.62 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.27 - 4.24 (m, 1H), 4.20 - 4.15 (m, 1H), 3.92 - 3.89 (m, 5H), 3.53 - 3.51 (m, 2H), 3.02 (t, J = 5.6 Hz, 2H), 2.94 - 2.84 (m, 2H), 2.74 - 2.67 (m, 2H), 2.62 - 2.59 (m, 2H), 2.39 - 2.13 (m, 5H), 2.08 - 2.06 (m, 2H), 1.87 (s, 3H), 1.79 - 1.77 (m, 2H), 1.43 - 1.24 (m, 9H), 1.09 - 1.03 (m, 5H) Example 234. Preparation of Compound 298a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pip eridin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperidin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinate To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2-methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)o xy)phenyl)picolinate (150 mg, 201.36 μmol, 1.0 equiv.) in DCM (2 mL) was added AcOH (12.09 mg, 201.36 μmol, 11.53 μL, 1.0 equiv.) and 3-(1-methyl-7-(piperidin-4-yl)-1H-indazol-3-yl)piperidine-2, 6-dione (72.29 mg, 221.49 μmol, 1.1 equiv.). The mixture was stirred at 25 °C for 1 hour. Then NaBH(OAc) 3 (128.03 mg, 604.08 μmol, 3.0 equiv.) was added into the mixture, and the resulting mixture was stirred at 25 °C for 11 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was diluted with H2O (50 mL) and extracted with DCM (50 mL × 2). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was used in the next step without further purification to give tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r) -4-(4-(4-(3-(2,6-dioxopiperidin- 3-yl)-1-methyl-1H-indazol-7-yl)piperidin-1-yl)butyl)cyclohex yl)oxy)-2- methylphenyl)picolinate (100 mg, 94.76 μmol, 47.0 % yield) as a yellow oil. Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperidin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-7- yl)piperidin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picol inate (100 mg, 94.76 μmol, 1.0 equiv.) in TFA (1 mL) was added DCM (1 mL). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)piperidin-1-yl)butyl)cyclohexyl)oxy)-2-methy lphenyl)picolinic acid (31.22 mg, 29.96 μmol, 31.6% yield) as a white solid. MS (ESI) m/z: 1000.0 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.89 (s, 1H), 8.16 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 7.2 Hz, 1H), 7.55 (d, J = 8.0 Hz, 1H), 7.50 - 7.42 (m, 3H), 7.40 - 7.33 (m, 2H), 7.26 (d, J = 7.2 Hz, 1H), 7.07 (t, J = 7.6 Hz, 2H), 6.97 - 6.90 (m, 2H), 6.63 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.35 (dd, J = 5.2, 9.8 Hz, 1H), 4.21 (s, 3H), 3.96 - 3.89 (m, 2H), 4.00 (s, 1H), 3.08 (dd, J = 1.2, 9.4 Hz, 2H), 3.03 (t, J = 5.2 Hz, 2H), 2.71 - 2.65 (m, 2H), 2.36 - 2.31 (m, 4H), 2.20 - 2.11 (m, 2H), 2.11 - 2.06 (m, 2H), 1.88 (s, 5H), 1.80 (d, J = 12.0 Hz, 4H), 1.53 - 1.45 (m, 2H), 1.38 - 1.20 (m, 8H), 1.06 (d, J = 11.6 Hz, 2H) Example 235. Preparation of Compound 299a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip eridin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperidin-1-yl)butyl)cyclohexyl)oxy)-2-methy lphenyl)picolinate To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2-methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)o xy)phenyl)picolinate (100 mg, 134.24 μmol, 1 equiv.) and 3-(1-methyl-6-(piperidin-4-yl)-1H-indazol-3-yl)piperidine-2, 6- dione (52.58 mg, 161.09 μmol, 1.2 eq) in DCM (2 mL) was added NaBH(OAc)3 (85.35 mg, 402.72 μmol, 3 equiv.). The mixture was stirred at 25 °C for 15 hours. The reaction mixture was concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r) -4-(4-(4-(3-(2,6-dioxopiperidin- 3-yl)-1-methyl-1H-indazol-6-yl)piperidin-1-yl)butyl)cyclohex yl)oxy)-2- methylphenyl)picolinate (155 mg, 108.77 μmol, 81.0% yield) as a yellow solid. MS (ESI) m/z: 528.8 [M/2+H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperidin-1-yl)butyl)cyclohexyl)oxy)-2-methy lphenyl)picolinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (150 mg, 142.14 μmol, 1 equiv.) in DCM (1 mL) and TFA (1 mL) was stirred at 25 °C for 15 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-6-yl)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (35.5 mg, 34.0 μmol, 23.9% yield) as a yellow solid MS (ESI) m/z: 999.8 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 13.04 - 12.76 (m, 1H), 10.89 (s, 1H), 8.13 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.67 (d, J = 8.4 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.49 - 7.43 (m, 3H), 7.41 - 7.33 (m, 3H), 7.10 - 7.01 (m, 2H), 6.95 (dd, J = 8.8, 16.8 Hz, 2H), 6.62 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.34 (dd, J = 5.2, 10.0 Hz, 1H), 4.23 - 4.18 (m, 1H), 3.98 (s, 3H), 3.94 - 3.89 (m, 4H), 3.62 (d, J = 12.0 Hz, 2H), 3.10 (d, J = 8.8 Hz, 2H), 3.05 - 2.93 (m, 4H), 2.67 (dd, J = 1.6, 3.6 Hz, 1H), 2.65 - 2.59 (m, 2H), 2.38 - 2.30 (m, 2H), 2.23 - 2.13 (m, 2H), 2.12 - 2.06 (m, 4H), 1.98 - 1.92 (m, 2H), 1.87 (s, 3H), 1.80 (d, J = 11.6 Hz, 2H), 1.68 (s, 1H), 1.34 (d, J = 6.8 Hz, 2H), 1.27 (d, J = 5.2 Hz, 2H), 1.12 - 1.02 (m, 2H) Example 236. Preparation of Compound 300a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- ((3R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)-3-methylpiperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-((3R)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-3-methylpiperazin-1-yl)butyl)cyclohe xyl)oxy)-2- methylphenyl)picolinate To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2-methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)o xy)phenyl)picolinate (100 mg, 134.24 μmol, 1 equiv.) and 3-[1-methyl-6-[(2R)-2-methylpiperazin-1-yl]indazol-3- yl]piperidine-2,6-dione (45.83 mg, 134.24 μmol, 1 equiv.) in DCM (2 mL) was added NaBH(OAc)3 (227.61 mg, 1.07 mmol, 8 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , DCM: MeOH = 12:1) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1s,4r)-4-(4-((3R)- 4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)-3-m ethylpiperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (55 mg, 49.84 μmol, 37.1% yield) as a yellow oil. MS (ESI) m/z: 1071.2 [M+H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-((3R)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-3-methylpiperazin-1-yl)butyl)cyclohe xyl)oxy)-2- methylphenyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1s,4r)-4-(4-((3R)-4-(3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-6-yl)-3- methylpiperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)pi colinate (55 mg, 51.39 μmol, 1 equiv.) in DCM (1 mL) and TFA (1 mL) was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep- HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(4-((3R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl -1H-indazol-6-yl)-3- methylpiperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)pi colinic acid (19.08 mg, 18.36 μmol, 35.72% yield, 97.58% purity) as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ = 10.85 (s, 1H), 8.15 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.8 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.52 - 7.42 (m, 4H), 7.40 - 7.30 (m, 2H), 7.06 (d, J = 8.0 Hz, 1H), 7.00 - 6.86 (m, 3H), 6.78 (s, 1H), 6.61 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.27 - 4.22 (m, 1H), 4.15 - 4.08 (m, 1H), 3.94 - 3.90 (m, 2H), 3.88 (s, 3H), 3.06 - 3.00 (m, 3H), 2.88 (d, J = 4.0 Hz, 1H), 2.64 - 2.60 (m, 2H), 2.28 (s, 2H), 2.20 - 2.12 (m, 3H), 2.08 (s, 3H), 1.93 (s, 1H), 1.87 (s, 3H), 1.82 - 1.76 (m, 2H), 1.49 - 1.22 (m, 11H), 1.02 (d, J = 5.6 Hz, 5H) Example 237: Preparation of Compound 300b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(4- ((3S)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)-3-methylpiperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(4-((3S)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-3-methylpiperazin-1-yl)butyl)cyclohe xyl)oxy)-2- methylphenyl)picolinate To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2-methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)o xy)phenyl)picolinate (109.10 mg, 146.45 μmol, 1 equiv.) and 3-[1-methyl-6-[(2S)-2-methylpiperazin-1-yl]indazol-3- yl]piperidine-2,6-dione (50 mg, 146.45 μmol, 1 equiv.) in DCM (3 mL) was added NaBH(OAc)3 (186.24 mg, 878.72 μmol, 6 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , DCM: MeOH = 10:1) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(4- ((3S)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)-3-methylpiperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (60 mg, 53.34 μmol, 36.4% yield) as a brown solid. Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(4-((3S)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-3-methylpiperazin-1-yl)butyl)cyclohe xyl)oxy)-2- methylphenyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1r,4r)-4-(4-((3S)-4-(3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-6-yl)-3- methylpiperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)pi colinate (60 mg, 56.06 μmol, 1 equiv.) in DCM (2 mL) and TFA (2 mL) was stirred at 25 °C for 16 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by reverse- phase HPLC (with 0.1% formic acid) to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(4-((3S)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-3-methylpiperazin-1-yl)butyl)cyclohe xyl)oxy)-2- methylphenyl)picolinic acid (37.55 mg, 36.92 μmol, 65.8% yield) as a white solid. MS (ESI) m/z: 1014.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.84 (s, 1H), 8.16 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.53 - 7.40 (m, 4H), 7.40 - 7.31 (m, 2H), 7.09 - 7.03 (m, 1H), 6.96 - 6.85 (m, 3H), 6.77 (s, 1H), 6.64 - 6.58 (m, 1H), 5.01 - 4.91 (m, 2H), 4.29 - 4.15 (m, 2H), 4.14 - 4.04 (m, 1H), 3.96 - 3.84 (m, 5H), 3.04 - 3.01 (m, 2H), 2.90 - 2.84 (m, 1H), 2.71 (d, J = 11.2 Hz, 1H), 2.66 - 2.59 (m, 2H), 2.28 (d, J = 7.6 Hz, 5H), 2.18 - 2.03 (m, 5H), 1.93 - 1.86 (m, 3H), 1.82 - 1.74 (m, 2H), 1.48 - 1.42 (m, 2H), 1.39 - 1.22 (m, 7H), 1.10 - 0.98 (m, 5H) Example 238. Preparation of Compound 297b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(4- ((2S)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)-2-methylpiperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(4-((2S)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-2-methylpiperazin-1-yl)butyl)cyclohe xyl)oxy)-2- methylphenyl)picolinate A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(2-methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)oxy)phe nyl)picolinate (100 mg, 134.24 μmol, 1.0 equiv.), and 3-[1-methyl-6-[(3S)-3-methylpiperazin-1-yl]indazol-3-yl]pipe ridine- 2,6-dione (60.87 mg, 161.09 μmol, 1.2 equiv., HCl) in DCM (2 mL) was stirred at 25 °C for 1 hour. To this reaction mixture was added NaBH(OAc)3 (142.25 mg, 671.20 μmol, 5.0 equiv.), and the mixture was stirred at 25 °C for 3 hours. The reaction mixture was diluted with water (40 mL) and extracted with DCM (40 mL × 3). The combined organic layers were washed with brine (40 mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1r,4r)-4-(4-((2S)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl -1H-indazol-6-yl)-2- methylpiperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)pi colinate (115 mg, 107.4 μmol, 80.0% yield) as a yellow solid. MS (ESI) m/z: 536.1 [M/2 + H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(4-((2S)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-2-methylpiperazin-1-yl)butyl)cyclohe xyl)oxy)-2- methylphenyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1r,4r)-4-(4-((2S)-4-(3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-6-yl)-2- methylpiperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)pi colinate (115.00 mg, 107.44 μmol, 1.0 equiv.), DCM (1.5 mL), and TFA (0.5 mL) was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove TFA and DCM to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(4-((2S)-4 -(3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-2-methylpiperazin-1-yl)butyl)cyclohe xyl)oxy)-2- methylphenyl)picolinic acid (18.53 mg, 17.26 μmol, 16.0% yield) was obtained as an off-white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.85 (s, 1H), 8.18 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.66 - 7.59 (m, 1H), 7.50 - 7.41 (m, 4H), 7.39 - 7.32 (m, 2H), 7.10 - 7.01 (m, 1H), 6.95 - 6.87 (m, 3H), 6.85 - 6.79 (m, 1H), 6.62 (m, 1H), 4.97 (s, 2H), 4.25 (m, 1H), 4.21 - 4.13 (m, 1H), 3.93 - 3.86 (m, 5H), 3.56 - 3.48 (m, 3H), 3.08 - 2.96 (m, 3H), 2.93 - 2.84 (m, 2H), 2.71 (m, 1H), 2.62 - 2.59 (m, 2H), 2.25 - 2.13 (m, 3H), 2.10 - 2.04 (m, 2H), 1.93 (s, 1H), 1.87 (s, 3H), 1.81 - 1.73 (m, 2H), 1.46 - 1.18 (m, 11H), 1.08 (m, 3H). Example 239. Preparation of Compound 302 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[1-[2-[[3- (2,6-dioxo-3-piperidyl)-7-fluoro-1-methyl-indazol-6-yl]amino ]-2-oxo-ethyl]-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of (E)-4-bromo-2,3-difluorobenzaldehyde O-methyl oxime To a solution of O-methylhydroxylamine (6.24 g, 74.66 mmol, 1.1 equiv., HCl) in DME (150 mL) was added K 2 CO 3 (28.14 g, 203.62 mmol, 3.0 equiv.) and 4-bromo-2,3-difluoro- benzaldehyde (15.0 g, 67.87 mmol, 1.0 equiv.). The mixture was stirred at 40 °C for 3 hours. The reaction mixture was filtered and concentrated under reduced pressure to give (E)-4- bromo-2,3-difluorobenzaldehyde O-methyl oxime (52 g, crude) as a white solid. Step B. Procedure for preparation of 6-bromo-7-fluoro-1H-indazole To a solution of (E)-4-bromo-2,3-difluorobenzaldehyde O-methyl oxime (26 g, 103.98 mmol, 1.0 equiv.) in DME (260 mL) was added N 2 H 4 •H 2 O (19.42 g, 380.17 mmol, 18.82 mL, 98% purity, 3.66 equiv.). The mixture was stirred at 90 °C for 16 hours. The reaction mixture was cooled to 20 °C, concentrated under reduced pressure, added into H2O, filtered, and concentrated under reduced pressure to give a residue. The residue was triturated with petroleum ether at 20 °C for 10 min to give 6-bromo-7-fluoro-1H-indazole (44.0 g, 193.70 mmol, 93.1% yield) as a white solid. MS (ESI) m/z: 215.2 [M + H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 8.22 (d, J = 3.4 Hz, 1H), 7.56 (d, J = 8.5 Hz, 1H), 7.29 (dd, J = 5.8, 8.5 Hz, 1H) 19 F NMR (377 MHz, DMSO-d 6 ) δ = -125.15 (s, 1F) Step C. Procedure for preparation of 6-bromo-7-fluoro-3-iodo-1H-indazole To a solution of 6-bromo-7-fluoro-1H-indazole (44 g, 204.63 mmol, 1.0 equiv.) in DMF (400 mL) was added KOH (22.96 g, 409.26 mmol, 2.0 equiv.) and I 2 (83.10 g, 327.41 mmol, 65.95 mL, 1.6 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was quenched by addition of saturated Na2SO3 solution (20 mL) at 20 °C, diluted with H2O, filtered, and concentrated under reduced pressure to give a residue. The residue was triturated with H 2 O at 20 °C for 10 min to give 6-bromo-7-fluoro-3-iodo-1H-indazole (49.0 g, 134.96 mmol, 65.9% yield) as a light brown solid. MS (ESI) m/z: 341.0 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.40 - 7.34 (m, 1H), 7.25 - 7.20 (m, 1H) 19 F NMR (377 MHz, DMSO-d6) δ = -124.27 (s, 1F) Step D. Procedure for preparation of 6-bromo-7-fluoro-3-iodo-1-methyl-indazole To a solution of 6-bromo-7-fluoro-3-iodo-1H-indazole (49.0 g, 143.73 mmol, 1.0 equiv.) in DMF (500 mL) was added K2CO3 (59.59 g, 431.19 mmol, 3.0 equiv.) and MeI (40.80 g, 287.46 mmol, 17.90 mL, 2.0 equiv.). The mixture was stirred at 40 °C for 16 hours. The reaction mixture was quenched by addition of saturated NH4Cl solution (100 mL) at 0 °C, diluted with H2O, and extracted with EtOAc (1000 mL × 3). The combined organic layers were washed with H 2 O (3000 mL × 3), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether / ethyl acetate = 200/1 to 100/1) to give 6-bromo-7-fluoro-3-iodo-1-methyl-indazole (29.3 g, 81.75 mmol, 56.8% yield) as a white solid. MS (ESI) m/z: 357.0 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 7.42 - 7.34 (m, 1H), 7.21 (d, J = 8.8 Hz, 1H), 4.21 - 4.13 (m, 3H) 19 F NMR (377 MHz, DMSO-d 6 ) δ = -126.90 (s, 1F) Step E. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-bromo-7- fluoro-1-methyl-1H-indazole A mixture of 6-bromo-7-fluoro-3-iodo-1-methyl-indazole (29.3 g, 82.55 mmol, 1.0 equiv.), 2,6-dibenzyloxy-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)pyridine (37.89 g, 90.80 mmol, 1.1 equiv.), Cs 2 CO 3 (80.69 g, 247.64 mmol, 3.0 equiv.), and Pd(dppf)Cl 2 •CH 2 Cl 2 (3.37 g, 4.13 mmol, 0.05 equiv.) in THF (300 mL) and H 2 O (30 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 60 °C for 16 hours under N2 atmosphere. The mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether / ethyl acetate = 200/1 to 50/1) to give 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-bromo-7-fluoro-1-methyl -1H-indazole (30.35 g, 53.95 mmol, 65.3% yield) as a light yellow solid. MS (ESI) m/z: 520.2 [M+H] + 1 H NMR (400 MHz, CDCl 3 ) δ = 7.82 (d, J = 8.0 Hz, 1H), 7.42 - 7.38 (m, 2H), 7.36 - 7.29 (m, 3H), 7.27 - 7.19 (m, 6H), 6.99 (dd, J = 5.6, 8.4 Hz, 1H), 6.48 (d, J = 8.0 Hz, 1H), 5.40 (s, 2H), 5.37 (s, 2H), 4.23 - 4.18 (m, 3H) 19 F NMR (377 MHz, CDCl 3 ) δ = -128.97 (s, 1F) Step F. Procedure for preparation of N-benzyl-3-(2,6-dibenzyloxy-3-pyridyl)-7-fluoro-1- methyl-indazol-6-amine A mixture of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-bromo-7-fluoro-1-methyl -1H-indazole (5 g, 9.65 mmol, 1.0 equiv.), phenylmethanamine (1.55 g, 14.47 mmol, 1.58 mL, 1.5 equiv.), Cs2CO3 (9.43 g, 28.94 mmol, 3.0 equiv.), and tBuBrettPhos Pd G3 (412.07 mg, 482.28 μmol, 0.05 equiv.) in 2-methylbutan-2-ol (30 mL) was degassed and purged with N2 three times. The mixture was stirred at 80 °C for 16 hours under N 2 atmosphere. The mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~10% ethyl acetate/petroleum ether) to give N-benzyl-3-(2,6- dibenzyloxy-3-pyridyl)-7-fluoro-1-methyl-indazol-6-amine (8.5 g, 15.46 mmol, 80.1% yield) as a yellow oil. MS (ESI) m/z: 545.5 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 7.83 (d, J = 8.0 Hz, 1H), 7.48 - 7.43 (m, 2H), 7.41 - 7.27 (m, 9H), 7.26 - 7.16 (m, 4H), 7.14 (d, J = 8.8 Hz, 1H), 6.55 (d, J = 8.0 Hz, 1H), 6.49 (dd, J = 7.2, 8.8 Hz, 1H), 6.37 - 6.31 (m, 1H), 5.39 (d, J = 5.6 Hz, 4H), 4.43 (d, J = 6.4 Hz, 2H), 4.14 - 4.05 (m, 3H) 19 F NMR (377 MHz, DMSO-d 6 ) δ = -162.61 (s, 1F) Step G. Procedure for preparation of 3-(6-amino-7-fluoro-1-methyl-indazol-3- yl)piperidine-2,6-dione To a solution of N-benzyl-3-(2,6-dibenzyloxy-3-pyridyl)-7-fluoro-1-methyl-ind azol-6-amine (2.0 g, 3.67 mmol, 1.0 equiv.) in THF (40 mL) was added Pd/C (1.00 g, 939.67 μmol, 10% purity, 0.256 equiv.) and Pd(OH)2 (1.00 g, 1.42 mmol, 20% purity, 0.388 equiv.) under N2. The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H2 (15 psi) at 20 °C for 3 hours. The mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by reverse-phase HPLC (with 0.05% formic acid) to give 3-(6-amino-7-fluoro-1-methyl-indazol-3-yl)piperidine-2,6-dio ne (1336.60 mg, 4.39 mmol, 87.8% yield) as a gray solid. MS (ESI) m/z: 277.2 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 10.84 (s, 1H), 7.17 (d, J = 8.8 Hz, 1H), 6.63 (t, J = 8.0 Hz, 1H), 5.34 - 5.26 (m, 2H), 4.25 - 4.18 (m, 1H), 3.99 (s, 3H), 2.64 - 2.58 (m, 2H), 2.31 - 2.22 (m, 1H), 2.19 - 2.10 (m, 1H) 19 F NMR (377 MHz, DMSO-d6) δ = -163.31 (s, 1F) Step H. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-diox o-3-piperidyl)-7-fluoro-1- methyl-indazol-6-yl]amino]-2-oxo-ethyl]-4-piperidyl]propoxy] -2-methyl- phenyl]pyridine-2-carboxylate To a solution of 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]propyl]-1- piperidyl]acetic acid (118 mg, 152.07 μmol, 1 equiv.) and 3-(6-amino-7-fluoro-1-methyl- indazol-3-yl)piperidine-2,6-dione (50.41 mg, 182.49 μmol, 1.2 equiv.) in pyridine (1 mL) was added EDCI (43.73 mg, 228.11 μmol, 1.5 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was then added dropwise into water, and then filtered and concentrated to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- [3-[3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-7-fluoro-1-methyl-in dazol-6-yl]amino]-2-oxo-ethyl]- 4-piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate (137 mg, 132.4 μmol, 87.1% yield) as a brown solid. MS (ESI) m/z: 518.2 [M/2+H] + . Step I. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3-piperidy l)-7-fluoro-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-4-piperidyl]propoxy]-2-methyl-pheny l]pyridine-2-carboxylic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-7-fluoro-1-met hyl-indazol-6-yl]amino]-2-oxo- ethyl]-4-piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carbo xylate (137 mg, 132.47 μmol, 1 equiv.) in TFA (1 mL) and DCM (3 mL) was stirred at 25 °C for 2 hours. The residue was concentrated under reduced pressure, and the resulting residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[1-[2- [[3-(2,6-dioxo-3-piperidyl)-7-fluoro-1-methyl-indazol-6-yl]a mino]-2-oxo-ethyl]-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid (38.1 mg, 38.95 μmol, 29.4% yield) as an off-white solid. MS (ESI) m/z: 978.7 [M+H] + . 1H NMR (400 MHz, DMSO-d 6 ) δ = 12.88 (dd, J = 3.2, 6.0 Hz, 1H), 10.92 (s, 1H), 9.79 - 9.62 (m, 1H), 8.03 (d, J = 8.4 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.65 - 7.53 (m, 2H), 7.51 - 7.31 (m, 7H), 7.09 (t, J = 8.0 Hz, 1H), 6.97 (d, J = 8.8 Hz, 1H), 6.88 (d, J = 8.4 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.37 (dd, J = 5.2, 10.4 Hz, 1H), 4.10 (s, 3H), 4.00 - 3.94 (m, 2H), 3.92 (t, J = 5.6 Hz, 2H), 3.15 (d, J = 2.4 Hz, 2H), 3.04 - 3.01 (m, 2H), 2.95 - 2.88 (m, 2H), 2.20 - 2.14 (m, 4H), 1.90 (s, 3H), 1.83 - 1.70 (m, 5H), 1.45 - 1.38 (m, 2H), 1.28 (dd, J = 4.8, 13.2 Hz, 3H). Example 240. Preparation of Compound 303 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)ethyl)pip eridin-4-yl)propoxy)-2- methylphenyl)picolinic acid Step A. Procedure for preparation of (E)-3-(2, 6-bis(benzyloxy)pyridin-3-yl)-6-(2- ethoxyvinyl)-1-methyl-1H-indazole A mixture of 3-(2, 6-bis(benzyloxy)pyridin-3-yl)-6-bromo-1-methyl-1H-indazole (3 g, 6.00 mmol, 1 equiv.), (E)-2-(2-ethoxyvinyl)-4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolane (1.42 g, 7.19 mmol, 1.2 equiv.), K3PO4 (3.18 g, 14.99 mmol, 2.5 equiv.), Pd(OAc)2 (67.30 mg, 299.77 μmol, 0.05 equiv.), and dicyclohexyl-[2-(2, 6-dimethoxyphenyl)phenyl]phosphane (2.46 g, 6.00 mmol, 1 equiv.) in MeCN (30 ml) and H 2 O (10 ml) was degassed and purged with N 2 three times. The mixture was stirred at 60 °C for 2 hours under N2 atmosphere. The reaction was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 20% ethyl acetate/petroleum ether) to give (E)-3-(2, 6- bis(benzyloxy)pyridin-3-yl)-6-(2-ethoxyvinyl)-1-methyl-1H-in dazole (2.8 g, 5.70 mmol, 95.0 % yield) as a yellow oil. MS (ESI) m/z: 492.3 [M+H] + . Step B. Procedure for preparation of 2-(3-(2, 6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H- indazol-6-yl)acetaldehyde A mixture of (E)-3-(2, 6-bis(benzyloxy)pyridin-3-yl)-6-(2-ethoxyvinyl)-1-methyl-1H- indazole (2.8 g, 5.70 mmol, 1 equiv.) in HCOOH (30 mL) was stirred at 40 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give 2-(3-(2, 6- bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-6-yl)acetald ehyde (2.5 g, crude) as a yellow solid. MS (ESI) m/z: 464.2 [M+H] + . Step C. Procedure for preparation of 3-(2, 6-bis(benzyloxy)pyridin-3-yl)-6-(2, 2- dimethoxyethyl)-1-methyl-1H-indazole A mixture of 2-(3-(2, 6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-6-yl)aceta ldehyde (2.5 g, 5.39 mmol, 1 equiv.) and TsOH (92.88 mg, 539.34 μmol, 0.1 equiv.) in MeOH (10 ml) and toluene (20 ml) was degassed and purged with N 2 three times. The mixture was stirred at 130 °C for 12 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 3-(2, 6-bis(benzyloxy)pyridin-3-yl)-6-(2, 2-dimethoxyethyl)-1-methyl-1H-indazole (460 mg, 902.6 μmol, 16.7 % yield) as a yellow oil. MS (ESI) m/z: 510.4 [M+H] + . Step D. Procedure for preparation of 3-(6-(2, 2-dimethoxyethyl)-1-methyl-1H-indazol-3- yl)piperidine-2, 6-dione A mixture of 3-(2, 6-bis(benzyloxy)pyridin-3-yl)-6-(2, 2-dimethoxyethyl)-1-methyl-1H- indazole (460.00 mg, 902.68 μmol, 1equiv.), Pd/C (960.63 mg, 902.68 μmol, 10 % purity, 1 equiv.), and Pd(OH)2 (1.27 g, 902.68 μmol, 10 % purity, 1 equiv.) in THF (8 ml) was stirred at 50 ℃ for 12 hours under H2 atmosphere. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give 3-(6-(2, 2-dimethoxyethyl)-1-methyl-1H-indazol- 3-yl)piperidine-2, 6-dione (330 mg, crude) as a yellow solid. MS (ESI) m/z: 332.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.88 (s, 1H), 7.60 (d, J = 8.4 Hz, 1H), 7.44 (s, 1H), 7.02 (dd, J = 1.2, 8.4 Hz, 1H), 4.64 (t, J = 5.6 Hz, 1H), 4.34 (dd, J = 4.8, 10.0 Hz, 1H), 3.96 (s, 3H), 3.25 (s, 6H), 2.98 (d, J = 5.6 Hz, 2H), 2.71 - 2.60 (m, 2H), 2.37 - 2.16 (m, 2H) Step E. Procedure for preparation of 2-(3-(2, 6-dioxopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)acetaldehyde A mixture of 3-(6-(2, 2-dimethoxyethyl)-1-methyl-1H-indazol-3-yl)piperidine-2, 6-dione (100 mg, 301.78 μmol, 1 equiv.) in HCOOH (2 ml) was stirred at 100 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give 2-(3-(2, 6-dioxopiperidin- 3-yl)-1-methyl-1H-indazol-6-yl)acetaldehyde (70 mg, crude) as a green solid. MS (ESI) m/z: 286.2 [M+H] + . Step F. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-(3-(2,6-dioxopipe ridin-3-yl)-1-methyl-1H- indazol-6-yl)ethyl)piperidin-4-yl)propoxy)-2-methylphenyl)pi colinic acid A mixture of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(3-(piperidin-4-yl)propoxy)phenyl)picolinic acid (150 mg, 226.65 μmol, 1 equiv.), 2- (3-(2, 6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)acetaldehyde (71.13 mg, 249.32 μmol, 1.1 equiv.), and 4A MS (30 mg) in DCM (4 ml) was stirred at 25 °C for 12 hours. Afterwards, NaBH(OAc)3 (96.07 mg, 453.30 μmol, 2 equiv.) was added into the mixture, and then the mixture was stirred at 25 ℃ for 0.5 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2- (3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)ethyl) piperidin-4-yl)propoxy)-2- methylphenyl)picolinic acid (29.55 mg, 28.42 μmol, 12.54 % yield) as a yellow solid. MS (ESI) m/z: 931.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.88 (s, 1H), 8.16 (s, 1H), 8.01 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.64 - 7.59 (m, 3H), 7.47 - 7.42 (m, 3H), 7.40 - 7.34 (m, 2H), 7.09 (t, J = 8.0 Hz, 1H), 7.00 (d, J = 8.0 Hz, 1H), 6.93 (d, J = 8.8 Hz, 1H), 6.87 (d, J = 8.0 Hz, 1H), 6.64 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.33 (dd, J = 5.2, 9.6 Hz, 1H), 3.98 - 3.90 (m, 7H), 3.02 (d, J = 4.8 Hz, 2H), 2.94 - 2.88 (m, 2H), 2.76 - 2.68 (m, 2H), 2.63 (dd, J = 6.0, 11.6 Hz, 2H), 2.22 - 2.09 (m, 4H), 1.90 (s, 3H), 1.79 - 1.66 (m, 4H), 1.46 - 1.25 (m, 4H), 1.24 - 1.03 (m, 3H) Example 241. Preparation of Compound 304 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)methyl)piperid in-4-yl)propoxy)-2- methylphenyl)picolinic acid Step A. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-6- vinyl-1H-indazole A mixture of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-bromo-1-methyl-1H-indaz ole (5 g, 9.99 mmol, 1 equiv.), trifluoro(vinyl)-l4-borane potassium salt (1.34 g, 9.99 mmol, 1 equiv.), Pd(dppf)Cl 2 (219.34 mg, 299.77 μmol, 0.03 equiv.), and K 3 PO 4 (3.18 g, 14.99 mmol, 1.5 equiv.) in dioxane (27.3 mL) and H 2 O (2.7 mL) was degassed and purged with N 2 three times. The mixture was stirred at 80 °C for 2 hours under N2 atmosphere. The reaction was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 15% ethyl acetate/petroleum ether) to give 3-(2,6- bis(benzyloxy)pyridin-3-yl)-1-methyl-6-vinyl-1H-indazole (4.4 g, 9.83 mmol, 98.39% yield) as a yellow oil. MS (ESI) m/z: 448.2 [M+H] + Step B. Procedure for preparation of 1-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H- indazol-6-yl)ethane-1,2-diol A mixture of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-6-vinyl-1H-indaz ole (1.90 g, 4.25 mmol, 1 equiv.), K2OsO4.2H2O (156.43 mg, 424.56 μmol, 0.1 equiv.), and NMO (1.49 g, 12.74 mmol, 1.34 mL, 3 equiv.) in acetone (35 mL) and H2O (3.5 mL) was degassed and purged with N 2 three times. The mixture was stirred at 65 °C for 2 hours under N 2 atmosphere. The reaction was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 66% ethyl acetate / petroleum ether) to give 1-(3-(2,6- bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-6-yl)ethane- 1,2-diol (2.2 g, 4.57 mmol, 53.81% yield) as a yellow oil. MS (ESI) m/z: 482.3 [M+H] + Step C. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H- indazole-6-carbaldehyde A mixture of 1-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-6- yl)ethane-1,2-diol (2.2 g, 4.57 mmol, 1 equiv.) and NaIO 4 (1.95 g, 9.14 mmol, 506.32 μL, 2 equiv.) in acetone (20 mL) and H2O (2 mL) was stirred at 25 °C for 2 hours. The mixture was quenched by sat. Na2SO3 (20 mL) and exacted with DCM (30 mL, 10 mL × 3). The combined organic phase was concentrated to a residue. The residue was purified by flash silica gel chromatography (0 ~ 30% ethyl acetate / petroleum ether) to give 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl- 1H-indazole-6-carbaldehyde (2 g, 4.45 mmol, 97.39% yield) as a yellow oil. MS (ESI) m/z: 450.2 [M+H] + Step D. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-(1,3-dioxolan- 2-yl)-1-methyl-1H-indazole A mixture of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazole-6-ca rbaldehyde (700 mg, 1.56 mmol, 1 equiv.), ethylene glycol (483.30 mg, 7.79 mmol, 434.24 μL, 5 equiv.), and TsOH (26.82 mg, 155.73 μmol, 0.1 equiv.) in toluene (15 mL) was degassed and purged with N 2 three times. The mixture was stirred at 120 °C for 12 hours under N 2 atmosphere. The reaction was filtered and concentrated under reduced pressure to give 3-(2,6- bis(benzyloxy)pyridin-3-yl)-6-(1,3-dioxolan-2-yl)-1-methyl-1 H-indazole (500 mg, 1.01 mmol, 65.05% yield) as a yellow solid. MS (ESI) m/z: 494.2 [M+H] + . Step E. Procedure for preparation of 3-(6-(1,3-dioxolan-2-yl)-1-methyl-1H-indazol-3- yl)piperidine-2,6-dione A mixture of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-(1,3-dioxolan-2-yl)-1-m ethyl-1H-indazole (500 mg, 1.01 mmol, 1 equiv.), Pd/C (600 mg, 563.80 μmol, 10% purity, 0.557 equiv.), and Pd(OH) 2 (600 mg, 427.23 μmol, 10% purity, 0.422 equiv.) in THF (15 mL) was degassed and purged with N 2 three times. The mixture was stirred at 25 °C for 12 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 66% ethyl acetate / petroleum ether) to give 3-(6-(1,3-dioxolan-2-yl)-1-methyl-1H-indazol-3- yl)piperidine-2,6-dione (260 mg, 816.31 μmol, 80.58% yield) as a yellow oil. MS (ESI) m/z: 316.1 [M+H] + . Step F. Procedure for preparation of 3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazole-6- carbaldehyde A mixture of 3-(6-(1,3-dioxolan-2-yl)-1-methyl-1H-indazol-3-yl)piperidine -2,6-dione (100 mg, 317.13 μmol, 1 equiv.) in HCOOH (3 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 2 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give 3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazole-6-carbaldehyde (100 mg, crude) as a yellow oil. MS (ESI) m/z: 272.0 [M+H] + . Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-((3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H- indazol-6-yl)methyl)piperidin-4-yl)propoxy)-2-methylphenyl)p icolinic acid The pH of a mixture containing 3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazole-6- carbaldehyde (100 mg, 368.64 μmol, 1 equiv.), 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(2-methyl-3-(3-(piperidin-4-y l)propoxy)phenyl)picolinic acid (243.97 mg, 368.64 μmol, 1 equiv.), and DCM (3 mL) was adjusted to ~6 with DIEA (0.5 mL). The mixture was stirred at 40°C for 2 hours, and then NaBH(OAc) 3 (234.39 mg, 1.11 mmol, 3 equiv.) was added. The reaction mixture was then stirred at 40°C for 0.5 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-((3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H-indazol- 6-yl)methyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinic acid (25.22 mg, 26.94 μmol, 7.31% yield) as a yellow solid. MS (ESI) m/z: 917.8 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.99 - 12.77 (m, 1H), 10.94 - 10.85 (m, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.81 - 7.73 (m, 2H), 7.63 (d, J = 7.6 Hz, 2H), 7.48 - 7.41 (m, 3H), 7.36 (td, J = 7.6, 11.2 Hz, 2H), 7.18 (d, J = 8.4 Hz, 1H), 7.09 (t, J = 8.0 Hz, 1H), 6.96 (d, J = 8.8 Hz, 1H), 6.87 (d, J = 8.4 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.38 (dd, J = 5.2, 9.6 Hz, 1H), 4.00 (s, 3H), 3.98 - 3.88 (m, 4H), 3.03 (t, J = 5.6 Hz, 2H), 2.75 - 2.59 (m, 4H), 2.40 - 2.31 (m, 4H), 2.21 - 2.14 (m, 1H), 1.89 (s, 3H), 1.85 - 1.69 (m, 4H), 1.52 - 1.20 (m, 6H) Example 242. Preparation of Compound 317 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-((3- (2,4-dioxotetrahydropyrimidin-1(2H)-yl)-1-methyl-1H-indazol- 6-yl)amino)-2- oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-((3-(2,4-dioxotet rahydropyrimidin-1(2H)-yl)- 1-methyl-1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)pr opoxy)-2- methylphenyl)picolinate To a solution of 2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-2-methylpheno xy)propyl)piperidin-1- yl)acetic acid (100 mg, 128.87 μmol, 1 equiv.) in pyridine (3 mL) was added EDCI (32.12 mg, 167.54 μmol, 1.3 equiv.) and 1-(6-amino-1-methyl-1H-indazol-3-yl)dihydropyrimidine- 2,4(1H,3H)-dione (43.4 mg, 167.54 μmol, 1.3 equiv.). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure followed by the addition of DCM (50 mL) and water (50 mL). The aqueous layer was washed with DCM ,(15 mL × 2), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1- (2-((3-(2,4-dioxotetrahydropyrimidin-1(2H)-yl)-1-methyl-1H-i ndazol-6-yl)amino)-2- oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinate (150 mg, 125.34 μmol, 97.26% yield) as a yellow oil. MS (ESI) m/z: 1017.7 [M+H] + Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-((3-(2,4-dioxotet rahydropyrimidin-1(2H)-yl)- 1-methyl-1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)pr opoxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-((3-(2,4-dioxotetrahydropyrimidin-1( 2H)-yl)-1-methyl-1H-indazol-6- yl)amino)-2-oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl) picolinate (150 mg, 147.46 μmol, 1 equiv.) in DCM (1 mL) was added TFA (3.07 g, 26.93 mmol, 2 mL, 182.59 equiv.). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-(1-(2-((3-(2,4- dioxotetrahydropyrimidin-1(2H)-yl)-1-methyl-1H-indazol-6-yl) amino)-2-oxoethyl)piperidin- 4-yl)propoxy)-2-methylphenyl)picolinic acid (54.02 mg, 56.94 μmol, 38.61% yield) as a white solid. MS (ESI) m/z: 961.6 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 13.07 - 12.69 (m, 1H), 10.54 (s, 1H), 10.22 - 9.76 (m, 1H), 8.13 (s, 1H), 8.08 - 7.98 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.65 - 7.54 (m, 2H), 7.51 - 7.43 (m, 3H), 7.40 - 7.32 (m, 2H), 7.18 (d, J = 9.2 Hz, 1H), 7.13 - 7.05 (m, 1H), 6.97 (d, J = 8.4 Hz, 1H), 6.88 (d, J = 8.4 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 3.96 (t, J = 6.0 Hz, 2H), 3.94 - 3.88 (m, 7H), 3.11 - 3.06 (m, 1H), 3.03 ( t, J = 5.6 Hz, 4H), 2.75 ( t, J = 6.8 Hz, 2H), 2.40 - 2.20 (m, 2H), 1.90 (s, 3H), 1.81 - 1.69 (m, 4H), 1.45 - 1.30 (m, 5H), 1.17 (t, J = 7.2 Hz, 1H) Example 243. Preparation of Compound 310a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1R,4r)-4- ((2R)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 6-yl)piperazin-1-yl)butan-2- yl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1R,4r)-4- ((2R*)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)piperazin-1-yl)butan-2- yl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of methyl (1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexane-1-carboxylate To a solution of methyl (1s,4s)-4-hydroxycyclohexane-1-carboxylate (5 g, 31.61 mmol, 1 equiv.) and 3-bromo-2-methyl-phenol (7.09 g, 37.93 mmol, 1.2 equiv.) in toluene (50 mL) was added 2-(tributyl-phosphanylidene)acetonitrile (9.15 g, 37.93 mmol, 1.2 equiv.). The mixture was stirred at 110 °C for 16 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by reverse-phase HPLC (with 0.1 % formic acid) to give methyl (1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexane-1- carboxylate (4 g, 12.22 mmol, 19.34% yield) as a yellow oil. 1 H NMR (400 MHz, CDCl3) δ = 7.14 (d, J = 8.0 Hz, 1H), 6.97 (t, J = 8.0 Hz, 1H), 6.80 (d, J = 8.4 Hz, 1H), 4.22 - 4.11 (m, 1H), 3.69 (s, 3H), 2.43 - 2.34 (m, 1H), 2.30 (s, 3H), 2.19 - 2.13 (m, 2H), 2.10 - 2.03 (m, 2H), 1.62 - 1.50 (m, 4H) Step B. Procedure for preparation of (1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexane- 1-carboxylic acid To a solution of methyl (1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexane-1-carboxylate (3.95 g, 12.07 mmol, 1 equiv.) in THF (100 mL) and H2O (10 mL) was added LiOH•H2O (1.52 g, 36.21 mmol, 3 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The pH of the residue was adjusted to ~3 by progressively adding diluted 1M HCl. The reaction mixture was partitioned between DCM (20 mL) and H 2 O (5 mL). The organic phase was separated, dried over Na 2 SO 4 , and concentrated under reduced pressure to give (1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexane-1-carboxylic acid (3.6 g, 10.84 mmol, 44.92% yield) as a yellow solid. MS (ESI) m/z: 314.1 [M+H] + . 1 H NMR (400 MHz, CDCl3) δ = 7.15 (d, J = 8.0 Hz, 1H), 6.98 (t, J = 8.0 Hz, 1H), 6.80 (d, J = 8.4 Hz, 1H), 4.21 - 4.15 (m, 1H), 2.48 - 2.38 (m, 1H), 2.30 (s, 3H), 2.19 - 2.08 (m, 4H), 1.65 - 1.52 (m, 4H) Step C. Procedure for preparation of (1r,4r)-4-(3-bromo-2-methylphenoxy)-N-methoxy- N-methylcyclohexane-1-carboxamide To a solution of (1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexane-1-carboxylic acid (3.4 g, 10.86 mmol, 1 equiv.) and N-methoxymethanamine (3.18 g, 32.57 mmol, 3 equiv. HCl) in DCM (5 mL) was added HATU (6.19 g, 16.28 mmol, 1.5 equiv.) and TEA (5.49 g, 54.28 mmol, 7.56 mL, 5 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 10% ethyl acetate/petroleum ether) to give (1r,4r)-4-(3-bromo-2- methylphenoxy)-N-methoxy-N-methylcyclohexane-1-carboxamide (3.3 g, 8.69 mmol, 80.04% yield, 93.8% purity) as a yellow solid. MS (ESI) m/z: 356.1 [M+H] + . 1 H NMR (400 MHz, CDCL 3 ) δ = 7.15 (d, J = 8.0 Hz, 1H), 6.99 (t, J = 8.0 Hz, 1H), 6.82 (d, J = 8.0 Hz, 1H), 4.17 (d, J = 11.6 Hz, 1H), 3.73 (s, 3H), 3.20 (s, 3H), 2.80 - 2.70 (m, 1H), 2.30 (s, 3H), 2.24 (dd, J = 2.8, 12.4 Hz, 2H), 1.96 - 1.89 (m, 2H), 1.72 - 1.62 (m, 2H), 1.57 - 1.46 (m, 2H) Step D. Procedure for preparation of 1-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)ethan-1-one A mixture of (1r,4r)-4-(3-bromo-2-methylphenoxy)-N-methoxy-N-methylcycloh exane-1- carboxamide (3.1 g, 8.70 mmol, 1 equiv.) in THF (30 mL) was degassed and purged with N2 three times. To this mixture was slowly added MeMgBr (3 M, 5.80 mL, 2 equiv.). The reaction mixture was stirred at -78 °C for 0.5 hours under N 2 atmosphere. The mixture was slowly warmed to 25 °C and stirred for 15.5 hours under N2 atmosphere. The reaction mixture was quenched by the addition of saturated NH4Cl (10 mL) and extracted with DCM (50 mL × 2). The combined organic layers were washed with brine (5 mL × 2), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give 1-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)ethan-1-one (2.7 g, crude) as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.16 - 7.13 (m, 1H), 7.10 - 7.02 (m, 2H), 4.27 (d, J = 4.8, 9.2 Hz, 1H), 2.45 - 2.38 (m, 1H), 2.25 - 2.21 (m, 3H), 2.12 (s, 3H), 2.09 - 2.03 (m, 2H), 1.95 - 1.86 (m, 2H), 1.47 - 1.35 (m, 4H) Step E. Procedure for preparation of ethyl (E)-3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)but-2-enoate A mixture of ethyl 2-diethoxyphosphorylacetate (2.16 g, 9.64 mmol, 1.91 mL, 1.2 equiv.) in THF (9 mL) was degassed and purged with N 2 three times. To this mixture was slowly added LiHMDS (1 M, 12.05 mL, 1.5 equiv.). The reaction mixture was stirred at -78 °C for 0.5 hours under N2 atmosphere. To the reaction mixture was added 1-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)ethan-1-one (2.5 g, 8.03 mmol, 1 equiv.) in THF (20 mL). The mixture was slowly warm to -25 °C and stirred for 2.5 hours under N 2 atmosphere. The reaction mixture was quenched by the addition of saturated NH4Cl (10 mL) and extracted with ethyl acetate (10 mL × 2). The combined organic layers were washed with brine (5 mL × 2), dried over Na 2 SO 4 , and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 8% ethyl acetate/petroleum ether) to give ethyl (E)-3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)but-2-e noate (1.7 g, 4.27 mmol, 53.11% yield) as a yellow oil. MS (ESI) m/z: 383.1 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 7.19 - 7.13 (m, 1H), 7.11 - 7.01 (m, 2H), 5.67 (s, 1H), 4.33 - 4.23 (m, 1H), 4.06 (q, J = 7.2 Hz, 2H), 3.33 - 3.27 (m, 1H), 2.21 (s, 2H), 2.10 (s, 6H), 1.74 (d, J = 6.8 Hz, 2H), 1.62 - 1.48 (m, 1H), 1.43 (t, J = 10.0 Hz, 3H), 1.19 (t, J = 7.2 Hz, 3H) Step F. Procedure for preparation of ethyl 3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butanoate To a solution of ethyl (E)-3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)but-2-e noate (1.7 g, 4.46 mmol, 1 equiv.) in EtOH (30 mL) was added PtO2 (126.55 mg, 445.84 μmol, 80% purity, 0.1 equiv.) under N 2 . The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H2 (15psi) at 25°C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give ethyl 3-((1r,4r)-4-(3- bromo-2-methylphenoxy)cyclohexyl)butanoate (1.7 g, crude) as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.16 - 7.11 (m, 1H), 7.10 - 6.98 (m, 2H), 4.26 - 4.14 (m, 1H), 4.06 (q, J = 7.2 Hz, 2H), 2.36 (dd, J = 5.2, 14.8 Hz, 1H), 2.29 - 2.15 (m, 3H), 2.11 - 2.02 (m, 3H), 1.87 - 1.75 (m, 1H), 1.68 (d, J = 10.4 Hz, 2H), 1.38 - 1.26 (m, 2H), 1.26 - 1.06 (m, 6H), 0.89 - 0.80 (m, 3H) Step G. Procedure for preparation of 3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butan-1-ol To a solution of ethyl 3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butanoate (1.7 g, 4.43 mmol, 1 equiv.) in THF (17 mL) was added LiAlH4 (168.33 mg, 4.43 mmol, 1 equiv.). The mixture was stirred at 0 °C for 2 hours. The reaction mixture was quenched with Na 2 SO 4 •10 H 2 O (100 mg) and slowly warmed to 20 °C. The mixture was filtered and washed with DCM (50 mL). Then the organic layers were concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butan-1-ol (600 mg, 1.64 mmol, 36.91% yield) as a yellow oil. Step H. Procedure for preparation of (R)-3-((1r,4R)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butan-1-ol 3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butan-1-ol (480 mg, 1.41 mmol, 1 equiv.) was separated by SFC to give (R)-3-((1r,4R)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butan- 1-ol (90 mg, 240.24 μmol, 17.08% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ = 7.15 - 7.10 (m, 1H), 7.09 - 7.00 (m, 2H), 4.31 (t, J = 5.2 Hz, 1H), 4.25 - 4.13 (m, 1H), 3.50 - 3.34 (m, 2H), 2.20 (s, 3H), 2.08 (d, J = 11.6 Hz, 2H), 1.69 - 1.60 (m, 2H), 1.56 - 1.42 (m, 2H), 1.38 - 1.28 (m, 3H), 1.21 - 1.10 (m, 3H), 0.80 (d, J = 6.8 Hz, 3H) Step I. Procedure for preparation of (R)-3-((1r,4R)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butanal To a solution of (R)-3-((1r,4R)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butan-1 -ol (90 mg, 263.71 μmol, 1 equiv.) in DCM (2 mL) was added DMP (111.85 mg, 263.71 μmol, 81.70 μL, 1 equiv.).The mixture was stirred at 0 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, petroleum ether : ethyl acetate = 6:1) to give (R)-3-((1r,4R)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butanal (50 mg, 130.87 μmol, 49.63% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 9.67 (s, 1H), 7.16 - 7.12 (m, 1H), 7.10 - 7.00 (m, 2H), 4.29 - 4.16 (m, 1H), 2.25 - 2.16 (m, 4H), 2.08 (d, J = 10.8 Hz, 2H), 2.01 - 1.90 (m, 1H), 1.69 (d, J = 12.0 Hz, 2H), 1.24 - 1.11 (m, 6H), 0.86 (d, J = 6.4 Hz, 3H) Step J. Procedure for preparation of 3-(6-(4-((R)-3-((1r,4R)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butyl)piperazin-1-yl)-1-methyl-1H-i ndazol-3-yl)piperidine- 2,6-dione A solution of (R)-3-((1r,4R)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butanal (40 mg, 117.90 μmol, 1 equiv.) and 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (42.46 mg, 129.69 μmol, 1.1 equiv.) in DCM (1 mL) at 25°C was stirred at 25 °C for 15.5 hours. NaBH(OAc)3 (74.96 mg, 353.70 μmol, 3 equiv.) was added to the reaction mixture, and the resulting mixture was stirred at 25 °C for 0.5 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , DCM : MeOH = 10:1) to give 3-(6-(4-((R)-3-((1r,4R)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butyl)piperazin-1-yl)-1-methyl-1H-i ndazol-3-yl)piperidine-2,6- dione (40 mg, 60.86 μmol, 51.62% yield) as a yellow oil. MS (ESI) m/z: 652.3 [M+H] + . Step K. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-((2R)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)butan-2-yl)cyclohexyl) oxy)-2- methylphenyl)picolinate To a solution of 3-(6-(4-((R)-3-((1r,4R)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butyl)piperazin-1-yl)-1-methyl-1H-i ndazol-3-yl)piperidine-2,6- dione (40 mg, 61.48 μmol, 1 equiv.) and tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)pyridine-2- carboxylate (45.19 mg, 73.77 μmol, 1.2 equiv.) in dioxane (2 mL) and H2O (0.1 mL) was added [2-(2-aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-buty l- phosphane;methanesulfonate (4.48 mg, 6.15 μmol, 0.1 equiv.) and KF (10.71 mg, 184.43 μmol, 4.32 μL, 3 equiv.). The mixture was stirred at 100 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 10% dichloromethane : methanol) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1R,4r)-4-((2R)-4- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1-yl)butan-2- yl)cyclohexyl)oxy)-2-methylphenyl)picolinate (35 mg, 29.06 μmol, 47.27% yield) as a yellow solid. MS (ESI) m/z: 529.2 [1/2M+H] + . Step L. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-((2R)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)butan-2-yl)cyclohexyl) oxy)-2- methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-((2R*)-4-(4-(3 -(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)butan-2-yl)cyclohexyl) oxy)-2- methylphenyl)picolinic acid] A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1R,4r)-4-((2R)-4-(4-(3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-6- yl)piperazin-1-yl)butan-2-yl)cyclohexyl)oxy)-2-methylphenyl) picolinate (35 mg, 33.13 μmol, 1 equiv.) in DCM (0.5 mL) and TFA (0.2 mL) was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1R,4r)-4-((2R)-4-(4-(3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-6- yl)piperazin-1-yl)butan-2-yl)cyclohexyl)oxy)-2-methylphenyl) picolinic acid [6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1R,4r)-4-((2R*)- 4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)p iperazin-1-yl)butan-2- yl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] (20.05 mg, 19.22 μmol, 58.02% yield) as a yellow solid. MS (ESI) m/z: 501.0 [M/2+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.97 - 12.74 (m, 1H), 10.87 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.57 (d, J = 8.8 Hz, 1H), 7.51 - 7.43 (m, 3H), 7.40 - 7.32 (m, 2H), 7.12 - 7.05 (m, 1H), 7.02 - 6.91 (m, 4H), 6.62 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.28 (dd, J = 5.2, 9.6 Hz, 1H), 4.22 - 4.15 (m, 2H), 3.97 (s, 4H), 3.92 (s, 5H), 3.64 (d, J = 11.2 Hz, 2H), 3.09 - 2.99 (m, 4H), 2.69 - 2.60 (m, 2H), 2.39 - 2.21 (m, 2H), 2.21 - 2.10 (m, 3H), 1.87 (s, 3H), 1.79 - 1.67 (m, 3H), 1.58 - 1.48 (m, 1H), 1.46 - 1.32 (m, 3H), 1.31 - 1.18 (m, 3H), 0.88 (d, J = 6.4 Hz, 3H) Example 244. Preparation of Compound 310b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1S,4r)-4- ((2S)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 6-yl)piperazin-1-yl)butan-2- yl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1S,4r)-4- ((2S*)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)piperazin-1-yl)butan-2- yl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of (S)-3-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butanal To a solution of (S)-3-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butan-1 -ol (110 mg, 322.31 μmol, 1 equiv.) in DCM (1 mL) was added DMP (150.38 mg, 354.55 μmol, 109.84 μL, 1.1 equiv.) at 0 °C. The mixture was stirred at 0 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , petroleum ether : ethyl acetate = 6:1) to give (S)-3-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butanal (80 mg, 211.73 μmol, 65.69% yield) as a yellow oil. Step B. Procedure for preparation of 3-(6-(4-((S)-3-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butyl)piperazin-1-yl)-1-methyl-1H-i ndazol-3-yl)piperidine- 2,6-dione To a solution of (S)-3-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butanal (70 mg, 206.33 μmol, 1.0 equiv.) in DCM (1 mL) was added 3-(1-methyl-6-piperazin-1-yl-indazol-3- yl) piperidine-2, 6-dione (97.59 mg, 268.23 μmol, 1.3 equiv., HCl) at 25 °C. The mixture was stirred at 25 °C for 16 hours. NaBH(OAc)3 (174.92 mg, 825.31 μmol, 4.0 equiv.) was then added to the mixture at 25 °C. The reaction mixture was stirred at 25 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH = 20:1) to give 3-(6-(4-((S)-3-((1r,4s)- 4-(3-bromo-2-methylphenoxy)cyclohexyl)butyl)piperazin-1-yl)- 1-methyl-1H-indazol-3- yl)piperidine-2,6-dione (100 mg, 153.69 μmol, 74.49% yield) as a yellow oil. MS (ESI) m/z: 650.3 [M+H] + . Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-((2S)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)butan-2-yl)cyclohexyl) oxy)-2- methylphenyl)picolinate A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) pyridine-2-carboxylate (101.68 mg, 165.99 μmol, 1.2 equiv.), 3-(6-(4-((S)-3-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butyl)piperazin-1-yl)-1-methyl-1H-i ndazol-3-yl)piperidine-2,6- dione (90 mg, 138.32 μmol, 1.0 equiv.), KF (24.11 mg, 414.97 μmol, 9.72 μL, 3.0 equiv.), and [2-(2-aminophenyl) phenyl]palladium (1 + );bis (1-adamantyl)-butyl- phosphane;methanesulfonate (20.15 mg, 27.66 μmol, 0.2 equiv.) in dioxane (1 mL) and H 2 O (0.1 mL) was degassed and purged with N2 three times. The mixture was stirred at 100 °C for 16 hours under a N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 10% DCM / MeOH) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-((2S)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)butan-2-yl)cyclohexyl) oxy)-2- methylphenyl)picolinate (120 mg, 96.33 μmol, 69.64% yield) as a yellow solid. MS (ESI) m/z: 1057.7 [M+H] + . Step D. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-((2S)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)butan-2-yl)cyclohexyl) oxy)-2- methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-((2S*)-4-(4-(3 -(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)butan-2-yl)cyclohexyl) oxy)-2- methylphenyl)picolinic acid] A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1s,4r)-4-((2S)-4-(4-(3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-6- yl)piperazin-1-yl)butan-2-yl)cyclohexyl)oxy)-2-methylphenyl) picolinate (110 mg, 104.14 μmol, 1.0 equiv.) in TFA (0.5 mL) and DCM (0.5 mL) was stirred at 25 °C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-((2S)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)butan-2-yl)cyclohexyl) oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1S,4r)- 4-((2S*)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indaz ol-6-yl)piperazin-1-yl)butan-2- yl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] (24.29 mg, 22.70 μmol, 21.80% yield) as a white solid. MS (ESI) m/z: 501.1 [M/2+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.85 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.52 - 7.42 (m, 4H), 7.40 - 7.32 (m, 2H), 7.11 - 7.04 (m, 1H), 6.98 - 6.89 (m, 3H), 6.84 (s, 1H), 6.61 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.29 - 4.13 (m, 2H), 3.95 - 3.85 (m, 5H), 3.22 (s, 2H), 3.02 (t, J = 5.6 Hz, 2H), 2.70 - 2.55 (m, 5H), 2.42 - 2.23 (m, 4H), 2.17 - 2.07 (m, 3H), 1.87 (s, 3H), 1.74 - 1.65 (m, 2H), 1.63 - 1.55 (m, 1H), 1.48 - 1.14 (m, 8H), 0.86 (d, J = 6.8 Hz, 3H) Example 245. Preparation of Compound 321a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- ((3R)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-y l)pyrrolidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1S,4r)-4-(4- ((3R*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7- yl)pyrrolidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of 4-((1r, 4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butanal A solution of DMSO (274.73 mg, 3.52 mmol, 274.73 μL, 4 equiv.) in DCM (20 mL) was added dropwise to a solution of (COCl) 2 (223.15 mg, 1.76 mmol, 153.89 μL, 2 equiv.) in DCM (20 mL) at -70 °C under N2 atmosphere. The mixture was stirred at -70 °C for 1 hour. After which time, 4-((1r, 4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butan-1-ol (300 mg, 879.04 μmol, 1 equiv.) in DCM (20 mL) was added dropwise at -70 °C. The solution was stirred for 1 hour at -70 °C. Then TEA (533.69 mg, 5.27 mmol, 734.10 μL, 6 equiv.) was added into the solution. The solution was stirred at -70 °C for 1 hour under N2 atmosphere. The mixture was diluted with H2O (10 mL) and extracted with DCM (10 mL × 3). The combined organic layers were filtered and concentrated under reduced pressure to give 4-((1r, 4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butanal (200 mg, crude) as a white solid. Step B. Procedure for preparation of 3-(7-((R)-1-(4-((1r, 4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butyl)pyrrolidin-3-yl)-1-methyl-1H- indazol-3-yl)piperidine- 2,6-dione To a solution of 4-((1r, 4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butanal (150 mg, 442.13 μmol, 1 equiv.) and 3-(1-methyl-7-((R)-pyrrolidin-3-yl)-1H-indazol-3-yl)piperidi ne-2,6-dione (165.73 mg, 530.56 μmol, 1.2 equiv.), NaOAc (36.27 mg, 442.13 μmol, 1 equiv.) in DCM (2 mL), was added NaBH(OAc) 3 (281.12 mg, 1.33 mmol, 3 equiv.). The mixture and stirred at 25 °C for 2.5 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography by prep-TLC (SiO2, DCM : MeOH =10:1) to give 3-(7-((R)-1-(4-((1r, 4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butyl)pyrrolidin-3-yl)-1-methyl-1H- indazol-3-yl)piperidine-2,6- dione (80 mg, 125.86 μmol, 28.47 % yield) as a white solid. MS (ESI) m/z: 635.4 [M+H] + . Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-(4-((3R)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)pyrrolidin-1-yl)butyl)cyclohexyl)oxy) -2-methylphenyl)picolinate A mixture of 3-(7-((R)-1-(4-((1r, 4S)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butyl)pyrrolidin-3-yl)-1-methyl-1H- indazol-3-yl)piperidine-2,6- dione (80.00 mg, 125.86 μmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)picolinate (92.51 mg, 151.03 μmol, 1.2 equiv.), [2-(2-aminophenyl)phenyl]palladium(1+); bis(1- adamantyl)-butyl-phosphane;methanesulfonate (9.17 mg, 12.59 μmol, 0.1 equiv.), and KF (1.5 M, 251.72 μL, 3 equiv.) in dioxane (3 mL) was degassed and purged with N 2 three times. The mixture was stirred at 100 °C for 12 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 10 % methanol : ethyl acetate) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1S,4r)-4-(4-((3R)- 3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pyrr olidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (98 mg, 94.11 μmol, 74.78 % yield) as a white solid. MS (ESI) m/z: 521.7 [M/2+H] + . Step D. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-((3R)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)pyrrolidin-1-yl)butyl)cyclohexyl)oxy) -2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3- (((1S,4r)-4-(4-((3R*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-7-yl)pyrrolidin- 1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1S,4r)-4-(4-((3R)-3-(3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-7- yl)pyrrolidin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)pico linate (90.00 mg, 86.43 μmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL) was stirred at 25 °C for 1 hour. The mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)- 3-(3-(((1s,4r)-4-(4-((3R)-3-(3-(2,6-dioxopiperidin-3-yl)-1-m ethyl-1H-indazol-7-yl)pyrrolidin- 1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r) -4-(4-((3R*)-3-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pyrrolidin-1-y l)butyl)cyclohexyl)oxy)-2- methylphenyl)picolinic acid] (43.68 mg, 42.41 μmol, 49.07 % yield) as an off-white solid. MS (ESI) m/z: 985.8 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.86 (d, J = 16.9 Hz, 1H), 10.89 (s, 1H), 8.14 (s, 1H), 8.01 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.58 (d, J = 8.0 Hz, 1H), 7.49 - 7.42 (m, 4H), 7.36 (td, J = 7.6, 11.5 Hz, 2H), 7.13 - 7.03 (m, 2H), 6.92 (dd, J = 8.8, 16.0 Hz, 2H), 6.62 (d, J = 7.2 Hz, 1H), 4.97 (s, 2H), 4.36 (dd, J = 4.8, 10.0 Hz, 1H), 4.24 - 4.16 (m, 5H), 3.91 (t, J = 5.8 Hz, 2H), 3.02 (t, J = 5.2 Hz, 4H), 2.87 - 2.74 (m, 2H), 2.70 - 2.60 (m, 2H), 2.45 - 2.27 (m, 3H), 2.18 - 1.99 (m, 4H), 1.87 (s, 3H), 1.78 (d, J = 12.4 Hz, 2H), 1.57 - 1.50 (m, 2H), 1.41 - 1.14 (m, 8H), 1.10 - 1.00 (m, 2H) Example 246. Preparation of Compound 321b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(4- ((3S)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-y l)pyrrolidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1R,4r)-4-(4- ((3S*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7- yl)pyrrolidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of 4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butanal A mixture of DMSO (457.88 mg, 5.86 mmol, 457.88 μL, 4.0 equiv.) in DCM (50 mL) was degassed and purged with N2 three times. To this solution was added oxalyl dichloride (371.91 mg, 2.93 mmol, 256.49 μL, 2.0 equiv.). The reaction mixture was stirred at -70 °C for 1 hour under N 2 atmosphere. A mixture of 4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butan-1-ol (500 mg, 1.47 mmol, 1.0 equiv.) in DCM (100 mL) was added, and the resulting mixture was stirred at -70 °C for 1.5 hours under N2 atmosphere. TEA (889.49 mg, 8.79 mmol, 1.22 mL, 6.0 equiv.) was added, and the mixture was stirred at -70 °C for 0.5 hour under N 2 atmosphere. The reaction mixture was diluted with H 2 O (50 mL) and extracted with DCM (50 mL × 2). The combined organic layers were concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 20 % ethyl acetate / petroleum ether) to give 4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butanal (200 mg, 560.03 μmol, 38.23 % yield) as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ = 9.66 (s, 1H), 7.17 - 7.11 (m, 1H), 7.10 - 7.00 (m, 2H), 4.23 (t, J = 10.1 Hz, 1H), 2.41 (t, J = 6.8 Hz, 2H), 2.21 (s, 3H), 2.04 (d, J = 10.8 Hz, 2H), 1.76 (d, J = 12.4 Hz, 2H), 1.58 - 1.50 (m, 2H), 1.41 - 1.30 (m, 2H), 1.29 - 1.23 (m, 1H), 1.22 - 1.15 (m, 2H), 1.08 - 0.98 (m, 2H) Step B. Procedure for preparation of 3-(7-((S)-1-(4-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butyl)pyrrolidin-3-yl)-1-methyl-1H- indazol-3-yl)piperidine- 2,6-dione To a solution of 4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butanal (100 mg, 294.75 μmol, 1.0 equiv.) in DCM (2 mL) was added 3-[1-methyl-7-[(3S)-pyrrolidin-3- yl]indazol-3-yl]piperidine-2,6-dione (110.49 mg, 353.70 μmol, 1.2 equiv.). The mixture was stirred at 25 °C for 1 hour. NaBH(OAc)3 (187.41 mg, 884.26 μmol, 3.0 equiv.) was added to the mixture which was stirred for 0.5 hour. The reaction mixture was diluted with H 2 O (5 mL) and extracted with DCM (5 mL × 2). The combined organic layers were concentrated under reduced pressure to give 3-(7-((S)-1-(4-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butyl)pyrrolidin-3-yl)-1-methyl-1H- indazol-3-yl)piperidine-2,6- dione (200 mg, 289.48 μmol, 98.21 % yield) as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ = 10.88 (s, 1H), 7.61 - 7.52 (m, 1H), 7.47 - 7.41 (m, 1H), 7.16 - 7.00 (m, 4H), 4.35 (dd, J = 5.6, 9.2 Hz, 1H), 4.22 (s, 3H), 3.20 (s, 1H), 2.68 - 2.61 (m, 4H), 2.33 (s, 1H), 2.21 (s, 3H), 2.17 - 2.12 (m, 1H), 2.07 - 2.02 (m, 2H), 1.91 (s, 5H), 1.78 (d, J = 10.4 Hz, 2H), 1.58 - 1.49 (m, 2H), 1.43 - 1.17 (m, 10H), 1.09 - 0.98 (m, 2H) MS (ESI) m/z: 635.5 [M+H] + . Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(4-((3S)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)pyrrolidin-1-yl)butyl)cyclohexyl)oxy) -2-methylphenyl)picolinate To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin e-2-carboxylate (106.00 mg, 173.06 μmol, 1.1 equiv.), 3-(7-((S)-1-(4-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butyl)pyrrolidin-3-yl)-1-methyl-1H- indazol-3-yl)piperidine-2,6- dione (100 mg, 157.32 μmol, 1.0 equiv.), and KF (1.5 M, 314.65 μL, 3.0 equiv.) in dioxane (2 mL), was added [2-(2-aminophenyl)phenyl]palladium(1 + );bis(1-adamantyl)-butyl-phosphane; methanesulfonate (22.91 mg, 31.46 μmol, 0.2 equiv.). The mixture was stirred and heated at 100 °C for 1.5 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 10 % DCM / MeOH gradient) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(4-((3S)-3-(3-(2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-7- yl)pyrrolidin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)pico linate (90 mg, crude) as a white solid. MS (ESI) m/z: 521.7 [M+H] + . Step D. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(4-((3S)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)pyrrolidin-1-yl)butyl)cyclohexyl)oxy) -2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3- (((1R,4r)-4-(4-((3S*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-7-yl)pyrrolidin- 1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(4-((3S)-3-(3-(2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-7- yl)pyrrolidin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)pico linate (90.00 mg, 86.43 μmol, 1.0 equiv.) in DCM (1 mL) was added TFA (1 mL). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(4-((3S)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)pyrrolidin-1-yl)butyl)cyclohexyl)oxy) -2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1R,4r)-4-(4- ((3S*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7- yl)pyrrolidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] (39.67 mg, 39.86 μmol, 46.12% yield) as a white solid. MS (ESI) m/z: 985.8 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.89 (s, 1H), 8.03 (d, J = 8.4 Hz, 1H), 7.79 (d, J = 8.4 Hz, 1H), 7.61 (t, J = 6.4 Hz, 2H), 7.50 - 7.41 (m, 4H), 7.40 - 7.31 (m, 2H), 7.13 (t, J = 7.6 Hz, 1H), 7.10 - 7.03 (m, 1H), 6.99 - 6.88 (m, 2H), 6.61 (d, J = 7.2 Hz, 1H), 4.97 (s, 2H), 4.40 - 4.33 (m, 1H), 4.29 - 4.14 (m, 5H), 3.94 - 3.88 (m, 2H), 3.08 - 2.97 (m, 4H), 2.61 (s, 3H), 2.32 (d, J = 1.6 Hz, 2H), 2.22 - 2.03 (m, 5H), 1.87 (s, 3H), 1.78 (d, J = 14.0 Hz, 2H), 1.63 - 1.54 (m, 2H), 1.45 - 1.30 (m, 5H), 1.29 - 1.21 (m, 3H), 1.17 - 0.97 (m, 3H) Example 247. Preparation of Compound 320a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3- ((3R)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-y l)pyrrolidin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1S,4r)-4-(3- ((3R*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7- yl)pyrrolidin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of tert-butyl (3R)-3-[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-7-yl]pyrrolidine-1-carboxylate A mixture of tert-butyl (3R)-3-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-7- yl]pyrrolidine-1-carboxylate (4.30 g, 7.28 mmol, 1 equiv.), Pd(OH) 2 (1 g, 7.12 mmol, 0.978 equiv.), AcOH (437.14 mg, 7.28 mmol, 416.72 μL, 1 equiv.), and Pd/C (1 g, 10% purity, 1.00 equiv.) in THF (25 mL) and EtOH (25 mL) was degassed and purged with H2 three times. The mixture was stirred at 20 °C for 16 hours under H 2 (15 Psi) atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give tert-butyl (3R)-3-[3-(2,6-dioxo- 3-piperidyl)-1-methyl-indazol-7-yl]pyrrolidine-1-carboxylate (2.7 g, 6.55 mmol, 89.92% yield) as a gray gum. MS (ESI) m/z: 357.3 [M+H] + Step B. Procedure for preparation of 3-[1-methyl-7-[(3R)-pyrrolidin-3-yl]indazol-3- yl]piperidine-2,6-dione To a solution of tert-butyl (3R)-3-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7- yl]pyrrolidine-1-carboxylate (2.7 g, 6.55 mmol, 1 equiv.) in DCM (20 mL) was added HCl/dioxane (4 M, 1.64 mL, 1 equiv.).The mixture was stirred at 20 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was triturated with MeCN at 20 °C for 10 min and filtered to give 3-[1-methyl-7- [(3R)-pyrrolidin-3-yl]indazol-3-yl]piperidine-2,6-dione (2.79 g, crude, HCl) as a yellow solid. MS (ESI) m/z: 313.1 [M+H] + . 1 H NMR (400 MHz, D 2 O) δ = 7.58 (d, J = 8.0 Hz, 1H), 7.38 (d, J = 7.2 Hz, 1H), 7.18 (t, J = 7.6 Hz, 1H), 4.40 (dd, J = 5.2, 11.6 Hz, 1H), 4.31 (q, J = 8.0 Hz, 1H), 4.19 (s, 3H), 3.76 (dd, J = 7.8, 11.6 Hz, 1H), 3.60 - 3.37 (m, 3H), 2.85 - 2.68 (m, 2H), 2.51 - 2.38 (m, 2H), 2.28 - 2.17 (m, 2H). Step C. Procedure for preparation of 3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propanal A mixture of 3-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propan-1-ol (800 mg, 2.44 mmol, 1.0 equiv.) and DMP (2.07 g, 4.89 mmol, 1.51 mL, 2.0 equiv.) in DCM (10 mL) was degassed and purged with N2 three times at 0 °C. The mixture was stirred at 25 °C for 6 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 56% ethyl acetate / petroleum ether) to give 3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propanal (600 mg, 1.84 mmol, 75.46% yield) as a white solid. Step D. Procedure for preparation of 3-(7-((R)-1-(3-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propyl)pyrrolidin-3-yl)-1-methyl-1H -indazol-3- yl)piperidine-2,6-dione A solution of 3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propanal (100 mg, 307.47 μmol, 1.0 equiv.), 3-[1-methyl-7-[(3R)-pyrrolidin-3-yl]indazol-3-yl]piperidine- 2,6-dione (105.65 mg, 338.21 μmol, 1.1 equiv.), NaOAc (37.83 mg, 461.20 μmol, 1.5 equiv.), and NaBH(OAc) 3 (325.82 mg, 1.54 mmol, 5.0 equiv.) in DCM (2 mL) was degassed and purged with N2 three times. The mixture was stirred at 25 °C for 3 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 8% MeOH / DCM) to give 3-(7- ((R)-1-(3-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)pro pyl)pyrrolidin-3-yl)-1- methyl-1H-indazol-3-yl)piperidine-2,6-dione (70 mg, 112.61 μmol, 36.63% yield) as a white solid. MS (ESI) m/z: 621.2 [M+H] + Step E. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-(3-((3R)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)pyrrolidin-1-yl)propyl)cyclohexyl)oxy )-2- methylphenyl)picolinate A mixture of 3-(7-((R)-1-(3-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propyl)pyrrolidin-3-yl)-1-methyl-1H -indazol-3-yl)piperidine-2,6- dione (70 mg, 112.61 μmol, 1.0 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)pyridine-2- carboxylate (82.78 mg, 135.13 μmol, 1.2 equiv.), Ad 2 nBuP Pd G 3 (cataCXium® A Pd G 3 ) (16.40 mg, 22.52 μmol, 0.2 equiv.), and KF (19.63 mg, 337.83 μmol, 7.91 μL, 3.0 equiv.) in dioxane (2 mL) and H2O (0.2 mL) was degassed and purged with N2 three times. The mixture was stirred at 80 °C for 12 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH = 10:1) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-(3-((3R)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)pyrrolidin-1-yl)propyl)cyclohexyl)oxy )-2-methylphenyl)picolinate (60 mg, 58.41 μmol, 51.87% yield) as a yellow oil. MS (ESI) m/z: 514.2 [1/2M+H] + Step F. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-((3R)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)pyrrolidin-1-yl)propyl)cyclohexyl)oxy )-2- methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-(3-((3R*)-3-(3 -(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)pyrrolidin-1-yl)propyl)cyclohexyl)oxy )-2- methylphenyl)picolinic acid] To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1S,4r)-4-(3-((3R)-3-(3-(2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-7- yl)pyrrolidin-1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl)pic olinate (60 mg, 58.41 μmol, 1.0 equiv.) in DCM (1 mL) was added TFA (0.3 mL). The mixture was stirred at 40 °C for 15 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-((3R)-3 -(3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)pyrrolidin-1-yl)propyl)cyclohexyl)oxy )-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1S,4r)- 4-(3-((3R*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indaz ol-7-yl)pyrrolidin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] (15.57 mg, 15.54 μmol, 26.60% yield) as a white solid. MS (ESI) m/z: 971.5 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 13.42 - 11.72 (m, 2H), 10.89 (s, 1H), 8.16 (s, 1H), 8.06 - 7.97 (m, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 6.4 Hz, 1H), 7.55 (d, J = 8.0 Hz, 1H), 7.49 - 7.40 (m, 4H), 7.39 - 7.31 (m, 2H), 7.12 - 7.01 (m, 2H), 6.96 - 6.86 (m, 2H), 6.63 (d, J = 6.4 Hz, 1H), 4.97 (s, 2H), 4.37 - 4.35 (m, 1H), 4.21 (s, 3H), 3.91 - 3.89 (m, 2H), 3.06 - 2.97 (m, 3H), 2.69 - 2.60 (m, 4H), 2.39 - 2.30 (m, 4H), 2.21 - 2.11 (m, 2H), 2.09 - 2.06 (m, 2H), 1.83 - 1.77 (m, 3H), 1.85 - 1.69 (m, 3H), 1.54 (d, J = 7.6 Hz, 2H), 1.42 - 1.19 (m, 6H), 1.12 - 0.99 (m, 2H). Example 248. Preparation of Compound 320b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(3- ((3S)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-y l)pyrrolidin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1R,4r)-4-(3- ((3S*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7- yl)pyrrolidin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of tert-butyl 3-[3-(2,6-dibenzyloxy-3-pyridyl)-1- methyl-indazol-7-yl]pyrrolidine-1-carboxylate A mixture of 7-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (20 g, 39.97 mmol, 1.0 equiv.), tert-butyl 3-bromopyrrolidine-1-carboxylate (10.00 g, 39.97 mmol, 1.0 equiv.), Ir[dF(CF 3 )ppy] 2 (dtbpy)(PF 6 ) (448.42 mg, 399.69 μmol, 0.01 equiv.), NiCl 2 .dtbbpy (795.38 mg, 2.00 mmol, 0.05 equiv.), quinuclidine (444.40 mg, 4.00 mmol, 0.1 equiv.), and TMP (11.29 g, 79.94 mmol, 13.57 mL, 2.0 equiv.) in MeCN (100 mL) was degassed three times. The reaction vial was then sealed with parafilm, placed 2 cm away from one blue LED, and irradiated at 25 °C for 14 hours. The mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by reverse-phase HPLC to give tert-butyl 3-[3-(2,6- dibenzyloxy-3-pyridyl)-1-methyl-indazol-7-yl]pyrrolidine-1-c arboxylate (8.73 g, 13.48 mmol, 33.73% yield) as a brown oil. MS (ESI) m/z: 591.4 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 8.14 (s, 1H), 7.84 (d, J = 8.0 Hz, 1H), 7.61 - 7.13 (m, 12H), 7.00 (t, J = 7.6 Hz, 1H), 6.59 (d, J = 8.0 Hz, 1H), 5.43 (s, 4H), 4.30 (s, 3H), 4.25 - 4.17 (m, 1H), 3.75 (t, J = 8.8 Hz, 1H), 3.49 - 3.39 (m, 3H), 2.33 (s, 1H), 2.12 - 2.04 (m, 1H), 1.43 (s, 9H) Step B. Procedure for preparation of tert-butyl (3R)-3-[3-(2,6-dibenzyloxy-3-pyridyl)-1- methyl-indazol-7-yl]pyrrolidine-1-carboxylate and tert-butyl (3S)-3-[3-(2,6-dibenzyloxy- 3-pyridyl)-1-methyl-indazol-7-yl]pyrrolidine-1-carboxylate The stereoisomers of tert-butyl 3-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-7- yl]pyrrolidine-1-carboxylate (9.59 g, 16.23 mmol, 1 equiv.) were separated by prep-HPLC to afford tert-butyl (3S)-3-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-7-yl] pyrrolidine-1- carboxylate (4.5 g, 7.55 mmol, 46.51% yield) as a brown oil and tert-butyl (3R)-3-[3-(2,6- dibenzyloxy-3-pyridyl)-1-methyl-indazol-7-yl]pyrrolidine-1-c arboxylate (4.3 g, 7.08 mmol, 43.63% yield) as a brown oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.84 (d, J = 8.0 Hz, 1H), 7.53 - 7.20 (m, 12H), 7.00 (t, J = 7.6 Hz, 1H), 6.59 (d, J = 8.0 Hz, 1H), 5.43 (s, 4H), 4.30 (s, 3H), 4.25 - 4.16 (m, 1H), 3.80 - 3.69 (m, 1H), 3.50 - 3.38 (m, 3H), 2.33 (s, 1H), 2.07 (s, 1H), 1.43 (s, 9H). 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.84 (d, J = 8..0 Hz, 1H), 7.58 - 7.16 (m, 12H), 7.00 (t, J = 7.6 Hz, 1H), 6.59 (d, J = 8.0 Hz, 1H), 5.43 (s, 4H), 4.30 (s, 3H), 4.24 - 4.14 (m, 1H), 3.80 - 3.70 (m, 1H), 3.49 - 3.38 (m, 3H), 2.32 (s, 1H), 2.14 - 2.00 (m, 1H), 1.48 - 1.37 (m, 9H) Step C. Procedure for preparation of tert-butyl (3S)-3-[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-7-yl]pyrrolidine-1-carboxylate To a solution of tert-butyl (3S)-3-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-7- yl]pyrrolidine-1-carboxylate (4.5 g, 7.62 mmol, 1 equiv.) and Pd(OH)2 (937.50 mg, 6.68 mmol, 0.876 equiv.), Pd/C (937.50 mg, 880.94 μmol, 10% purity, 0.116 equiv.), and AcOH (457.47 mg, 7.62 mmol, 436.10 μL, 1 equiv.) in THF (25 mL) and EtOH (25 mL) was degassed and purged with H2 three times. The mixture was stirred at 20 °C for 16 hours under H2 atmosphere. The mixture was filtered and concentrated under reduced pressure to give tert-butyl (3S)-3-[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-7-yl]pyrrolidine-1- carboxylate (3 g, 7.27 mmol, 95.47% yield) as a yellow gum. MS (ESI) m/z: 357.3 [M+H] + Step D. Procedure for preparation of 3-[1-methyl-7-[(3S)-pyrrolidin-3-yl]indazol-3- yl]piperidine-2,6-dion To a solution of tert-butyl (3S)-3-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-7- yl]pyrrolidine-1-carboxylate (3 g, 7.27 mmol, 1 eq) in DCM (20 mL) was added HCl/dioxane (4 M, 17.14 mL, 9.43 eq). The mixture was stirred at 20 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was triturated with MeCN (10 mL) at 20 °C for 10 min then filtered to give 3-[1-methyl-7-[(3S)- pyrrolidin-3-yl]indazol-3-yl]piperidine-2,6-dione (2.23 g, 6.72 mmol, 92.44% yield) as a white solid. MS (ESI) m/z: 313.0 [M+H] + . 1 H NMR (400 MHz, D2O) δ = 7.61 (d, J = 7.6 Hz, 1H), 7.40 (d, J = 7.2 Hz, 1H), 7.20 (t, J = 7.6 Hz, 1H), 4.43 (dd, J = 5.2, 11.6 Hz, 1H), 4.38 - 4.31 (m, 1H), 4.21 (s, 3H), 3.78 (dd, J = 7.6, 11.6 Hz, 1H), 3.63 - 3.53 (m, 1H), 3.52 - 3.38 (m, 2H), 2.86 - 2.69 (m, 2H), 2.58 - 2.38 (m, 2H), 2.33 - 2.16 (m, 2H). Step E. Procedure for preparation of 3-(7-((S)-1-(3-((1r,4R)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propyl)pyrrolidin-3-yl)-1-methyl-1H -indazol-3- yl)piperidine-2,6-dione A mixture of 3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propanal (100.00 mg, 307.47 μmol, 1 equiv.), 3-(1-methyl-7-((S)-pyrrolidin-3-yl)-1H-indazol-3-yl)piperidi ne-2,6- dione (115.25 mg, 368.96 μmol, 1.2 equiv.), and NaBH(OAc)3 (195.49 mg, 922.40 μmol, 3 equiv.) in DCM (4 mL) was degassed and purged with N 2 three times. The mixture was stirred at 25 °C for 10 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH = 10:1) to give 3-(7-((S)-1-(3-((1r,4R)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propyl)pyrrolidin-3-yl)-1-methyl-1H -indazol-3-yl)piperidine-2,6- dione (135 mg, 193.29 μmol, 62.87% yield) as a white oil. MS (ESI) m/z: 623.4 [M+H] + Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-((3S)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)pyrrolidin-1-yl)propyl)cyclohexyl)oxy )-2- methylphenyl)picolinate A mixture of 3-(7-((S)-1-(3-((1r,4R)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propyl)pyrrolidin-3-yl)-1-methyl-1H -indazol-3-yl)piperidine-2,6- dione (135 mg, 217.18 μmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)picolinate (172.94 mg, 282.33 μmol, 1.3 equiv.), Ad 2 nBuP Pd G 3 (cataCXium® A Pd G 3 ) (31.63 mg, 43.44 μmol, 0.2 equiv.), and KF (1.5 M, 434.36 μL, 3 equiv.) in dioxane (5 mL) was degassed and purged with N2 three times. The mixture was stirred at 80 °C for 10 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 - 9% ethyl acetate / methanol) to give compound tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-((3S)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)pyrrolidin-1-yl)propyl)cyclohexyl)oxy )-2-methylphenyl)picolinate (150 mg, 146.02 μmol, 67.23% yield) as a brown oil. MS (ESI) m/z: 514.7 [M/2+H] + Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-((3S)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)pyrrolidin-1-yl)propyl)cyclohexyl)oxy )-2- methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-(3-((3S*)-3-(3 -(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)pyrrolidin-1-yl)propyl)cyclohexyl)oxy )-2- methylphenyl)picolinic acid] A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1R,4r)-4-(3-((3S)-3-(3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-7- yl)pyrrolidin-1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl)pic olinate (140 mg, 136.28 μmol, 1 equiv.) in TFA (1.5 mL) and DCM (1.5 mL) was degassed and purged with N2 three times. The mixture was stirred at 25 °C for 2 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)- 3-(3-(((1r,4r)-4-(3-((3S)-3-(3-(2,6-dioxopiperidin-3-yl)-1-m ethyl-1H-indazol-7-yl)pyrrolidin- 1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r) -4-(3-((3S*)-3-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pyrrolidin-1-y l)propyl)cyclohexyl)oxy)-2- methylphenyl)picolinic acid] (78.67 mg, 79.21 μmol, 58.13% yield) as a white solid, MS (ESI) m/z: 486.5 [M/2+H] + . 1H NMR (400 MHz, DMSO-d 6 ) δ = 13.14 - 12.29 (m, 1H), 10.89 (s, 1H), 8.14 (s, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.4 Hz, 1H), 7.65 - 7.58 (m, 2H), 7.50 - 7.41 (m, 4H), 7.40 - 7.32 (m, 2H), 7.13 (t, J = 7.6 Hz, 1H), 7.06 (t, J = 8.0 Hz, 1H), 6.93 (dd, J = 8.8, 15.6 Hz, 2H), 6.62 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.37 (dd, J = 5.2, 10.0 Hz, 1H), 4.31 - 4.18 (m, 4H), 3.91 (r t, J = 5.4 Hz, 2H), 3.62 - 3.46 (m, 2H), 3.21 (s, 2H), 3.01 (d, J = 5.6 Hz, 4H), 2.69 - 2.61 (m, 2H), 2.43 - 2.29 (m, 2H), 2.19 - 2.06 (m, 4H), 1.87 (s, 3H), 1.80 (d, J = 11.6 Hz, 2H), 1.63 (s, 2H), 1.40 - 1.24 (m, 5H), 1.14 - 1.01 (m, 2H) Example 249: Preparation of Compound 305 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(3-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)propyl)pi peridin-4-yl)propoxy)-2- methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 4-(3-(3-bromo-2- methylphenoxy)propyl)piperidine-1-carboxylate A mixture of 3-bromo-2-methylphenol (2 g, 10.69 mmol, 1 equiv.), tert-butyl 4-(3- hydroxypropyl) piperidine-1-carboxylate (3.12 g, 12.83 mmol, 1.2 equiv.), and 2- (tributyl- phosphanylidene) acetonitrile (5.16 g, 21.39 mmol, 2 equiv.) in toluene. (30 mL) was degassed and purged with N 2 three times. The mixture was stirred at 120 °C for 12 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 7 % ethyl acetate / petroleum ether) to give tert-butyl 4-(3-(3-bromo-2-methylphenoxy)propyl)piperidine-1- carboxylate (4.31 g, 10.45 mmol, 97.74 % yield) as a white solid. MS (ESI) m/z: 357.9 [M+H] + . Step B. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(tert-butoxycarbonyl )piperidin-4-yl)propoxy)-2- methylphenyl)picolinate A mixture of tert-butyl 4-(3-(3-bromo-2-methylphenoxy)propyl)piperidine-1-carboxylat e (500 mg, 1.21 mmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)picolinate (891.28 mg, 1.46 mmol, 1.2 equiv.), Ad 2 nBuP Pd G 3 (cataCXium ® A Pd G 3 ) (264.92 mg, 363.76 μmol, 0.3 equiv.), and KF (105.67 mg, 1.82 mmol, 42.61 μL, 1.5 equiv.) in dioxane (2 mL) and H2O (0.2 mL) was degassed and purged with N2 three times. The mixture was stirred at 80 ℃ for 1 hour under N2 atmosphere. The reaction was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 17 % ethyl acetate / petroleum ether) to give tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(te rt-butoxycarbonyl)piperidin-4- yl)propoxy)-2-methylphenyl)picolinate (800 mg, 977.96 μmol, 80.65 % yield) as a white solid. MS (ESI) m/z: 818.9 [M+H] + . Step C. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(2-methyl-3-(3-(piperidin-4-y l)propoxy)phenyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-(1-(tert-butoxycarbonyl)piperidin-4-yl)propoxy)- 2-methylphenyl)picolinate (800 mg, 977.96 μmol, 1 equiv.) in TFA (4 mL) and DCM (4 mL) was stirred at 25 ℃ for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(2-methyl-3-(3- (piperidin-4-yl)propoxy)phenyl)picolinic acid (460 mg, 902.68 μmol, 16.74 % yield) as a yellow oil. MS (ESI) m/z: 662.6 [M+H] + . Step D. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-6-(3- ((tetrahydro-2H-pyran-2-yl)oxy)prop-1-yn-1-yl)-1H-indazole A mixture of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-bromo-1-methyl-1H-indaz ole (3 g, 6.00 mmol, 1 equiv.), 2-(prop-2-yn-1-yloxy)tetrahydro-2H-pyran (840.43 mg, 6.00 mmol, 842.96 μL, 1 equiv.), CuI (228.36 mg, 1.20 mmol, 0.2 equiv.), Pd(PPh3)4 (692.80 mg, 599.54 μmol, 0.1 equiv.), and DMF (20 mL) in DIEA (20 mL) was degassed and purged with N 2 three times. The mixture was stirred at 80 ℃ for 16 hours under N2 atmosphere. The reaction was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 13 % ethyl acetate / petroleum ether) to give 3-(2,6- bis(benzyloxy)pyridin-3-yl)-1-methyl-6-(3-((tetrahydro-2H-py ran-2-yl)oxy)prop-1-yn-1-yl)- 1H-indazole (1.2 g, 2.14 mmol, 35.76 % yield) as a yellow oil. MS (ESI) m/z: 560.3 [M+H] + . Step E. Procedure for preparation of 3-(1-methyl-6-(3-((tetrahydro-2H-pyran-2- yl)oxy)propyl)-1H-indazol-3-yl)piperidine-2,6-dione A mixture of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-6-(3-((tetrahydr o-2H-pyran-2- yl)oxy)prop-1-yn-1-yl)-1H-indazole (800 mg, 1.43 mmol, 1 equiv.), Pd/C (450 mg, 1.43 mmol, 10 % purity, 1 equiv.), and Pd(OH)2 (450 mg, 1.43 mmol, 30 % purity, 1 equiv.) in THF (4 mL) was degassed and purged with H 2 three times. The mixture was stirred at 25 ℃ for 12 hours under H 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, petroleum ether : ethyl acetate = 1 : 1) to give 3-(1-methyl-6-(3-((tetrahydro-2H-pyran-2-yl)oxy)propyl)-1H-i ndazol- 3-yl)piperidine-2,6-dione (300 mg, 778.30 μmol, 54.45 % yield) as a white solid. MS (ESI) m/z: 386.3 [M+H] + . Step F. Procedure for preparation of 3-(6-(3-hydroxypropyl)-1-methyl-1H-indazol-3- yl)piperidine-2,6-dione A mixture of 3-(1-methyl-6-(3-((tetrahydro-2H-pyran-2-yl)oxy)propyl)-1H-i ndazol-3- yl)piperidine-2,6-dione (300 mg, 778.30 μmol, 1 equiv.) in MeOH (4 mL) was added HCl (12 M, 12.97 μL, 0.2 equiv.) at 0 °C. The mixture was stirred at 25 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give 3-(6-(3-hydroxypropyl)- 1-methyl-1H-indazol-3-yl)piperidine-2,6-dione (340 mg, crude) as a yellow oil. MS (ESI) m/z: 302.0 [M+H] + . Step G. Procedure for preparation of 3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 6-yl)propanal A mixture of 3-(6-(3-hydroxypropyl)-1-methyl-1H-indazol-3-yl)piperidine-2 ,6-dione (220 mg, 730.07 μmol, 1 equiv.) and DMP (464.48 mg, 1.10 mmol, 339.29 μL, 1.5 equiv.) in DCM (4 mL) was stirred at 25 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 20 % ethyl acetate / petroleum ether) to give 3-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)propanal (240 mg, 577.30 μmol, 79.07 % yield) as a yellow solid. MS (ESI) m/z: 300.1 [M+H] + . Step H. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(3-(3-(2,6-dioxopipe ridin-3-yl)-1-methyl-1H- indazol-6-yl)propyl)piperidin-4-yl)propoxy)-2-methylphenyl)p icolinic acid A mixture of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(3-(piperidin-4-yl)propoxy)phenyl)picolinic acid (241.20 mg, 364.46 μmol, 1.0 equiv.), 3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)prop anal (120 mg, 400.90 μmol, 1.1 equiv.), and 4A MS (30 mg) in DCM (6 mL) was stirred at 25 °C for 12 hours. To the reaction mixture was added NaBH(OAc) 3 (154.49 mg, 728.92 μmol, 2 equiv.). The resulting mixture was stirred at 25 ℃ for 0.5 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(3- (3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)propyl )piperidin-4-yl)propoxy)-2- methylphenyl)picolinic acid (89.66 mg, 89.27 μmol, 24.49 % yield) as a yellow solid. MS (ESI) m/z: 945.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.88 (s, 1H), 8.15 (d, J = 2.4 Hz, 1H), 8.01 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.4 Hz, 1H), 7.62 (dd, J = 4.0, 6.8 Hz, 2H), 7.46 - 7.39 (m, 4H), 7.39 - 7.30 (m, 2H), 7.07 (t, J = 7.2 Hz, 1H), 6.99 (d, J = 8.8 Hz, 1H), 6.94 - 6.90 (m, 1H), 6.86 (d, J = 8.0 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 5.01 - 4.94 (m, 2H), 4.33 (dd, J = 5.2, 9.6 Hz, 1H), 3.95 (s, 5H), 3.91 (t, J = 5.6 Hz, 2H), 3.12 (d, J = 3.6 Hz, 2H), 3.02 (d, J = 4.8 Hz, 2H), 2.72 (d, J = 7.2 Hz, 3H), 2.69 - 2.61 (m, 5H), 2.20 - 2.14 (m, 1H), 1.89 (s, 5H), 1.73 (d, J = 2.4 Hz, 4H), 1.48 - 1.30 (m, 4H), 1.25 - 1.14 (m, 2H) Example 250. Preparation of Compound 307a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4- ((2S)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 6-yl)piperazin-1-yl)-2- methylbutyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1S,4r)-4- ((2S*)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)piperazin-1-yl)-2- methylbutyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of methyl 2-(4-hydroxycyclohexyl)acetate To a solution of 2-(4-hydroxycyclohexyl)acetic acid (10.0 g, 63.21 mmol, 1 equiv.) in MeOH (100 mL) was added H2SO4 (18.60 g, 189.64 mmol, 10.11 mL, 3.0 equiv.). The mixture was stirred at 70 °C for 3 hours. The reaction mixture was diluted with H 2 O (100 mL) and extracted with DCM (150 mL × 3). The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give methyl 2-(4-hydroxycyclohexyl)acetate (10.0 g, crude) as a yellow oil. Step B. Procedure for preparation of methyl 2-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]acetate A mixture of methyl 2-(4-hydroxycyclohexyl)acetate (10 g, 58.06 mmol, 1.0 equiv.), 3-bromo- 2-methyl-phenol (21.72 g, 116.13 mmol, 2.0 equiv.), and 2-(tributyl- phosphanylidene)acetonitrile (16.82 g, 69.68 mmol, 1.2 equiv.) in toluene (100 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 120 °C for 16 hours under a N 2 atmosphere. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~4% ethyl acetate/petroleum ether) to give methyl 2-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]acetate (14.5 g, crude) as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.16 - 7.11 (m, 1H), 7.06 (t, J = 8.0 Hz, 1H), 6.82 - 6.78 (m, 1H), 4.63 (s, 1H), 3.60 - 3.57 (m, 3H), 2.26 (s, 3H), 2.22 (d, J = 13.2 Hz, 2H), 1.93 - 1.74 (m, 3H), 1.62 - 1.48 (m, 3H), 1.46 - 1.22 (m, 3H) Step C. Procedure for preparation of 2-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]acetic acid To a solution of methyl 2-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]acetate (14.00 g, 41.03 mmol, 1.0 equiv.) in THF (100 mL) and H 2 O (30 mL) was added LiOH•H 2 O (5.16 g, 123.08 mmol, 3.0 equiv.). The mixture was stirred at 30 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to remove THF (100 mL). The residue was diluted with H 2 O (20 mL), and its pH was adjusted with HCl (1 M) to 4. The resulting mixture was extracted with EtOAc (100 mL × 3). The combined organic layers were washed with brine (100 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate = 50/1 to 10/1) to give 2-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]acetic acid (7.75 g, crude) as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.03 (s, 1H), 7.14 - 7.11 (m, 1H), 7.07 (t, J = 8.0 Hz, 1H), 7.00 - 6.96 (m, 1H), 4.64 (s, 1H), 2.26 (s, 3H), 2.15 (d, J = 7.2 Hz, 2H), 1.92 - 1.84 (m, 2H), 1.83 - 1.71 (m, 2H), 1.55 - 1.50 (m, 2H), 1.43 - 1.25 (m, 3H) Step D. Procedure for preparation of 2-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-N- methoxy-N-methyl-acetamide To a solution of 2-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]acetic acid (7.75 g, 23.68 mmol, 1.0 equiv.) in DCM (80 mL) was added HATU (10.81 g, 28.42 mmol, 1.2 equiv.), DIEA (15.31 g, 118.42 mmol, 20.63 mL, 5 equiv.) and N-methoxymethanamine (6.93 g, 71.05 mmol, 3.0 equiv., HCl). The mixture was stirred at 20 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~5% ethyl acetate/petroleum ether) to give 2-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]-N-methoxy-N-methyl-acetamide (7.0 g, 18.38 mmol, 77.61% yield) as a yellow oil. MS (ESI) m/z: 369.9 [M+H] + Step E. Procedure for preparation of 2-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-N-methoxy-N-methylacetamide Stereoisomers of 2-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]-N-methoxy-N-methy l- acetamide (7.0 g, 18.90 mmol, 1.0 equiv.) was separated by prep-HPLC to give 2-((1r,4r)-4- (3-bromo-2-methylphenoxy)cyclohexyl)-N-methoxy-N-methylaceta mide (1.6 g, 4.23 mmol, 22.38% yield) as a yellow solid. MS (ESI) m/z: 370.2 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.16 - 7.12 (m, 1H), 7.07 (t, J = 8.0 Hz, 1H), 7.03 - 6.99 (m, 1H), 4.28 - 4.16 (m, 1H), 3.65 (s, 3H), 3.08 (s, 3H), 2.29 (d, J = 6.4 Hz, 2H), 2.21 (s, 3H), 2.08 - 2.00 (m, 2H), 1.82 - 1.69 (m, 3H), 1.43 - 1.31 (m, 2H), 1.20 - 1.05 (m, 2H) Step F. Procedure for preparation of 1-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propan-2-one To a solution of 2-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-N-methoxy- N- methylacetamide (1.6 g, 4.32 mmol, 1.0 equiv.) in THF (16 mL) was added MeMgBr (3 M, 2.88 mL, 2 equiv.) dropwise at -70 °C under N2 atmosphere. The mixture was stirred at 20 °C for 2 hours under N 2 atmosphere. The reaction mixture was added dropwise into saturated NH 4 Cl solution 20 mL at 0 °C and extracted with EtOAc (30 mL × 3). The combined organic layers were washed with brine (50 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give 1-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propan-2- one (1.4 g, crude) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.17 - 7.11 (m, 1H), 7.06 (t, J = 8.0 Hz, 1H), 7.03 - 6.98 (m, 1H), 4.27 - 4.16 (m, 1H), 2.34 (d, J = 6.4 Hz, 2H), 2.21 (s, 3H), 2.07 (s, 3H), 2.04 - 1.98 (m, 2H), 1.82 - 1.66 (m, 3H), 1.44 - 1.31 (m, 2H), 1.12 - 1.00 (m, 2H) Step G. Procedure for preparation of ethyl (E)-4-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-3-methylbut-2-enoate A solution of ethyl 2-diethoxyphosphorylacetate (1.16 g, 5.17 mmol, 1.02 mL, 1.2 equiv.) in THF (15 mL) was degassed and purged with N2 three times. The mixture was treated with LiHMDS (1 M, 6.46 mL, 1.5 equiv.) dropwise at -70 °C and stirred at -70 °C for 1 hour. Then, 1-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propan-2-on e (1.4 g, 4.30 mmol, 1.0 equiv.) was added at -70 °C slowly. The mixture was warmed to 20 °C and stirred at 20 °C for 15 hours under N 2 atmosphere. The reaction mixture was added dropwise into saturated NH 4 Cl solution (20 mL) at 0 °C, and then extracted with EtOAc (30 mL × 3). The combined organic layers were washed with brine (50 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~5% ethyl acetate/petroleum ether) to give ethyl (E)-4-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-3-methylbut-2-enoate (450 mg, 1.11 mmol, 25.86% yield) as a light yellow oil. MS (ESI) m/z: 395.1 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.16 - 7.11 (m, 1H), 7.10 - 6.99 (m, 2H), 5.76 - 5.59 (m, 1H), 4.30 - 4.17 (m, 1H), 4.12 - 4.01 (m, 2H), 2.20 (s, 3H), 2.11 - 2.01 (m, 6H), 1.86 (s, 1H), 1.68 (d, J = 12.8 Hz, 2H), 1.63 - 1.53 (m, 1H), 1.42 - 1.30 (m, 2H), 1.19 (t, J = 7.2 Hz, 3H), 1.15 - 0.99 (m, 2H) Step H. Procedure for preparation of ethyl 4-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-3-methylbutanoate To a solution of ethyl (E)-4-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-3-meth ylbut- 2-enoate (630 mg, 1.59 mmol, 1.0 equiv.) in EtOH (15 mL) was added PtO2 (72.37 mg, 318.72 μmol, 0.2 equiv.) under N 2 . The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H 2 (15 psi) at 20 °C for 2 hours. The mixture was filtered to give a filtrate, which was concentrated under reduced pressure to give a residue. The residue was purified by reversed-phase HPLC (with 0.1% NH3•H2O) to give ethyl 4-((1r,4r)- 4-(3-bromo-2-methylphenoxy)cyclohexyl)-3-methylbutanoate (440 mg, 1.01 mmol, 63.18% yield) as a yellow oil. MS (ESI) m/z: 397.3 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.16 - 7.11 (m, 1H), 7.10 - 7.00 (m, 2H), 4.34 (t, J = 4.8 Hz, 1H), 4.28 - 4.18 (m, 1H), 4.09 - 4.01 (m, 2H), 2.20 (s, 3H), 2.13 - 2.00 (m, 4H), 1.98 - 1.88 (m, 1H), 1.78 - 1.67 (m, 2H), 1.40 - 1.30 (m, 3H), 1.22 - 1.08 (m, 5H), 1.02 - 0.95 (m, 1H), 0.87 (d, J = 6.8 Hz, 3H) Step I. Procedure for preparation of 4-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)- 3-methylbutan-1-ol A solution of ethyl 4-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-3-methylbu tanoate (440 mg, 1.11 mmol, 1.0 equiv.) in THF (5 mL) was degassed and purged with N2 three times. LiAlH 4 (42.03 mg, 1.11 mmol, 1.0 equiv.) was then added. The mixture was stirred at 0 °C for 1 hour under N 2 atmosphere. To this mixture was added Na 2 SO 4 •10 H 2 O (2 g) at 0 °C. The resulting mixture was filtered to give a filtrate. The filtrate was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, petroleum ether: ethyl acetate = 8:1) to give 4-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-3-methylbu tan-1- ol (300 mg, 806.85 μmol, 72.86% yield) as a light yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.16 - 7.11 (m, 1H), 7.10 - 7.03 (m, 1H), 7.03 - 6.99 (m, 1H), 4.32 - 4.17 (m, 2H), 3.48 - 3.34 (m, 2H), 2.21 (s, 3H), 2.04 (d, J = 10.8 Hz, 2H), 1.81 - 1.67 (m, 2H), 1.66 - 1.55 (m, 1H), 1.48 - 1.30 (m, 4H), 1.24 - 1.10 (m, 2H), 1.07 - 0.92 (m, 3H), 0.83 (d, J = 6.8 Hz, 3H) Step J. Procedure for preparation of (S)-4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-3-methylbutan-1-ol Stereoisomers of 4-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-3-methylbu tan-1-ol (300 mg, 844.34 μmol, 1 equiv.) were separated by prep-HPLC to give (S)-4-((1r,4s)-4-(3- bromo-2-methylphenoxy)cyclohexyl)-3-methylbutan-1-ol (150 mg, 408.03 μmol, 48.33% yield) as a light yellow oil. Step K. Procedure for preparation of (S)-4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-3-methylbutanal A solution of (COCl)2 (48.58 mg, 382.77 μmol, 33.51 μL, 2.0 equiv.) in DCM (2 mL) was degassed and purged with N 2 three times. The mixture was treated with DMSO (59.81 mg, 765.53 μmol, 59.81 μL, 4.0 equiv.) dropwise at -70 °C, and then the mixture was stirred at -70 °C for 3 hours. (S)-4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-3-meth ylbutan-1-ol (68 mg, 191.38 μmol, 1.0 equiv.) in DCM (0.5 mL) was added dropwise at -70 °C. The mixture was stirred at -70 °C for 1 hour, and then TEA (116.20 mg, 1.15 mmol, 159.83 μL, 6.0 equiv.) was added dropwise at -70 °C. The resulting mixture was stirred at 20 °C for 0.5 hour. The reaction mixture was diluted with DCM (10 mL) and extracted with H 2 O (10 mL × 3). The combined organic layers were washed with saturated brine (20 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give (S)-4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-3-methylbutanal (55 mg, crude) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 9.66 (s, 1H), 7.16 - 7.11 (m, 1H), 7.11 - 6.99 (m, 2H), 4.29 - 4.17 (m, 1H), 2.42 - 2.36 (m, 1H), 2.21 (s, 4H), 2.12 - 2.00 (m, 3H), 1.82 - 1.69 (m, 2H), 1.42 - 1.29 (m, 3H), 1.15 - 1.08 (m, 2H), 1.05 - 0.94 (m, 2H), 0.88 (d, J = 6.8 Hz, 3H) Step L. Procedure for preparation of 3-(6-(4-((S)-4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-3-methylbutyl)piperazin-1-yl)-1-me thyl-1H-indazol-3- yl)piperidine-2,6-dione A solution of (S)-4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-3-meth ylbutanal (110 mg, 311.36 μmol, 1.0 equiv.) and 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6- dione (124.61 mg, 342.49 μmol, 1.1 equiv., HCl) in DCM (2 mL) was stirred at 20 °C for 16 hours. The mixture was then treated with NaBH(OAc) 3 (197.97 mg, 934.07 μmol, 3.0 equiv.). Afterwards, the mixture was stirred at 20 °C for 0.5 hour. The reaction mixture was diluted with H 2 O (10 mL) and extracted with DCM (10 mL × 3). The combined organic layers were washed with brine (20 mL × 1), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~4% methanol / dichloromethane) to give 3-(6-(4-((S)-4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-3-methylbutyl)piperazin-1-yl)-1-me thyl-1H-indazol-3- yl)piperidine-2,6-dione (128 mg, 180.06 μmol, 57.83% yield) as a yellow oil. MS (ESI) m/z: 666.1 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.85 (s, 1H), 7.52 (d, J = 8.0 Hz, 1H), 7.17 - 7.11 (m, 1H), 7.10 - 6.99 (m, 2H), 6.97 - 6.81 (m, 2H), 4.31 - 4.17 (m, 2H), 3.90 (s, 3H), 3.40 - 3.11 (m, 6H), 2.71 - 2.53 (m, 4H), 2.39 - 2.24 (m, 2H), 2.24 - 2.11 (m, 4H), 2.05 (d, J = 10.0 Hz, 2H), 1.81 - 1.71 (m, 2H), 1.68 - 1.50 (m, 2H), 1.38 - 1.29 (m, 4H), 1.27 - 1.14 (m, 2H), 1.12 - 0.93 (m, 3H), 0.88 (d, J = 5.6 Hz, 3H) Step M. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-((2S)-4-(4 -(3-(2,6-dioxopiperidin-3-yl)- 1-methyl-1H-indazol-6-yl)piperazin-1-yl)-2-methylbutyl)cyclo hexyl)oxy)-2- methylphenyl)picolinate A mixture of 3-(6-(4-((S)-4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexy l)-3- methylbutyl)piperazin-1-yl)-1-methyl-1H-indazol-3-yl)piperid ine-2,6-dione (128 mg, 192.58 μmol, 1.0 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (129.76 mg, 211.83 μmol, 1.1 equiv.), KF (33.56 mg, 577.73 μmol, 13.53 μL, 3.0 equiv.), and Ad 2 nBuP Pd G 3 (14.02 mg, 19.26 μmol, 0.1 equiv.) in dioxane (3 mL) and H 2 O (0.3 mL) was degassed and purged with N 2 three times. The mixture was stirred at 100 °C for 3 hours under N2 atmosphere. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, petroleum ether : ethyl acetate = 1:1) to give tert- butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)- 4-((2S)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazo l-6-yl)piperazin-1-yl)-2- methylbutyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (90 mg, crude) as a yellow oil. MS (ESI) m/z: 1071.1 [M+H] + Step N. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-((2S)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-2-methylbutyl)cyclohe xyl)oxy)-2- methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-((2S*)-4-(4-(3 -(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-2-methylbutyl)cyclohe xyl)oxy)-2- methylphenyl)picolinic acid] To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-((2S)-4-(4-(3-(2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-6- yl)piperazin-1-yl)-2-methylbutyl)cyclohexyl)oxy)-2-methylphe nyl)picolinate (90 mg, 84.08 μmol, 1 equiv.) in DCM (2 mL) was added TFA (1 mL). The mixture was stirred at 40 °C for 10 min. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-((2S)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-2-methylbutyl)cyclohe xyl)oxy)-2- methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin - 2(1H)-yl)-3-(3-(((1S,4r)-4-((2S*)-4-(4-(3-(2,6-dioxopiperidi n-3-yl)-1-methyl-1H-indazol-6- yl)piperazin-1-yl)-2-methylbutyl)cyclohexyl)oxy)-2-methylphe nyl)picolinic acid] (35.31 mg, 33.77 μmol, 40.16% yield) as a yellow solid. MS (ESI) m/z: 1015.3 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 13.23 - 12.06 (m, 2H), 10.85 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.53 - 7.41 (m, 4H), 7.40 - 7.30 (m, 2H), 7.11 - 7.02 (m, 1H), 6.99 - 6.87 (m, 3H), 6.84 (s, 1H), 6.61 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.32 - 4.13 (m, 2H), 3.98 - 3.82 (m, 5H), 3.23 (s, 4H), 3.02 (s, 2H), 2.60 (s, 6H), 2.35 - 2.26 (m, 2H), 2.22 - 2.00 (m, 4H), 1.87 (s, 3H), 1.82 - 1.71 (m, 2H), 1.59 - 1.46 (m, 2H), 1.42 - 1.13 (m, 6H), 1.08 - 1.01 (m, 2H), 0.87 (d, J = 5.6 Hz, 3H) Example 251. Preparation of Compound 307b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4- ((2R)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 6-yl)piperazin-1-yl)-2- methylbutyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1R,4r)-4- ((2R*)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)piperazin-1-yl)-2- methylbutyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of (R)-4-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-3-methylbutanal A solution of (COCl) 2 (85.73 mg, 675.47 μmol, 59.13 μL, 2 equiv.) in DCM (2 mL) was cooled to -78 °C, and DMSO (105.55 mg, 1.35 mmol, 105.55 μL, 4 equiv.) was added dropwise. The mixture was stirred at -78~25 °C for 2 hours under N2 atmosphere. Then (R)-4-((1r,4R)-4-(3- bromo-2-methylphenoxy)cyclohexyl)-3-methylbutan-1-ol (120 mg, 337.73 μmol, 1 equiv.) was added dropwise, and the mixture was stirred at -78~25 °C for 2 hours under N2 atmosphere. TEA (205.05 mg, 2.03 mmol, 282.05 μL, 6 equiv.) was added dropwise to the mixture at -78 °C, and the mixture was warmed to 25 °C and stirred at 25 °C for 1 hour under N2 atmosphere. The reaction mixture was diluted with H2O (50 mL) and extracted with DCM (20 mL × 3). The combined organic layers were washed with H 2 O (10 mL × 3), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give (R)-4-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-3-methylbutanal (110 mg, crude) was obtained as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 9.66 (s, 1H), 7.17 - 6.78 (m, 3H), 4.29 - 4.12 (m, 1H), 2.43 - 2.34 (m, 1H), 2.21 (s, 3H), 2.12 - 1.99 (m, 3H), 1.82 - 1.67 (m, 2H), 1.36 (d, J = 10.8 Hz, 2H), 1.24 - 0.97 (m, 6H), 0.88 (d, J = 6.8 Hz, 3H). Step B. Procedure for preparation of 3-(6-(4-((R)-4-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-3-methylbutyl)piperazin-1-yl)-1-me thyl-1H-indazol-3- yl)piperidine-2,6-dione A solution of (R)-4-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-3-meth ylbutanal (110 mg, 311.36 μmol, 1 equiv.) and 3-(1-methyl-6-(piperazin-1-yl)-1H-indazol-3-yl)piperidine- 2,6-dione (122.32 mg, 373.63 μmol, 1.2 equiv.) in DCM (2 mL) was stirred at 0 °C for 2 hours. To this mixture was added NaBH(OAc) 3 (197.97 mg, 934.07 μmol, 3 equiv.). The resulting reaction mixture was stirred at 0 °C for 12 hours. The reaction mixture was quenched by addition of H 2 O (5 mL) at 25 °C, and extracted with DCM (5 mL × 3). The combined organic layers were washed with brine (5 mL × 3), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give 3-(6-(4-((R)-4-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexy l)-3- methylbutyl)piperazin-1-yl)-1-methyl-1H-indazol-3-yl)piperid ine-2,6-dione (200 mg, crude) as a yellow oil. MS (ESI) m/z: 664.2 [M+H] + . Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-((2R)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-2-methylbutyl)cyclohe xyl)oxy)-2- methylphenyl)picolinate A mixture of 3-(6-(4-((r)-4-((1r,4R)-4-(3-bromo-2-methylphenoxy)cyclohexy l)-3- methylbutyl)piperazin-1-yl)-1-methyl-1H-indazol-3-yl)piperid ine-2,6-dione (180 mg, 270.81 μmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pic olinate (199.06 mg, 324.97 μmol, 1.2 equiv.), Ad2nBup-Pd-G3(cataCXium® A Pd G3 ) (19.72 mg, 27.08 μmol, 0.1 equiv.), and KF (47.20 mg, 812.43 μmol, 60.18 μL, 3 equiv.) in dioxane (5 mL) and H2O (0.5 mL) was degassed and purged with N 2 three times. The mixture was stirred at 100 °C for 2 hours under a N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH = 20:1) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1r,4r)-4-((2R)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl -1H-indazol-6-yl)piperazin-1-yl)- 2-methylbutyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (170 mg, 150.89 μmol, 55.72% yield) as a yellow oil. MS (ESI) m/z: 1070.6 [M+H] + . Step D. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-((2R)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-2-methylbutyl)cyclohe xyl)oxy)-2- methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-((2R*)-4-(4-(3 -(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-2-methylbutyl)cyclohe xyl)oxy)-2- methylphenyl)picolinic acid] A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1r,4r)-4-((2R)-4-(4-(3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-6- yl)piperazin-1-yl)-2-methylbutyl)cyclohexyl)oxy)-2-methylphe nyl)picolinate (170 mg, 158.83 μmol, 1 equiv.) in TFA (1 mL) and DCM (2 mL) was stirred at 25 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-((2R)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-2-methylbutyl)cyclohe xyl)oxy)-2- methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin - 2(1H)-yl)-3-(3-(((1R,4r)-4-((2R*)-4-(4-(3-(2,6-dioxopiperidi n-3-yl)-1-methyl-1H-indazol-6- yl)piperazin-1-yl)-2-methylbutyl)cyclohexyl)oxy)-2-methylphe nyl)picolinic acid] (48.43 mg, 47.06 μmol, 29.63% yield) as a white solid. MS (ESI) m/z: 1014.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.99 - 12.73 (m, 1H), 10.85 (s, 1H), 8.13 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 6.8 Hz, 1H), 7.53 - 7.43 (m, 4H), 7.40 - 7.32 (m, 2H), 7.11 - 7.03 (m, 1H), 6.98 - 6.86 (m, 4H), 6.61 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.28 - 4.16 (m, 2H), 3.93 - 3.87 (m, 5H), 3.28 - 3.24 (m, 2H), 3.02 (s, 2H), 2.82 - 2.57 (m, 6H), 2.39 - 2.23 (m, 2H), 2.21 - 1.98 (m, 4H), 1.87 (s, 3H), 1.81 - 1.71 (m, 2H), 1.62 - 1.50 (m, 2H), 1.48 - 1.25 (m, 5H), 1.25 - 0.94 (m, 5H), 0.88 (d, J = 5.6 Hz, 3H). Example 252. Preparation of Compound 308a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4- ((3R)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 6-yl)piperazin-1-yl)-3- methylbutyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1S,4r)-4- ((3R*)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)piperazin-1-yl)-3- methylbutyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of 2-(4-(3-bromo-2-methylphenoxy)cyclohexyl)ethan- 1-ol To a solution of methyl 2-(4-(3-bromo-2-methylphenoxy)cyclohexyl)acetate (14.5 g, 42.49 mmol, 1 equiv.) in THF (150 mL) at 0 °C was added LiAlH4 (1.94 g, 50.99 mmol, 1.2 equiv.). The mixture was stirred at 25 °C for 16 hours under a N 2 atmosphere. The reaction mixture was quenched by the addition of saturated sodium carbonate solution (6.0 mL) at 0 °C, and then diluted with EtOAc (80 mL). The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 - 50% ethyl acetate/petroleum ether) and reverse-phase HPLC (with 0.1% formic acid) to give 2-(4-(3-bromo-2-methylphenoxy)cyclohexyl)ethan-1-ol (7.5 g, 23.58 mmol, 55.49% yield) as a purple oil. MS (ESI) m/z: 314.2 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.15 - 7.10 (m, 1H), 7.06 (t, J = 8.0 Hz, 1H), 7.00 - 6.96 (m, 1H), 4.63 (s, 1H), 4.33 - 4.29 (m, 1H), 3.46 - 3.42 (m, 2H), 2.28 - 2.27 (m, 1H), 2.26 (s, 3H), 2.20 (s, 1H), 1.91 - 1.83 (m, 2H), 1.53 (s, 1H), 1.49 (s, 2H), 1.37 (d, J = 6.4 Hz, 2H), 1.22 (s, 2H) Step B. Procedure for preparation of 2-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)ethan-1-ol The stereoisomers of 2-(4-(3-bromo-2-methylphenoxy)cyclohexyl)ethan-1-ol were separated by SFC to give 2-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)ethan-1-ol (700 mg, 2.23 mmol, 23.33% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.14 - 7.10 (m, 1H), 7.06 (t, J = 8.0 Hz, 1H), 6.99 - 6.95 (m, 1H), 4.63 (s, 1H), 4.30 (t, J = 5.2 Hz, 1H), 3.47 - 3.42 (m, 2H), 2.27 - 2.25 (m, 3H), 1.91 - 1.84 (m, 2H), 1.51 (dd, J = 10.0, 6.8 Hz, 5H), 1.40 - 1.35 (m, 2H), 1.32 - 1.23 (m, 2H) Step C. Procedure for preparation of 2-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)acetaldehyde To a solution of (COCl) 2 (405.22 mg, 3.19 mmol, 279.46 μL, 2 equiv.) in DCM (3 mL) was added DMSO (498.89 mg, 6.39 mmol, 498.89 μL, 4 equiv.) dropwise at -70 °C over 2 minutes. After addition, the mixture was stirred at this temperature for 30 minutes, and then 2-((1r,4r)- 4-(3-bromo-2-methylphenoxy)cyclohexyl)ethan-1-ol (500.00 mg, 1.60 mmol, 1 equiv.) in DCM (3 mL) was added dropwise at -70 °C. After addition, the mixture was stirred at this temperature for 30 minutes, and then TEA (969.15 mg, 9.58 mmol, 1.33 mL, 6 equiv.) was added into the mixture. The resulting mixture was stirred at 25°C for 1 hour. The reaction mixture was diluted with water (20 mL) and extracted with DCM (25 mL × 3). The combined organic layers were washed with brine (15 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 - 7% ethyl acetate/petroleum ether) to give 2-((1r,4r)-4- (3-bromo-2-methylphenoxy)cyclohexyl)acetaldehyde (370 mg, 1.19 mmol, 74.48% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 9.69 - 9.64 (m, 1H), 7.16 - 7.12 (m, 1H), 7.10 - 7.04 (m, 1H), 7.03 - 7.00 (m, 1H), 4.28 - 4.18 (m, 1H), 2.34 (dd, J = 6.8, 2.0 Hz, 2H), 2.21 (s, 3H), 2.04 (d, J = 9.6 Hz, 2H), 1.92 - 1.82 (m, 1H), 1.75 (d, J = 12.0 Hz, 2H), 1.45 - 1.36 (m, 2H), 1.21 - 1.14 (m, 2H) Step D. Procedure for preparation of ethyl (E)-4-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2-methylbut-2-enoate To a mixture of ethyl 2-(diethoxyphosphoryl)propanoate (424.82 mg, 1.78 mmol, 388.32 μL, 1.5 equiv.) was added NaH (104.62 mg, 2.62 mmol, 60% purity, 2.2 equiv.) in THF (5 mL) at -10 °C under N2 atmosphere for 3 hours. Then 2-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)acetaldehyde (370 mg, 1.19 mmol, 1 equiv.) was added at 0 °C under N2 atmosphere. The mixture was stirred at 25 °C for 16 hours under N2 atmosphere. The reaction mixture was quenched by addition water (30 mL) at 0 °C and extracted with EtOAc (20 mL × 3). The combined organic layers were washed with brine (20 mL × 3), dried over anhydrous sodium sulfate filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 - 7% ethyl acetate/petroleum ether) to give ethyl (E)-4-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2-meth ylbut-2- enoate (200 mg, 505.91 μmol, 42.55% yield) as a colorless oil. MS (ESI) m/z: 396.9 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 7.16 - 7.11 (m, 1H), 7.09 - 7.04 (m, 1H), 7.03 - 7.00 (m, 1H), 6.71 (t, J = 7.6 Hz, 1H), 4.28 - 4.19 (m, 1H), 4.12 (q, J = 7.2 Hz, 2H), 2.20 (s, 3H), 2.11 (t, J = 7.2 Hz, 2H), 2.04 (d, J = 10.0 Hz, 2H), 1.78 - 1.76 (m, 3H), 1.72 (s, 2H), 1.51 - 1.44 (m, 1H), 1.42 - 1.32 (m, 2H), 1.24 - 1.19 (m, 3H), 1.18 - 1.07 (m, 2H) Step E. Procedure for preparation of ethyl 4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2-methylbutanoate A mixture of ethyl (E)-4-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2-meth ylbut-2- enoate (200 mg, 505.91 μmol, 1 equiv.) and PtO 2 (57.44 mg, 50.59 μmol, 20% purity, 0.1 equiv.) in EtOH (10 mL) was degassed and purged with N2 three times. The mixture was stirred at 25 °C for 16 hours under H2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give ethyl 4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2- methylbutanoate (170 mg, 408.72 μmol, 80.79% yield) as a colorless oil. MS (ESI) m/z: 397.2 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 7.15 - 7.10 (m, 1H), 7.06 (t, J = 8.0 Hz, 1H), 7.03 - 6.98 (m, 1H), 4.38 (s, 1H), 4.26 - 4.17 (m, 1H), 4.09 - 4.02 (m, 2H), 3.44 (q, J = 7.2 Hz, 2H), 2.41 - 2.31 (m, 1H), 2.22 - 2.17 (m, 3H), 2.03 (d, J = 9.6 Hz, 2H), 1.78 - 1.70 (m, 2H), 1.59 - 1.50 (m, 1H), 1.44 - 1.37 (m, 1H), 1.36 - 1.28 (m, 2H), 1.16 (s, 4H), 1.05 (d, J = 2.0 Hz, 4H) Step F. Procedure for preparation of 4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)- 2-methylbutan-1-ol A solution of ethyl 4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2-methylbu tanoate (170 mg, 427.84 μmol, 1 equiv.) in THF (5 mL) at 0 °C was stirred at 0 °C for 15 minutes. After 15 minutes, LiAlH4 (32.47 mg, 855.68 μmol, 2 equiv.) was added, and the mixture was stirred at 25 °C for 16 hours under N 2 atmosphere. The reaction mixture was quenched by the addition of saturated sodium carbonate solution (0.15 mL) at 0 °C and then diluted with EtOAc 20 mL. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , petroleum ether : ethyl acetate = 3:1) and prep-HPLC to give 4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2- methylbutan-1-ol (100 mg, 280.58 μmol, 65.58% yield, 99.693% purity) as a yellow oil. MS (ESI) m/z: 356.4 [M+H] + 1 H NMR (400 MHz, CDCl 3 ) δ = 7.14 (d, J = 8.0 Hz, 1H), 6.97 (t, J = 8.0 Hz, 1H), 6.81 (d, J = 8.0 Hz, 1H), 4.14 - 4.04 (m, 1H), 3.55 - 3.49 (m, 1H), 3.48 - 3.42 (m, 1H), 2.32 - 2.29 (m, 3H), 2.17 - 2.10 (m, 2H), 1.86 (td, J = 10.0, 2.8 Hz, 2H), 1.65 - 1.55 (m, 1H), 1.49 - 1.43 (m, 4H), 1.31 - 1.21 (m, 3H), 1.18 - 1.11 (m, 1H), 1.07 - 0.98 (m, 2H), 0.93 (d, J = 6.4 Hz, 3H) Step G. Procedure for preparation of (R)-4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2-methylbutan-1-ol The stereoisomers of 4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2-methylbu tan-1- ol were separated by SFC to give (R)-4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2- methylbutan-1-ol (40 mg, 112.58 μmol, 40.00% yield) was obtained as a colorless oil. Step H. Procedure for preparation of (R)-4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2-methylbutanal To a solution of (R)-4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2-meth ylbutan-1-ol (40 mg, 112.58 μmol, 1 equiv.) in DCM (1 mL) was added Dess-Martin periodinane (95.50 mg, 225.16 μmol, 69.76 μL, 2 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was diluted with water (10 mL) and extracted with DCM (10 mL × 3). The combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure to give (R)-4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2-meth ylbutanal (45 mg, crude) as a yellow oil. Step I. Procedure for preparation of 3-(6-(4-((R)-4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2-methylbutyl)piperazin-1-yl)-1-me thyl-1H-indazol-3- yl)piperidine-2,6-dione To a solution of (R)-4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2-meth ylbutanal (45 mg, 127.37 μmol, 1 equiv.) in DCM (2 mL) was added 3-(1-methyl-6-piperazin-1-yl- indazol-3-yl)piperidine-2,6-dione (41.70 mg, 127.37 μmol, 1 equiv.) at 25 °C. The reaction mixture was stirred for 15 hours. To this mixture, NaBH(OAc)3 (80.99 mg, 382.12 μmol, 3 equiv.) was added at 0 °C. The resulting mixture was stirred at 25 °C for 1 hour. The residue was concentrated under reduced pressure to give a residue. The residue was purified by prep- TLC (SiO 2 , DCM: MeOH = 10:1) to give 3-(6-(4-((R)-4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2-methylbutyl)piperazin-1-yl)-1-me thyl-1H-indazol-3- yl)piperidine-2,6-dione (60 mg, crude) as a white solid. MS (ESI) m/z: 664.5 [M+H] + . Step J. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-((3R)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-3-methylbutyl)cyclohe xyl)oxy)-2- methylphenyl)picolinate A mixture of 3-(6-(4-((R)-4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexy l)-2- methylbutyl)piperazin-1-yl)-1-methyl-1H-indazol-3-yl)piperid ine-2,6-dione (41 mg, 61.68 μmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (37.78 mg, 61.68 μmol, 1 equiv.), KF (10.75 mg, 185.05 μmol, 4.34 μL, 3 equiv.), and [2-(2- aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-butyl-phos phane;methanesulfonate (4.49 mg, 6.17 μmol, 0.1 equiv.) in dioxane (5 mL) and H2O (0.5 mL) was degassed and purged with N 2 three times. The mixture was stirred at 100 °C for 2 hours under N 2 atmosphere. The reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (10 mL × 3). The combined organic layers were dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH = 10:1) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- (3-(((1s,4r)-4-((3R)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-met hyl-1H-indazol-6-yl)piperazin-1- yl)-3-methylbutyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (60 mg, 56.06 μmol, 90.88% yield) as a white solid. MS (ESI) m/z: 536.3 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.85 (d, J = 2.8 Hz, 1H), 10.83 (s, 1H), 8.01 (d, J = 7.6 Hz, 1H), 7.77 (d, J = 7.2 Hz, 1H), 7.60 (d, J = 6.4 Hz, 1H), 7.52 - 7.41 (m, 4H), 7.40 - 7.30 (m, 2H), 7.11 - 7.03 (m, 1H), 6.98 - 6.88 (m, 3H), 6.83 (s, 1H), 6.55 (d, J = 6.8 Hz, 1H), 4.97 (s, 2H), 4.29 - 4.16 (m, 2H), 3.88 (s, 5H), 3.21 (s, 4H), 3.06 - 3.00 (m, 2H), 2.69 - 2.58 (m, 4H), 2.33 (s, 3H), 2.22 - 2.14 (m, 3H), 2.09 - 2.01 (m, 3H), 1.84 (s, 3H), 1.81 - 1.72 (m, 2H), 1.70 - 1.59 (m, 2H), 1.27 - 1.17 (m, 4H), 1.07 (s, 3H), 0.99 (s, 9H), 0.88 (d, J = 6.0 Hz, 3H) Step K. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-((3R)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-3-methylbutyl)cyclohe xyl)oxy)-2- methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-((3R*)-4-(4-(3 -(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-3-methylbutyl)cyclohe xyl)oxy)-2- methylphenyl)picolinic acid] A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1s,4r)-4-((3R)-4-(4-(3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-6- yl)piperazin-1-yl)-3-methylbutyl)cyclohexyl)oxy)-2-methylphe nyl)picolinate (55 mg, 51.39 μmol, 1 equiv.) in DCM (3 mL) and TFA (1 mL) was stirred at 25 °C for 12 hours. The residue was concentrated under reduced pressure to give a residue. The residue was purified by prep- HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-((3R)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl -1H-indazol-6-yl)piperazin-1-yl)- 3-methylbutyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r) -4-((3R*)-4-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin-1-yl )-3- methylbutyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] (20.39 mg, 19.74 μmol, 80.10% yield) as a yellow solid. MS (ESI) m/z: 1014.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.85 (s, 1H), 8.17 (s, 1H), 8.03 (d, J = 7.2 Hz, 1H), 7.81 - 7.75 (m, 1H), 7.62 (d, J = 6.4 Hz, 1H), 7.51 - 7.41 (m, 4H), 7.40 - 7.32 (m, 2H), 7.10 - 7.02 (m, 1H), 6.96 - 6.87 (m, 3H), 6.84 (s, 1H), 6.61 (d, J = 6.8 Hz, 1H), 5.02 - 4.92 (m, 2H), 4.28 - 4.16 (m, 2H), 3.93 - 3.87 (m, 5H), 3.21 (s, 4H), 3.02 (s, 2H), 2.63 - 2.58 (m, 3H), 2.22 - 2.01 (m, 7H), 1.87 (s, 3H), 1.79 (d, J = 2.8 Hz, 2H), 1.66 (dt, J = 2.4, 4.4 Hz, 1H), 1.50 - 1.30 (m, 4H), 1.27 - 1.17 (m, 3H), 1.14 - 0.95 (m, 4H), 0.88 (d, J = 6.0 Hz, 3H). Example 253. Preparation of Compound 309 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[5-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-2-pyridyl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of 1-bromo-2-methyl-3-prop-2-ynoxy-benzene A mixture of 3-bromo-2-methyl-phenol (4 g, 21.39 mmol, 1 equiv.), 3-bromoprop-1-yne (3.82 g, 25.66 mmol, 2.77 mL, 1.2 equiv.), K 2 CO 3 (8.87 g, 64.16 mmol, 3 equiv.), and KI (355.02 mg, 2.14 mmol, 0.1 equiv.) in THF (30 mL) was degassed and purged with N2 three times. The mixture was stirred at 60 °C for 16 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 - 3% ethyl acetate/petroleum ether) to give 1-bromo-2-methyl-3-prop- 2-ynoxy-benzene (4.12 g, 18.30 mmol, 85.59% yield) as a colorless oil. 1 H NMR (400 MHz, CDCl 3 ) δ = 7.24 - 7.19 (m, 1H), 7.03 (t, J = 8.0 Hz, 1H), 6.92 (d, J = 8.4 Hz, 1H), 4.72 (d, J = 2.4 Hz, 2H), 2.53 (s, 1H), 2.35 (s, 3H) Step B. Procedure for preparation of methyl 2-[6-[3-(3-bromo-2-methyl-phenoxy)prop-1- ynyl]-3-pyridyl]acetate A mixture of 1-bromo-2-methyl-3-prop-2-ynoxy-benzene (1.82 g, 8.08 mmol, 1.5 equiv.), methyl 2-(6-chloro-3-pyridyl)acetate (1 g, 5.39 mmol, 1 equiv.), K2CO3 (2.23 g, 16.16 mmol, 3 equiv.), acetonitrile;dichloropalladium (27.95 mg, 107.75 μmol, 0.02 equiv.), and XPhos (154.11 mg, 323.26 μmol, 0.06 equiv.) in MeCN (10 mL) was degassed and purged with N2 three times. The mixture was stirred at 60 °C for 1 hour under N2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (25 mL × 3). The combined organic layers were washed with brine (15 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 - 25% ethyl acetate/petroleum ether) to give methyl 2- [6-[3-(3-bromo-2-methyl-phenoxy)prop-1-ynyl]-3-pyridyl]aceta te (180 mg, 480.99 μmol, 8.93% yield) as a brown oil. MS (ESI) m/z: 375.7 [M+H] + 1 H NMR (400 MHz, CDCl3) δ = 7.66 - 7.58 (m, 1H), 7.44 - 7.36 (m, 1H), 7.23 - 7.17 (m, 1H), 7.07 - 6.98 (m, 3H), 4.96 - 4.91 (m, 2H), 3.72 (s, 3H), 3.65 (s, 2H), 2.36 (s, 3H) Step C. Procedure for preparation of methyl 2-[6-[3-(3-bromo-2-methyl- phenoxy)propyl]-3-pyridyl]acetate A mixture of methyl 2-[6-[3-(3-bromo-2-methyl-phenoxy)prop-1-ynyl]-3-pyridyl]ace tate (160 mg, 427.55 μmol, 1 equiv.) and Rh/Al2O3 (88.00 mg, 42.75 μmol, 5% purity, 0.1 equiv.) in EtOAc (10 mL) was degassed and purged with H2 three times. The reaction mixture was stirred at 25 °C for 3 hours under H 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 - 5% DCM/MeOH) to give methyl 2-[6-[3-(3-bromo-2-methyl- phenoxy)propyl]-3-pyridyl]acetate (65 mg, 171.84 μmol, 40.19% yield) as a brown oil. MS (ESI) m/z: 379.9 [M+H] + 1 H NMR (400 MHz, CDCl3) δ = 8.46 (s, 1H), 7.65 - 7.58 (m, 1H), 7.19 (d, J = 7.2 Hz, 1H), 7.15 (d, J = 8.0 Hz, 1H), 7.02 - 6.96 (m, 1H), 6.75 (d, J = 8.4 Hz, 1H), 4.01 (t, J = 6.0 Hz, 2H), 3.63 (s, 3H), 3.03 (t, J = 7.2 Hz, 2H), 2.33 - 2.30 (m, 3H), 2.28 - 2.21 (m, 2H), 1.31 - 1.25 (m, 2H) Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[5-(2-methoxy-2-oxo -ethyl)-2- pyridyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-2-carb oxylate (116.60 mg, 190.35 μmol, 1.2 equiv.), methyl 2-[6-[3-(3-bromo-2-methyl-phenoxy)propyl]-3-pyridyl]acetate (60 mg, 158.62 μmol, 1 equiv.), Ad2nBuP Pd G3 (11.55 mg, 15.86 μmol, 0.1 equiv.), and KF (1.5 M, 317.24 μL, 3 equiv.) were taken up into a microwave tube in dioxane (1 mL), and the tube was sealed. The sealed tube was heated at 100 °C for 1 hour under microwave. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (25 mL × 3). The combined organic layers were washed with brine (15 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM : MeOH = 15:1) to give tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[5-(2- methoxy-2-oxo-ethyl)-2- pyridyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate (50 mg, 63.78 μmol, 40.21% yield) as a brown oil. MS (ESI) m/z: 784.7 [M+H] + Step E. Procedure for preparation of 2-[6-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyr idyl]-2-methyl- phenoxy]propyl]-3-pyridyl]acetic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[5-(2-methoxy-2-oxo-ethyl)-2-pyridyl]propoxy]-2- methyl-phenyl]pyridine-2- carboxylate (50 mg, 63.78 μmol, 1 equiv.) and LiOH·H 2 O (8.03 mg, 191.34 μmol, 3 equiv.) in THF (2 mL) and H2O (0.5 mL) was degassed and purged with N2 three times. The mixture was stirred at 25 °C for 2 hours under N2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with DCM (25 mL × 3). The combined organic layers were washed with brine (15 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give 2-[6-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]propyl]-3- pyridyl]acetic acid (50 mg, crude) as a brown solid. MS (ESI) m/z: 770.7 [M+H] + Step F. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[5-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-2-pyridyl]propoxy]-2-methyl-phenyl] pyridine-2-carboxylate A mixture of 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (25.16 mg, 97.41 μmol, 1.5 equiv.), 2-[6-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H-isoquinolin-2- yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]propyl ]-3-pyridyl]acetic acid (50 mg, 64.94 μmol, 1 equiv.), and EDCI (18.67 mg, 97.41 μmol, 1.5 equiv.) in pyridine (2 mL) was degassed and purged with N 2 three times. The mixture was stirred at 25 °C for 3 hours under N2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (25 mL × 3). The combined organic layers were washed with brine (15 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[5-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-2-pyridyl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylate (70 mg, crude) as a green-black oil. MS (ESI) m/z: 1010.4 [M+H] + Step G. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[5-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-2-pyridyl]propoxy]-2-methyl-phenyl]py ridine-2-carboxylic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[5-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indaz ol-6-yl]amino]-2-oxo-ethyl]-2- pyridyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate (70 mg, 69.30 μmol, 1 equiv.) in DCM (2 mL) and TFA (2 mL) was degassed and purged with N2 three times. The mixture was stirred at 40 °C for 24 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[5- [2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]- 2-oxo-ethyl]-2- pyridyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid (16.1 mg, 15.81 μmol, 22.82% yield) as a white solid. MS (ESI) m/z: 954.6 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.90 - 12.40 (m, 1H), 10.91 - 10.82 (m, 1H), 10.44 - 10.38 (m, 1H), 8.47 - 8.42 (m, 1H), 8.18 - 8.15 (m, 1H), 8.07 - 7.98 (m, 2H), 7.79 (d, J = 7.6 Hz, 1H), 7.69 - 7.58 (m, 3H), 7.50 - 7.41 (m, 3H), 7.40 - 7.32 (m, 2H), 7.26 (d, J = 7.6 Hz, 1H), 7.13 - 7.04 (m, 2H), 6.98 - 6.92 (m, 1H), 6.90 - 6.82 (m, 1H), 6.67 - 6.61 (m, 1H), 4.98 (s, 2H), 4.34 - 4.27 (m, 1H), 4.05 - 3.98 (m, 2H), 3.95 - 3.86 (m, 5H), 3.70 (s, 2H), 3.05 - 2.99 (m, 2H), 2.91 (t, J = 7.2 Hz, 2H), 2.71 - 2.61 (m, 2H), 2.16 (d, J = 8.0 Hz, 2H), 1.91 (s, 3H), 1.24 (s, 2H) Example 254. Preparation of Compound 311a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4s)-4-(3- (1-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip eridin-4- yl)propyl)cyclohexyl)oxy)-2-methylphenyl) picolinic acid Step A. Procedure for preparation of methyl 2-(4-hydroxycyclohexyl)acetate To a solution of 2-(4-hydroxycyclohexyl)acetic acid (10 g, 63.21 mmol, 1 equiv.) in MeOH (100 mL) was added H 2 SO 4 (12.40 g, 126.43 mmol, 6.74 mL, 2 equiv.) at 0 °C. The mixture was stirred at 60 °C for 3 hours. The reaction mixture was quenched with NaHCO3 (300 mL) and extracted with EtOAc (50 mL × 6). The combined organic layers were washed with saturated salt solution (100 mL × 1), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give methyl 2-(4-hydroxycyclohexyl)acetate (13.5 g, crude) as a light yellow oil. Step B. Procedure for preparation of methyl 2-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]acetate A mixture of methyl 2-(4-hydroxycyclohexyl)acetate (12 g, 69.68 mmol, 1 equiv.), 3-bromo- 2-methyl-phenol (15.64 g, 83.61 mmol, 1.2 equiv.), and 2-(tributyl-λ5- phosphanylidene)acetonitrile (25.23 g, 104.52 mmol, 1.5 equiv.) in toluene (120 mL) was degassed and purged with N2 three times. The mixture was stirred at 120 °C for 12 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~5% ethyl acetate/petroleum ether) to give methyl 2-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]acetate (20 g, crude) as a yellow oil. Step C. Procedure for preparation of 2-[4-(3-bromo-2-methyl- phenoxy)cyclohexyl]ethanol To a solution of methyl 2-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]acetate (20 g, 58.61 mmol, 1 equiv.) in THF (10 mL) was added LiAlH 4 (2.67 g, 70.33 mmol, 1.2 equiv.) at 0 °C. The mixture was stirred at 0 °C for 1 hour. The reaction mixture was quenched by sodium sulfate decahydrate (10 g) at 0°C, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~20% ethyl acetate/petroleum ether) to give 2-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]ethanol (7 g, crude) as a yellow oil. 1 H NMR (400 MHz, CDCl 3 ) δ = 7.16 - 7.10 (m, 1H), 6.97 (t, J = 8.0 Hz, 1H), 6.83 - 6.75 (m, 1H), 4.55 - 4.04 (m, 1H), 3.75 - 3.71 (m, 2H), 2.35 - 2.30 (m, 3H), 2.18 - 2.10 (m, 1H), 2.05 - 1.98 (m, 2H), 1.92 - 1.84 (m, 1H), 1.60 (d, J = 2.0 Hz, 1H), 1.57 - 1.53 (m, 3H), 1.47 - 1.34 (m, 3H), 1.14 - 1.01 (m, 1H). Step D. Procedure for preparation of 2-((1s,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)acetaldehyde To a solution of DMSO (6.98 g, 89.39 mmol, 6.98 mL, 4 equiv.) in DCM (20 mL) was added oxalyl dichloride (5.67 g, 44.70 mmol, 3.91 mL, 2 equiv.) in DCM (5 mL). The mixture was stirred at -70 °C for 1 hour.2-[4-(3-bromo-2-methyl-phenoxy)cyclohexyl]ethanol (7 g, 22.35 mmol, 1 equiv.) in DCM (5 mL) was added to the mixture at -70 °C and stirred for 1 hour. TEA (13.57 g, 134.09 mmol, 18.66 mL, 6 equiv.) was added into the mixture at -70 °C. The mixture was stirred at -70 °C for 1 hour. The reaction mixture was quenched by 50 mL H2O and extracted with DCM 150 mL (50 mL × 3). The combined organic layers were washed with saturated salt solution (20 mL × 1), dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~5% ethyl acetate/petroleum ether) to give 2-((1s,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)acetaldehyde (4 g, crude) as a light yellow oil. 1 H NMR (400 MHz, CDCl3) δ = 9.79 (t, J = 2.0 Hz, 1H), 7.15 (d, J = 8.0 Hz, 1H), 6.97 (t, J = 8.0 Hz, 1H), 6.80 (d, J = 8.0 Hz, 1H), 4.16 - 4.05 (m, 1H), 2.39 - 2.34 (m, 2H), 2.30 (s, 3H), 2.19 - 2.11 (m, 2H), 2.01 - 1.97 (m, 1H), 1.89 (d, J = 13.2 Hz, 2H), 1.55 - 1.50 (m, 2H), 1.21 - 1.08 (m, 2H). Step E. Procedure for preparation of tert-butyl 4-((E)-3-((1s,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)prop-1-en-1-yl)piperidine-1-carboxy late To a solution of (1-tert-butoxycarbonyl-4-piperidyl)methyl-triphenyl-phosphon ium;iodide (453.04 mg, 771.18 μmol, 1.2 equiv.) in THF (10 mL) was added LDA (2 M, 963.97 μL, 3 equiv.) at 0°C. The mixture was stirred for 2 hours at 0 °C. 2-((1s,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)acetaldehyde (200 mg, 642.65 μmol, 1 equiv.) in THF (5 mL) was added into the mixture at 0 °C. The mixture was stirred at 25 °C for 12 hours. The reaction mixture was diluted with NH 4 Cl (30 mL) and extracted with EtOAc (20 mL × 3). The combined organic layers were washed with aqueous NaCl (30 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by reverse- phase HPLC (with 0.1% formic acid) to give tert-butyl 4-((E)-3-((1s,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)prop-1-en-1-yl)piperidine-1-carboxy late (270 mg, 460.52 μmol, 35.83% yield) as a yellow oil. MS (ESI) m/z: 436.3 [M-56+H] + Step F. Procedure for preparation of tert-butyl 4-(3-((1s,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propyl)piperidine-1-carboxylate To a solution of tert-butyl 4-((E)-3-((1s,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)prop -1- en-1-yl)piperidine-1-carboxylate (270 mg, 548.24 μmol, 1 equiv.) in EtOH (10 mL) was added PtO 2 (99.59 mg, 438.59 μmol, 0.8 equiv.) under N 2 atmosphere. The suspension was degassed and purged with H 2 three times. The mixture was stirred under H 2 (15 Psi) at 25 °C for 1 hour. The reaction solution was filtered through the diatom layer under a moderate N2 atmosphere to give tert-butyl 4-(3-((1s,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propyl)p iperidine-1- carboxylate (250 mg, crude) as a yellow oil. MS (ESI) m/z: 438.3 [M+H-56] + Step G. Procedure for preparation of 4-(3-((1s,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propyl)piperidine A mixture of tert-butyl 4-(3-((1s,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propyl)piperidine-1-carboxylate (250 mg, 505.56 μmol, 1 equiv.) in HCl/dioxane (10 mL) was degassed and purged with N 2 three times. The mixture was stirred at 25 °C for 2 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by reverse-phase HPLC (with 0.1% formic acid) to give 4-(3-((1s,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propyl)piperidine (220 mg, crude) as a yellow oil. MS (ESI) m/z: 394.3 [M+H] + Step H. Procedure for preparation of 3-(6-iodo-1-methyl-indazol-3-yl)piperidine-2,6- dione A solution of 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (1 g, 3.87 mmol, 1 equiv.) in HCl (12 M, 1.61 mL, 5 equiv.) and H 2 O (5 mL) was stirred for 30 min. The mixture was cooled to 0°C, and NaNO2 (280.51 mg, 4.07 mmol, 1.05 equiv.) in H2O (5 mL) was added. The mixture was stirred at 0°C for 30 min. KI (1.29 g, 7.74 mmol, 2 equiv.) in H2O (10 mL) was added to the mixture at 0°C. The mixture was stirred at 25°C for 12 hours. The reaction mixture was quenched with H2O (60 mL) and extracted with EtOAc (10 mL × 3). The combined organic layers were washed with brine (20 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~40% ethyl acetate/petroleum ether) to give 3-(6-iodo-1-methyl- indazol-3-yl)piperidine-2,6-dione (400 mg, 975.20 μmol, 25.19% yield, 90% purity) as a yellow solid. MS (ESI) m/z: 370.1 [M+H] + Step I. Procedure for preparation of 3-(6-(4-(3-((1s,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propyl)piperidin-1-yl)-1-methyl-1H- indazol-3-yl)piperidine- 2,6-dione A mixture of 3-(6-iodo-1-methyl-indazol-3-yl)piperidine-2,6-dione (101.09 mg, 273.84 μmol, 1.2 equiv.), 4-(3-((1s,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propyl)p iperidine-1- carboxylate (90 mg, 228.20 μmol, 1 equiv.), Cs2CO3 (223.06 mg, 684.61 μmol, 3 equiv.), and 1,3-bis[2,6-bis(1-propylbutyl)phenyl]-4,5-dichloro-2H-imidaz ol-1-ium-2-ide;3- chloropyridine;dichloropalladium (44.40 mg, 45.64 μmol, 0.2 equiv.) in dioxane (4 mL) was degassed and purged with N 2 three times. The mixture was stirred at 90 °C for 5 hours under N2 atmosphere in a microwave reactor. The reaction mixture was quenched with H2O (20 mL) and extracted with EtOAc (10 mL × 3). The combined organic layers were washed with brine (20 mL × 1), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~35% ethyl acetate/petroleum ether) to give 3-(6-(4-(3-((1s,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propyl)piperidin-1-yl)-1-methyl-1H- indazol-3-yl)piperidine-2,6- dione (20 mg, 27.06 μmol, 11.86% yield) as a yellow solid. MS (ESI) m/z: 635.4 [M+H] + Step J. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4s)-4-(3-(1-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperidin-4-yl)propyl)cyclohexyl)oxy) -2- methylphenyl)picolinate A mixture of 3-(6-(4-(3-((1s,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)pr opyl)piperidin- 1-yl)-1-methyl-1H-indazol-3-yl)piperidine-2,6-dione (20 mg, 31.46 μmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-2-carboxylate (23.13 mg, 37.76 μmol, 1.2 equiv.), KF (3.66 mg, 62.93 μmol, 1.47 μL, 2 equiv.), and Ad2nBuP Pd G3 (2.29 mg, 3.15 μmol, 0.1 equiv.) in dioxane (3 mL) and H 2 O (0.5 mL) was degassed and purged with N 2 three times. The mixture was stirred at 100 °C for 1 hour under N2 atmosphere in a microwave reactor. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , petroleum ether : ethyl acetate = 0 : 1) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1r,4s)-4-(3-(1-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperidin -4-yl)propyl)cyclohexyl)oxy)- 2-methylphenyl)picolinate (15 mg, 10.08 μmol, 32.05% yield) as a yellow oil. MS (ESI) m/z: 1041.6 [M+H] + Step K. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4s)-4-(3-(1-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperidin-4-yl)propyl)cyclohexyl)oxy) -2-methylphenyl) picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1r,4s)-4-(3-(1-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)piperidin-4- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (15 mg, 14.41 μmol, 1 equiv.) in DCM (2 mL) and TFA (2 mL) was degassed and purged with N 2 three times. The mixture was stirred at 25 °C for 12 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1r,4s)-4-(3-(1-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperidin -4-yl)propyl)cyclohexyl)oxy)- 2-methylphenyl) picolinic acid (12.54 mg, 11.97 μmol, 83.09% yield) as a yellow solid. MS (ESI) m/z: 985.8 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.91 - 12.82 (m, 1H), 12.67 - 12.44 (m, 1H), 10.84 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.65 - 7.59 (m, 1H), 7.51 - 7.42 (m, 4H), 7.41 - 7.30 (m, 2H), 7.11 - 7.02 (m, 1H), 6.96 (d, J = 8.8 Hz, 1H), 6.93 - 6.83 (m, 3H), 6.60 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.61 (s, 1H), 4.30 - 4.21 (m, 1H), 3.96 - 3.85 (m, 5H), 3.80 - 3.71 (m, 2H), 3.05 - 3.00 (m, 2H), 2.75 - 2.58 (m, 4H), 2.20 - 2.11 (m, 2H), 1.94 - 1.87 (m, 5H), 1.78 - 1.72 (m, 2H), 1.56 - 1.47 (m, 4H), 1.34 - 1.23 (m, 12H) Example 255. Preparation of Compound 313 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-4-piperidyl]propoxy]-2- (trifluoromethyl)phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 4-[3-[3-bromo-2- (trifluoromethyl)phenoxy]propyl]piperidine-1-carboxylate To a solution of 1-bromo-3-fluoro-2-(trifluoromethyl)benzene (500 mg, 2.06 mmol, 1 equiv.) in MeCN (6 mL) was added t-BuOK (692.68 mg, 6.17 mmol, 3 equiv.) and tert-butyl 4-(3- hydroxypropyl)piperidine-1-carboxylate (500.71 mg, 2.06 mmol, 1 equiv.). The mixture was stirred at 60 °C for 12 hours. The reaction was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~7% ethyl acetate/petroleum ether) to give tert-butyl 4-[3-[3-bromo-2- (trifluoromethyl)phenoxy]propyl]piperidine-1-carboxylate (857 mg, 1.82 mmol, 88.66% yield) as a white solid. MS (ESI) m/z: 412.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 7.50 - 7.44 (m, 1H), 7.38 (d, J = 8.0 Hz, 1H), 7.27 (d, J = 8.4 Hz, 1H), 4.08 (t, J = 6.4 Hz, 2H), 3.91 (d, J = 12.0 Hz, 2H), 2.75 - 2.57 (m, 2H), 1.78 - 1.69 (m, 2H), 1.63 (d, J = 11.6 Hz, 2H), 1.38 (s, 9H), 1.37 - 1.30 (m, 3H), 1.01 - 0.88 (m, 2H). Step B. Procedure for preparation of 4-[3-[3-bromo-2- (trifluoromethyl)phenoxy]propyl]piperidine A solution of tert-butyl 4-[3-[3-bromo-2-(trifluoromethyl)phenoxy]propyl]piperidine-1 - carboxylate (857 mg, 1.84 mmol, 1 equiv.) in HCl/dioxane (10 mL) was stirred at 25 °C for 12 hours. The reaction was concentrated under reduced pressure to give 4-[3-[3-bromo-2- (trifluoromethyl)phenoxy]propyl]piperidine (730 mg, 1.80 mmol, 97.9% yield) as a yellow solid. MS (ESI) m/z: 366.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 9.34 - 8.36 (m, 2H), 7.52 - 7.44 (m, 1H), 7.38 (d, J = 8.0 Hz, 1H), 7.28 (d, J = 8.4 Hz, 1H), 4.10 (t, J = 6.0 Hz, 2H), 3.22 (d, J = 10.8 Hz, 2H), 2.89 - 2.71 (m, 2H), 1.83 - 1.68 (m, 4H), 1.63 - 1.47 (m, 1H), 1.41 - 1.23 (m, 4H). Step C. Procedure for preparation of ethyl 2-[4-[3-[3-bromo-2- (trifluoromethyl)phenoxy]propyl]-1-piperidyl]acetate To a solution of 4-[3-[3-bromo-2-(trifluoromethyl)phenoxy]propyl]piperidine (730 mg, 1.81 mmol, 1 equiv., HCl) in MeCN (8 mL) was added K2CO3 (751.65 mg, 5.44 mmol, 3 equiv.) and ethyl 2-bromoacetate (272.48 mg, 1.63 mmol, 180.57 μL, 0.9 equiv.). The mixture was stirred at 60 °C for 1 hour. The reaction was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~45% ethyl acetate/petroleum ether) to give ethyl 2-[4-[3-[3-bromo-2-(trifluoromethyl)phenoxy]propyl]- 1-piperidyl]acetate (650 mg, 1.42 mmol, 78.07% yield, 98.48% purity) as a yellow solid. MS (ESI) m/z: 452.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 7.50 - 7.44 (m, 1H), 7.37 (d, J = 8.0 Hz, 1H), 7.27 (d, J = 8.4 Hz, 1H), 4.06 (q, J = 7.2 Hz, 4H), 3.15 (s, 2H), 2.79 (d, J = 11.6 Hz, 2H), 2.15 - 2.05 (m, 2H), 1.76 - 1.68 (m, 2H), 1.61 (d, J = 11.2 Hz, 2H), 1.37 - 1.29 (m, 2H), 1.18 (t, J = 7.2 Hz, 4H), 1.11 (dd, J = 3.6, 12.0 Hz, 2H). Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-(2-ethoxy-2-oxo- ethyl)-4- piperidyl]propoxy]-2-(trifluoromethyl)phenyl]pyridine-2-carb oxylate Ethyl 2-[4-[3-[3-bromo-2-(trifluoromethyl)phenoxy]propyl]-1-piperi dyl]acetate (200 mg, 442.18 μmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)pyridine-2-carboxylate (297.94 mg, 486.40 μmol, 1.1 equiv.), Ad 2 nBuP Pd G 3 (64.41 mg, 88.44 μmol, 0.2 equiv.), and KF (1.5 M, 884.36 μL, 3 equiv.) were taken up into a microwave tube in dioxane (5 mL), and the tube was sealed. The sealed tube was heated at 100 °C for 1 hour in a microwave reactor. The reaction was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~7% dichloromethane : methanol) to give tert- butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[1-(2- ethoxy-2-oxo-ethyl)-4-piperidyl]propoxy]-2-(trifluoromethyl) phenyl]pyridine-2-carboxylate (310 mg, 275.54 μmol, 62.31% yield) as a brown oil. MS (ESI) m/z: 430.0 [M/2+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.87 (s, 1H), 8.02 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.60 (d, J = 8.0 Hz, 1H), 7.50 - 7.44 (m, 3H), 7.41 - 7.32 (m, 4H), 6.95 (d, J = 8.8 Hz, 1H), 6.65 (d, J = 7.6 Hz, 1H), 4.10 - 4.03 (m, 6H), 3.88 (t, J = 6.0 Hz, 2H), 3.18 - 3.12 (m, 3H), 3.03 (t, J = 5.6 Hz, 2H), 2.78 (d, J = 8.4 Hz, 2H), 2.27 - 2.23 (m, 1H), 2.09 (s, 2H), 1.75 - 1.70 (m, 3H), 1.59 (s, 2H), 1.36 - 1.33 (m, 2H), 1.05 - 0.99 (m, 12H). Step E. Procedure for preparation of 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyr idyl]-2- (trifluoromethyl)phenoxy]propyl]-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[3-[1-(2-ethoxy-2-oxo-ethyl)-4-piperidyl]propoxy] -2- (trifluoromethyl)phenyl]pyridine-2-carboxylate (310 mg, 361.31 μmol, 1 equiv.) in THF (6 mL) and H2O (2 mL) was added LiOH•H2O (75.81 mg, 1.81 mmol, 5 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction was concentrated under reduced pressure and diluted with HCl (1M). The pH of the reaction mixture was adjusted to 2~3. The resulting solid was filtered and dried. The solid was purified by prep-TLC (SiO2, DCM: MeOH = 8:1) to give 2- [4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-2-tert- butoxycarbonyl-3-pyridyl]-2-(trifluoromethyl)phenoxy]propyl] -1-piperidyl]acetic acid (140 mg, 145.71 μmol, 40.33% yield) as a yellow solid. MS (ESI) m/z: 830.6 [M+H] + . Step F. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-4-piperidyl]propoxy]-2-(trifluorome thyl)phenyl]pyridine-2- carboxylate A solution of 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H-isoquinolin- 2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-(trifluoromethyl)ph enoxy]propyl]-1-piperidyl]acetic acid (75 mg, 90.37 μmol, 1 equiv.) and 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (70.02 mg, 271.11 μmol, 3 equiv.) in pyridine (1 mL) was stirred at 40 °C for 12 hours. To this mixture was added EDCI (25.99 mg, 135.55 μmol, 1.5 equiv.). The resulting mixture was stirred at 40 °C for 12 hours. The reaction mixture was diluted with water (5 mL) and extracted with DCM (5 mL x 3). The combined organic layers were dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by reversed- phase HPLC (with 0.1% formic acid) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-diox o-3-piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-4-piperidyl]propoxy]-2-(trifluorome thyl)phenyl]pyridine-2- carboxylate (45 mg, 40.67 μmol, 45.0% yield) as a white solid. MS (ESI) m/z: 1070.4 [M+H] + . Step G. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-piperidyl]propoxy]-2-(trifluorometh yl)phenyl]pyridine-2- carboxylic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indaz ol-6-yl]amino]-2-oxo-ethyl]-4- piperidyl]propoxy]-2-(trifluoromethyl)phenyl]pyridine-2-carb oxylate (40 mg, 37.38 μmol, 1 equiv.) in TFA (1 mL) and DCM (1 mL) was stirred at 25 °C for 12 hours. The solution was concentrated under reduced pressure to give a residue. The residue was purified by reverse- phase HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indaz ol-6-yl]amino]-2-oxo-ethyl]-4- piperidyl]propoxy]-2-(trifluoromethyl)phenyl]pyridine-2-carb oxylic acid (31.12 mg, 30.06 μmol, 80.4% yield) as a yellow solid. MS (ESI) m/z: 1015.4 [M/2+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.87 (s, 1H), 9.82 (s, 1H), 8.08 - 7.98 (m, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 8.4 Hz, 2H), 7.50 - 7.43 (m, 3H), 7.40 - 7.31 (m, 3H), 7.25 - 7.15 (m, 2H), 7.00 - 6.93 (m, 1H), 6.68 (d, J = 7.6 Hz, 1H), 5.06 - 4.92 (m, 2H), 4.32 (d, J = 5.2, 9.6 Hz, 1H), 4.09 (t, J = 5.6 Hz, 2H), 3.97 - 3.88 (m, 5H), 3.12 (s, 2H), 3.02 (s, 2H), 2.90 - 2.84 (m, 4H), 2.20 - 2.13 (m, 4H), 1.79 - 1.73 (m, 2H), 1.67 (d, J = 7.2 Hz, 2H), 1.44 - 1.37 (m, 2H), 1.32 - 1.24 (m, 3H) Example 256. Preparation of Compound 314 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2- oxoethyl)piperidin-4- yl)propoxy)phenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 4-(3-(3-bromophenoxy)propyl)piperidine- 1-carboxylate A mixture of 3-bromophenol (500 mg, 2.89 mmol, 1 equiv.), tert-butyl 4-(3- hydroxypropyl)piperidine-1-carboxylate (703.27 mg, 2.89 mmol, 1 equiv.), and 2-(tributyl- phosphanylidene)acetonitrile (837.03 mg, 3.47 mmol, 1.2 equiv.) in toluene (6 mL) was degassed and purged with N2 three times. The mixture was stirred at 120 °C for 16 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 20 % ethyl acetate / petroleum ether) to give tert-butyl 4-(3-(3-bromophenoxy)propyl)piperidine-1- carboxylate (850 mg, 2.13 mmol, 73.8 % yield) as a white oil. MS (ESI) m/z: 342.1 [M+H] + . Step B. Procedure for preparation of 4-(3-(3-bromophenoxy)propyl)piperidine A mixture of tert-butyl 4-(3-(3-bromophenoxy)propyl)piperidine-1-carboxylate (850 mg, 2.13 mmol, 1 equiv.) in TFA (4 mL) and DCM (4 mL) was stirred at 40 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give 4-(3-(3- bromophenoxy)propyl)piperidine (630 mg, 2.11 mmol, 99.0 % yield) as a yellow oil. MS (ESI) m/z: 298.1 [M+H] + . Step C. Procedure for preparation of ethyl 2-(4-(3-(3-bromophenoxy)propyl)piperidin-1- yl)acetate A mixture of 4-(3-(3-bromophenoxy)propyl)piperidine (630 mg, 2.11 mmol, 1 equiv.), ethyl 2- bromoacetate (352.80 mg, 2.11 mmol, 233.80 μL, 1 equiv.), and K 2 CO 3 (1.46 g, 10.56 mmol, 5 equiv.) in MeCN (8 mL) was stirred at 60 °C for 2 hours. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 40 % ethyl acetate / petroleum ether) to give ethyl 2-(4- (3-(3-bromophenoxy)propyl)piperidin-1-yl)acetate (720 mg, 1.87 mmol, 88.6 % yield) as a yellow oil. MS (ESI) m/z: 384.1 [M+H] + . Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3, 4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-ethoxy-2-oxoeth yl) piperidin-4- yl)propoxy)phenyl)picolinate A mixture of ethyl 2-(4-(3-(3-bromophenoxy)propyl)piperidin-1-yl)acetate (150 mg, 390.31 μmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) picolinate (286.90 mg, 468.38 μmol, 1.2 equiv.), K2CO3 (1.5 M, 390.31 μL, 1.5 eq), and [2-(2-aminophenyl)phenyl]palladium(1+); bis(1-adamantyl)-butyl-phosphane; methanesulfonate (56.85 mg, 78.06 μmol, 0.2 equiv.) in dioxane (2 mL) was stirred at 100 °C for 1.5 hours in a microwave reactor. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 40 % ethyl acetate / petroleum ether) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3, 4-dihydroisoquinolin-2(1H)- yl)-3-(3-(3-(1-(2-ethoxy-2-oxoethyl) piperidin-4-yl)propoxy)phenyl)picolinate (200 mg, 253.17 μmol, 64.6 % yield) as a yellow solid. MS (ESI) m/z: 790.6 [M+H] + . Step E. Procedure for preparation of 2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyri din-3- yl)phenoxy)propyl)piperidin-1-yl)acetic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3, 4-dihydroisoquinolin-2(1H)- yl)-3-(3-(3-(1-(2-ethoxy-2-oxoethyl) piperidin-4-yl)propoxy)phenyl)picolinate (200 mg, 253.17 μmol, 1 equiv.) and LiOH·H 2 O (53.12 mg, 1.27 mmol, 5 equiv.) in THF (4 mL) and H2O (2 mL) was stirred at 40 °C for 2 hours. The reaction mixture was diluted with H2O (5 mL) and concentrated to provide a turbid liquid. Then the pH of the turbid liquid was adjusted to ~3 with HCl (1 M, 5 mL). The resulting mixture was filtered, dried, and extracted with DCM / MeOH (10 : 1). The organic layers were combined and concentrated under reduced pressure to give 2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin-2(1H)-yl)-2- (tert-butoxycarbonyl)pyridin-3-yl)phenoxy)propyl)piperidin-1 -yl)acetic acid (180 mg, 236.24 μmol, 93.31 % yield) as a yellow solid. MS (ESI) m/z: 762.6 [M+H] + . Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)propoxy)phenyl )picolinate A mixture of 2-(4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)phenoxy)propyl)piper idin-1-yl)acetic acid (100 mg, 131.25 μmol, 1 equiv.), 3-(6-amino-1-methyl-1H-indazol-3-yl)piperidine-2,6-dione (40.68 mg, 157.50 μmol, 1.2 equiv.), and EDCI (32.71 mg, 170.62 μmol, 1.3 equiv.) in pyridine (1 mL) was stirred at 40 °C for 2 hours. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 10 % ethyl acetate / petroleum ether) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-(1-(2-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2-oxoet hyl)piperidin-4- yl)propoxy)phenyl)picolinate (80 mg, 79.83 μmol, 60.82 % yield) as a black solid. MS (ESI) m/z: 1002.7 [M+H] + . Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)propoxy)phenyl )picolinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2- oxoethyl)piperidin-4-yl)propoxy)phenyl)picolinate (80 mg, 79.83 μmol, 1 equiv.) in TFA (0.6 mL) and DCM (0.6 mL) was stirred at 40 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep–HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino )-2-oxoethyl)piperidin-4- yl)propoxy)phenyl)picolinic acid (30.38 mg, 31.68 μmol, 39.69 % yield) as a white solid. MS (ESI) m/z: 946.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 13.07 - 12.74 (m, 1H), 10.88 (s, 1H), 9.85 (s, 1H), 8.14 (s, 1H), 8.07 - 8.01 (m, 2H), 7.80 (d, J = 7.6 Hz, 1H), 7.68 - 7.61 (m, 3H), 7.50 - 7.43 (m, 2H), 7.39 - 7.33 (m, 2H), 7.27 (t, J = 8.4 Hz, 1H), 7.22 (d, J = 7.6 Hz, 1H), 6.96 (d, J = 9.2 Hz, 1H), 6.91 - 6.85 (m, 3H), 4.98 (s, 2H), 4.33 (dd, J = 5.2, 10.0 Hz, 1H), 3.98 - 3.94 (m, 2H), 3.93 - 3.88 (m, 5H), 3.17 - 3.14 (m, 2H), 3.02 (t, J = 5.2 Hz, 2H), 2.89 (d, J = 10.0 Hz, 2H), 2.68 - 2.63 (m, 2H), 2.33 (s, 3H), 2.20 - 2.16 (m, 2H), 1.78 - 1.67 (m, 4H), 1.40 - 1.34 (m, 2H), 1.29 (s, 2H) Example 258. Preparation of Compound 316a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3- (4-(3-(2,4-dioxotetrahydropyrimidin-1(2H)-yl)-1-methyl-1H-in dazol-6-yl)piperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of ethyl (E)-3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)acrylate To a mixture of NaH (2.22 g, 55.52 mmol, 60% purity, 1.5 equiv.) in THF (300 mL) was added dropwise ethyl 2-diethoxyphosphorylacetate (11.62 g, 51.82 mmol, 10.28 mL, 1.4 equiv.) at 0 °C under N 2 . The mixture was stirred at 0 °C for 1 hour under N 2 . To this mixture was added (1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexane-1-carbaldehyd e (11 g, 37.01 mmol, 1.0 equiv.). The resulting reaction mixture was stirred at 25 °C for 2 hours under N2 atmosphere. The reaction mixture was quenched with aqueous NH 4 Cl (250 mL) at 0 °C and extracted with ethyl acetate (250 mL × 2). The combined organic layers were washed with brine (250 mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0-10% ethyl acetate / petroleum ether) to give compound ethyl (E)-3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)acrylat e (24 g, 65.35 mmol, 88.2% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ = 7.18 - 7.12 (m, 1H), 7.10 - 7.03 (m, 2H), 6.85 (m, 1H), 5.90 - 5.72 (m, 1H), 4.31 - 4.22 (m, 1H), 4.11 (q, J = 7.2 Hz, 2H), 2.30 - 2.23 (m, 1H), 2.21 (s, 3H), 2.08 (m, 2H), 1.81 (m, 2H), 1.51 - 1.25 (m, 4H), 1.21 (t, J = 7.2 Hz, 3H). Step B. Procedure for preparation of ethyl 3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propanoate A mixture of ethyl (E)-3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)acrylat e (12 g, 32.67 mmol, 1.0 equiv.) and PtO2 (2.97 g, 13.07 mmol, 0.4 equiv.) in EtOH (200 mL) was degassed and purged with N2 three times. The mixture was stirred at 25 °C for 16 hours under H 2 (15 Psi) atmosphere. The mixture was filtered washed with ethyl acetate (100 mL). The filtrate was concentrated under reduced pressure to give ethyl 3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propanoate (24 g, 64.99 mmol, 99.4% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.15 - 7.11 (m, 1H), 7.06 (t, J = 8.0 Hz, 1H), 7.02 - 6.98 (m, 1H), 4.26 - 4.16 (m, 1H), 4.04 (q, J = 7.2 Hz, 2H), 2.32 - 2.24 (m, 2H), 2.20 (s, 3H), 2.03 (m, 2H), 1.74 (m, 2H), 1.45 (q, J = 7.2 Hz, 2H), 1.37 - 1.23 (m, 3H), 1.17 (t, J = 7.2 Hz, 3H), 1.08 - 1.01 (m, 2H). Step C. Procedure for preparation of 3-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propan-1-ol To a solution of ethyl 3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propanoate (12 g, 32.49 mmol, 1.0 equiv.) in THF (150 mL) was added LiAlH 4 (1.23 g, 32.49 mmol, 1.0 equiv.). The mixture was degassed and purged with N2 three times. The reaction mixture was then stirred at 0 °C for 2 hours under N 2 atmosphere. The reaction mixture was quenched by the addition of Na 2 SO 4 •10H 2 O (20 g) at 0 °C under a N 2 atmosphere, filtered, and concentrated under reduced pressure to give 3-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propan-1- ol (20.8 g, 63.56 mmol, 97.8% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.15 - 7.11 (m, 1H), 7.06 (t, J = 8.0 Hz, 1H), 7.03 - 6.98 (m, 1H), 4.34 (m, 1H), 4.26 - 4.17 (m, 1H), 3.37 (m, 2H), 2.20 (s, 3H), 2.08 - 1.99 (m, 2H), 1.81 - 1.71 (m, 2H), 1.45 - 1.39 (m, 2H), 1.38 - 1.29 (m, 2H), 1.20 (m, 7.2 Hz, 3H), 1.08 - 0.96 (m, 2H) Step D. Procedure for preparation of 3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propanal A mixture of 3-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propan-1-ol (50 mg, 152.79 μmol, 1.0 equiv.) and DMP (77.76 mg, 183.34 μmol, 56.80 μL, 1.2 equiv.) in DCM (1 mL) was degassed and purged with N2 three times. The mixture was stirred at 25 °C for 20 hours under N 2 atmosphere. The mixture was filtered and washed with DCM (20 mL). The filtrate was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, petroleum ether : ethyl acetate = 3:1) to give 3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propanal (30 mg, 92.24 μmol, 60.3% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 9.67 (s, 1H), 7.16 - 7.11 (m, 1H), 7.09 - 6.99 (m, 2H), 4.29 - 4.16 (m, 1H), 2.47 - 2.41 (m, 2H), 2.20 (s, 3H), 2.04 (m, 2H), 1.75 (m, 2H), 1.45 (q, J = 7.2 Hz, 2H), 1.38 - 1.23 (m, 3H), 1.10 - 0.98 (m, 2H). Step E. Procedure for preparation of 1-(6-(4-(3-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propyl)piperazin-1-yl)-1-methyl-1H- indazol-3- yl)dihydropyrimidine-2,4(1H,3H)-dione A solution of 3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propanal (200 mg, 614.93 μmol, 1.0 equiv.), 1-(1-methyl-6-piperazin-1-yl-indazol-3-yl)hexahydropyrimidin e-2,4-dione (288.46 mg, 737.92 μmol, 1.2 equiv.), and KOAc (60.35 mg, 614.93 μmol, 1.0 equiv.) in DCM (6 mL) was stirred at 25 °C for 1 hour. To this solution was added NaBH(OAc)3 (390.99 mg, 1.84 mmol, 3.0 equiv.), and the resulting mixture was stirred at 25 °C for 2 hours. After completion, the reaction mixture was poured into water (20 mL), and extracted with ethyl acetate (20 mL × 3). The combined organic layers were washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , DCM:MeOH = 10:1) to give 1-(6-(4-(3-((1r,4s)- 4-(3-bromo-2-methylphenoxy)cyclohexyl)propyl)piperazin-1-yl) -1-methyl-1H-indazol-3- yl)dihydropyrimidine-2,4(1H,3H)-dione (120 mg, 166.28 μmol, 27.04% yield) as a yellow solid. MS (ESI) m/z: 637.4 [M+H] + . Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,4- dioxotetrahydropyrimidin- 1(2H)-yl)-1-methyl-1H-indazol-6-yl)piperazin-1-yl)propyl)cyc lohexyl)oxy)-2- methylphenyl)picolinate A mixture of 1-(6-(4-(3-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)pr opyl)piperazin- 1-yl)-1-methyl-1H-indazol-3-yl)dihydropyrimidine-2,4(1H,3H)- dione (120 mg, 188.20 μmol, 1.0 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine- 2-carboxylate (172.92 mg, 282.30 μmol, 1.5 equiv.), Ad2nBuP Pd G 3 (13.71 mg, 18.82 μmol, 0.1 equiv.), and KF (32.80 mg, 564.61 μmol, 13.23 μL, 3.0 equiv.) in dioxane (4 mL) and H2O (0.4 mL) was degassed and purged with N2 three times. The mixture was stirred at 100 °C for 2 hours under N2 atmosphere in a microwave reactor. After completion, the reaction mixture was poured into water (50 mL) and extracted with DCM (50 mL × 3). The combined organic layers were washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , DCM:MeOH = 10:1) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- (3-(((1s,4r)-4-(3-(4-(3-(2,4-dioxotetrahydropyrimidin-1(2H)- yl)-1-methyl-1H-indazol-6- yl)piperazin-1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl)pico linate (110 mg, 99.80 μmol, 53.03% yield) as a yellow solid. MS (ESI) m/z: 1043.8 [M+H] + . Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,4- dioxotetrahydropyrimidin- 1(2H)-yl)-1-methyl-1H-indazol-6-yl)piperazin-1-yl)propyl)cyc lohexyl)oxy)-2- methylphenyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,4-dioxotetrahydropyrimidin- 1(2H)-yl)-1-methyl-1H-indazol- 6-yl)piperazin-1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl)pi colinate (110 mg, 105.44 μmol, 1.0 equiv.) in DCM (1 mL) and TFA (2 mL) was stirred at 40 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,4-dioxotetrahydropyrimidin- 1(2H)-yl)-1-methyl-1H-indazol- 6-yl)piperazin-1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl)pi colinic acid (71.61 mg, 65.81 μmol, 62.42% yield) as a white solid. MS (ESI) m/z: 987.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.99 - 12.75 (m, 1H), 10.51 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (m, 1H), 7.62 (m, 1H), 7.53 - 7.46 (m, 2H), 7.45 (m, 2H), 7.40 - 7.33 (m, 2H), 7.11 - 7.04 (m, 1H), 6.98 - 6.91 (m, 4H), 6.62 (m, 1H), 4.98 (m, 2H), 4.26 - 4.17 (m, 1H), 3.94 - 3.88 (m, 7H), 3.03 (m, 2H), 2.74 (m, 2H), 2.60 (m, 2H), 2.46 - 2.30 (m, 7H), 2.10 (m, 2H), 1.87 (s, 3H), 1.81 (m, J = 10.4 Hz, 2H), 1.72 - 1.62 (m, 2H), 1.41 - 1.30 (m, 3H), 1.28 - 1.18 (m, 3H), 1.14 - 1.05 (m, 2H) Example 259. Preparation of Compound 328a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1- yl)propyl)cyclohexyl)oxy)-2-(trifluoromethyl)phenyl)picolini c acid Step A. Procedure for preparation of ethyl (E)-3-((1r,4r)-4-(3-bromo-2- (trifluoromethyl)phenoxy)cyclohexyl)acrylate To a solution of (1r,4r)-4-(3-bromo-2-(trifluoromethyl)phenoxy)cyclohexane-1- carbaldehyde (368.69 mg, 1.64 mmol, 326.28 μL, 1.5 equiv.) in THF (20 mL) was added LiHMDS (1 M, 2.19 mL, 2 equiv.) at -10 °C. The reaction mixture was stirred for 1 hour. Then (1r,4r)-4-(3- bromo-2-(trifluoromethyl)phenoxy)cyclohexane-1-carbaldehyde (385 mg, 1.10 mmol, 1 equiv.) was added, and the mixture was stirred at 0 °C for 1 hour under N2 atmosphere. The reaction mixture was quenched with NH 4 Cl (20 mL) at 0 °C and extracted with ethyl acetate (20 mL). The combined organic layers were washed with brine (20 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0-4% ethyl acetate/petroleum ether) to give ethyl (E)-3-((1r,4r)-4-(3-bromo-2-(trifluoromethyl)phenoxy)cyclohe xyl)acrylate (430 mg, 918.70 μmol, 83.52% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.50 - 7.41 (m, 1H), 7.40 - 7.32 (m, 2H), 6.84 (dd, J = 6.4, 16.0 Hz, 1H), 5.83 (d, J = 16.0 Hz, 1H), 4.57 - 4.41 (m, 1H), 4.11 (q, J = 7.2 Hz, 2H), 2.25 (d, J = 6.8 Hz, 1H), 2.07 (d, J = 10.0 Hz, 2H), 1.82 (d, J = 12.0 Hz, 2H), 1.50 - 1.38 (m, 2H), 1.37 - 1.25 (m, 2H), 1.20 (t, J = 7.2 Hz, 3H). Step B. Procedure for preparation of ethyl 3-((1r,4r)-4-(3-bromo-2- (trifluoromethyl)phenoxy)cyclohexyl)propanoate To a solution of ethyl (E)-3-((1r,4r)-4-(3-bromo-2- (trifluoromethyl)phenoxy)cyclohexyl)acrylate (430 mg, 1.02 mmol, 1 equiv.) in EtOH (4 mL) was added PtO2 (23.18 mg, 102.08 μmol, 0.1 equiv.) under N2 atmosphere. The suspension was degassed and purged with H 2 three times. The mixture was stirred under H 2 (15 Psi) at 25 °C for 12 hours. The reaction solution was filtered through the diatom layer under a moderate N2 atmosphere. The residue was purified by flash silica gel chromatography (0-6% ethyl acetate/petroleum ether) to give ethyl 3-((1r,4r)-4-(3-bromo-2- (trifluoromethyl)phenoxy)cyclohexyl)propanoate (300 mg, 637.90 μmol, 62.49% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.48 - 7.41 (m, 1H), 7.39 - 7.32 (m, 2H), 4.50 - 4.38 (m, 1H), 4.04 (q, J = 7.2 Hz, 2H), 2.29 (t, J = 7.6 Hz, 2H), 2.04 (d, J = 10.0 Hz, 2H), 1.75 (d, J = 12.0 Hz, 2H), 1.46 (q, J = 7.2 Hz, 2H), 1.36 - 1.28 (m, 2H), 1.26 - 1.22 (m, 1H), 1.17 (t, J = 7.2 Hz, 3H), 1.10 - 1.00 (m, 2H). Step C. Procedure for preparation of 3-((1r,4s)-4-(3-bromo-2- (trifluoromethyl)phenoxy)cyclohexyl)propan-1-ol To a solution of ethyl 3-((1r,4r)-4-(3-bromo-2- (trifluoromethyl)phenoxy)cyclohexyl)propanoate (150 mg, 354.39 μmol, 1 equiv.) in THF (10 mL) was added LiAlH4 (13.45 mg, 354.39 μmol, 1 equiv.) .The mixture was stirred at 0 °C for 1 hour. The reaction was quenched with saturated Na 2 SO 4 (3 mL). The organic phase was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0-20% ethyl acetate/petroleum ether) to give 3-((1r,4s)-4-(3-bromo-2- (trifluoromethyl)phenoxy)cyclohexyl)propan-1-ol (50 mg, 131.16 μmol, 37.01% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.48 - 7.41 (m, 1H), 7.38 - 7.32 (m, 2H), 4.50 - 4.38 (m, 1H), 4.34 (t, J = 5.2 Hz, 1H), 3.40 - 3.34 (m, 2H), 2.04 (d, J = 10.0 Hz, 2H), 1.76 (d, J = 12.4 Hz, 2H), 1.46 - 1.31 (m, 4H), 1.23 - 1.16 (m, 3H), 1.09 - 0.98 (m, 2H). Step D. Procedure for preparation of 3-((1r,4r)-4-(3-bromo-2- (trifluoromethyl)phenoxy)cyclohexyl)propanal A solution of (COCl) 2 (33.29 mg, 262.31 μmol, 22.96 μL, 2 equiv.) in DCM (5 mL) was cooled to -78 °C, and DMSO (40.99 mg, 524.62 μmol, 40.99 μL, 4 equiv.) was added dropwise. The mixture was stirred at -70 °C for 2 hours under N2 atmosphere. Then 3-((1r,4s)-4-(3-bromo-2- (trifluoromethyl)phenoxy)cyclohexyl)propan-1-ol (50 mg, 131.16 μmol, 1 equiv.) was added dropwise, and the mixture was stirred at -70 °C for 1 hour under N2 atmosphere. TEA (79.63 mg, 786.93 μmol, 109.53 μL, 6 equiv.) was added dropwise to the mixture at -70 °C, and the mixture was warmed to 25 °C and stirred at 25 °C for 1 hour under N 2 atmosphere. The reaction mixture was diluted with H2O (20 mL) and extracted with DCM (20 mL × 3). The combined organic layers were washed with brine (10 mL × 3), dried over Na2SO4, filtered, and concentrated under reduced pressure to give 3-((1r,4r)-4-(3-bromo-2- (trifluoromethyl)phenoxy)cyclohexyl)propanal (50 mg, crude) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 9.67 (t, J = 1.2 Hz, 1H), 7.48 - 7.41 (m, 1H), 7.39 - 7.33 (m, 2H), 4.50 - 4.38 (m, 1H), 2.45 (t, J = 1.6, 7.6 Hz, 2H), 2.08 - 2.00 (m, 2H), 1.79 - 1.71 (m, 2H), 1.45 (q, J = 7.2 Hz, 2H), 1.39 - 1.27 (m, 3H), 1.09 - 1.01 (m, 2H). Step E. Procedure for preparation of 3-(6-(4-(3-((1r,4s)-4-(3-bromo-2- (trifluoromethyl)phenoxy)cyclohexyl)propyl)piperazin-1-yl)-1 -methyl-1H-indazol-3- yl)piperidine-2,6-dione To a solution of 3-((1r,4r)-4-(3-bromo-2-(trifluoromethyl)phenoxy)cyclohexyl) propanal (60 mg, 158.22 μmol, 1 equiv.) and 3-(1-methyl-6-(piperazin-1-yl)-1H-indazol-3-yl)piperidine- 2,6-dione (69.08 mg, 189.87 μmol, 1.2 equiv., HCl) in DCM (3 mL) was added NaOAc (12.98 mg, 158.22 μmol, 1 equiv.). The mixture was stirred at 0 °C for 1 hour. To this mixture was then added NaBH(OAc) 3 (167.67 mg, 791.12 μmol, 5 equiv.). The mixture was stirred at 0 to 25 °C for 11 hours. The reaction mixture was quenched by addition of H 2 O (5 mL) at 25 °C and extracted with DCM (5 mL × 3). The combined organic layers were washed with brine (5 mL × 3), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0-7% MeOH/DCM) to give 3-(6- (4-(3-((1r,4s)-4-(3-bromo-2-(trifluoromethyl)phenoxy)cyclohe xyl)propyl)piperazin-1-yl)-1- methyl-1H-indazol-3-yl)piperidine-2,6-dione (110 mg, crude) as a yellow oil. MS (ESI) m/z: 690.2 [M+H] + . Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1-yl)propyl)cyclohexyl)oxy)-2- (trifluoromethyl)phenyl)picolinate A mixture of 3-(6-(4-(3-((1r,4s)-4-(3-bromo-2- (trifluoromethyl)phenoxy)cyclohexyl)propyl)piperazin-1-yl)-1 -methyl-1H-indazol-3- yl)piperidine-2,6-dione (110 mg, 159.28 μmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tet ramethyl-1,3,2-dioxaborolan-2- yl)picolinate (117.08 mg, 191.14 μmol, 1.2 equiv.), KF (27.76 mg, 477.85 μmol, 11.19 μL, 3 equiv.), and Ad2nBuP Pd G3 (23.20 mg, 31.86 μmol, 0.2 equiv.) in dioxane (2 mL) and H2O (0.2 mL) was degassed and purged with N2 three times. The mixture was stirred at 100 °C for 2 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH = 10: 1) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-6- yl)piperazin-1-yl)propyl)cyclohexyl)oxy)-2-(trifluoromethyl) phenyl)picolinate (110 mg, 89.36 μmol, 56.10% yield) as a yellow oil. MS (ESI) m/z: 1096.6 [M+H] + . Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1-yl)propyl)cyclohexyl)oxy)-2- (trifluoromethyl)phenyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)piperazin-1- yl)propyl)cyclohexyl)oxy)-2-(trifluoromethyl)phenyl)picolina te (110 mg, 100.34 μmol, 1 equiv.) in TFA (1 mL) and DCM (2 mL) was stirred at 25 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-6- yl)piperazin-1-yl)propyl)cyclohexyl)oxy)-2-(trifluoromethyl) phenyl)picolinic acid (31.76 mg, 28.28 μmol, 28.19% yield) as a white solid. MS (ESI) m/z: 1040.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 13.15 - 12.42 (m, 1H), 10.85 (s, 1H), 8.14 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.52 - 7.32 (m, 7H), 7.25 (d, J = 8.4 Hz, 1H), 7.01 - 6.88 (m, 2H), 6.85 (s, 1H), 6.64 (d, J = 7.6 Hz, 1H), 5.06 - 4.91 (m, 2H), 4.43 (t, J = 10.0 Hz, 1H), 4.26 (dd, J = 4.8, 9.2 Hz, 1H), 3.93 (t, J = 5.6 Hz, 2H), 3.89 (s, 3H), 3.25 (s, 4H), 3.02 (t, J = 5.6 Hz, 2H), 2.70 - 2.56 (m, 6H), 2.42 (d, J = 1.6 Hz, 2H), 2.35 - 2.24 (m, 2H), 2.19 - 2.13 (m, 1H), 2.12 - 2.04 (m, 2H), 1.87 - 1.75 (m, 2H), 1.52 (s, 2H), 1.42 - 1.32 (m, 2H), 1.25 (d, J = 7.2 Hz, 2H), 1.08 (q, J = 12.0 Hz, 2H). Example 260. Preparation of Compound 329a

6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1- yl)butyl)cyclohexyl)oxy)phenyl)picolinic acid Step A. Procedure for preparation of methyl (1r,4r)-4-(3-bromophenoxy)cyclohexane-1- carboxylate To a solution of 3-bromophenol (2.5 g, 14.45 mmol, 1.0 equiv.) in toluene (25 mL) was added methyl 4-hydroxycyclohexanecarboxylate (2.29 g, 14.45 mmol, 1.0 equiv.) and 2-(tributyl-λ 5 - phosphanylidene)acetonitrile (4.19 g, 17.34 mmol, 1.2 equiv.) at 25 °C. The reaction mixture was degassed and purged with N2 three times. The mixture was stirred at 120 °C for 16 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 5% ethyl acetate / petroleum ether) to give methyl (1r,4r)-4-(3-bromophenoxy)cyclohexane-1- carboxylate (6 g, 13.39 mmol, 46.34% yield) as a colorless oil. 1 H NMR (400 MHz, CDCl3-d) δ = 7.16 - 7.06 (m, 2H), 7.05 (s, 1H), 6.82 (d, J = 8.4 Hz, 1H), 4.23 - 4.16 (m, 1H), 3.70 (s, 3H), 2.43 - 2.33 (m, 1H), 2.17 (d, J = 12.4 Hz, 2H), 2.10 (s, 2H), 1.66 - 1.55 (m, 2H), 1.54 - 1.42 (m, 2H) Step B. Procedure for preparation of ((1r,4r)-4-(3-bromophenoxy)cyclohexyl)methanol To a solution of methyl (1r,4r)-4-(3-bromophenoxy)cyclohexane-1-carboxylate (3 g, 6.71 mmol, 1.0 equiv.) in THF (30 mL) was added LiAlH4 (305.36 mg, 8.05 mmol, 1.2 equiv.) at 0 °C under N 2 . The reaction mixture was stirred under N 2 at 0 °C for 1 hour. The reaction mixture was quenched with Na 2 SO 4 •10 H 2 O (1 g) and slowly warmed to 20 °C. The mixture was filtered, and the filter cake was washed with DCM (100 mL). The filtrate was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 20% ethyl acetate / petroleum ether) to give ((1r,4r)-4-(3- bromophenoxy)cyclohexyl)methanol (1.68 g, 5.68 mmol, 84.78% yield) as a yellow oil. 1 H NMR (400 MHz, CDCl3-d) δ = 7.15 - 7.10 (m, 1H), 7.08 - 7.04 (m, 2H), 6.85 - 6.80 (m, 1H), 4.20 - 4.10 (m, 1H), 3.51 (d, J = 6.4 Hz, 2H), 2.24 - 2.14 (m, 2H), 1.98 - 1.88 (m, 2H), 1.63 - 1.52 (m, 1H), 1.51 - 1.40 (m, 3H), 1.18 - 1.05 (m, 2H) Step C. Procedure for preparation of (1r,4r)-4-(3-bromophenoxy)cyclohexane-1- carbaldehyde A solution of DMSO (1.84 g, 23.56 mmol, 1.84 mL, 4.0 equiv.) in DCM (20 mL) was added dropwise to a solution of oxalyl dichloride (1.50 g, 11.78 mmol, 1.03 mL, 2.0 equiv.) in DCM (5 mL) at -70 °C under a N 2 atmosphere. The mixture was stirred at -70 °C for 30 minutes. Afterwards, ((1r,4r)-4-(3-bromophenoxy)cyclohexyl)methanol (1.68 g, 5.89 mmol, 1.0 equiv.) in DCM (5 mL) was added dropwise at -70 °C. The solution was stirred for 30 minutes at - 70 °C. Then, TEA (3.58 g, 35.35 mmol, 4.92 mL, 6.0 equiv.) was added into the solution. The solution was stirred at 25 °C for 1 hour under N2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with DCM (20 mL × 3). The combined organic layers were washed with brine (10 mL × 2), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give (1r,4r)-4-(3-bromophenoxy)cyclohexane-1-carbaldehyde (1.7 g, crude) as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 9.60 (s, 1H), 7.26 - 7.18 (m, 1H), 7.15 (t, J = 2.0 Hz, 1H), 7.12 - 7.06 (m, 1H), 6.96 (dd, J = 1.6, 8.4 Hz, 1H), 4.41 - 4.30 (m, 1H), 2.38 - 2.29 (m, 1H), 2.08 - 1.92 (m, 4H), 1.48 - 1.36 (m, 4H) Step D. Procedure for preparation of 1-(((1r,4r)-4-((E)-4-(benzyloxy)but-1-en-1- yl)cyclohexyl)oxy)-3-bromobenzene A mixture of 3-benzyloxypropyl (triphenyl) phosphonium;bromide (3.10 g, 6.30 mmol, 1.05 equiv.) in THF (15 mL) was degassed and purged with N 2 three times. To this solution was added LiHMDS (1 M, 7.20 mL, 1.2 equiv.) dropwise. The reaction mixture was stirred at - 70 °C for 2 hours under N2 atmosphere. (1r,4r)-4-(3-bromophenoxy)cyclohexane-1- carbaldehyde (1.7 g, 6.00 mmol, 1.0 equiv.) was added to the mixture, and stirred was continued under N 2 at 25 °C for 14 hours. The reaction mixture was quenched by aqueous NH 4 Cl (20 mL) at 0 °C and extracted with EtOAc (20 mL × 3). The combined organic layers were washed with brine (10 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 7% ethyl acetate/petroleum ether) to give 1-(((1r,4r)-4-((E)-4-(benzyloxy)but-1-en-1- yl)cyclohexyl)oxy)-3-bromobenzene (1.98 g, 4.77 mmol, 79.40% yield) as a colorless oil. 1 H NMR (400 MHz, CDCl 3 -d) δ = 7.40 - 7.33 (m, 4H), 7.32 - 7.27 (m, 1H), 7.16 - 7.09 (m, 1H), 7.07 (s, 2H), 6.83 (d, J = 8.4 Hz, 1H), 5.48 - 5.36 (m, 1H), 5.36 - 5.27 (m, 1H), 4.54 (s, 2H), 4.20 - 4.08 (m, 1H), 3.50 (t, J = 6.8 Hz, 2H), 2.41 (q, J = 7.2 Hz, 2H), 2.34 (dd, J = 3.6, 7.6 Hz, 1H), 2.14 (d, J = 10.4 Hz, 2H), 1.76 (d, J = 12.4 Hz, 2H), 1.53 - 1.41 (m, 2H), 1.29 - 1.16 (m, 2H) Step E. Procedure for preparation of 1-(((1s,4r)-4-(4-(benzyloxy)butyl)cyclohexyl)oxy)-3- bromobenzene To a solution of 1-(((1r,4r)-4-((E)-4-(benzyloxy)but-1-en-1-yl)cyclohexyl)oxy )-3- bromobenzene (1.68 g, 4.04 mmol, 1.0 equiv.) in EtOH (20 mL) was added PtO2 (183.69 mg, 808.93 μmol, 0.2 equiv.) under N 2 atmosphere. The suspension was degassed and purged with H2 three times. The mixture was stirred under H2 (15 Psi) at 25 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give 1-(((1s,4r)-4-(4- (benzyloxy)butyl)cyclohexyl)oxy)-3-bromobenzene (1.32 g, crude) as a yellow oil. Step F. Procedure for preparation of 4-((1r,4s)-4-(3-bromophenoxy)cyclohexyl)butan-1- ol To a solution of 1-(((1s,4r)-4-(4-(benzyloxy)butyl)cyclohexyl)oxy)-3-bromoben zene (1.32 g, 3.16 mmol, 1.0 equiv.) in DCM (15 mL) was added TMSI (1.90 g, 9.49 mmol, 1.29 mL, 3.0 equiv.) at 0 °C under N2. The mixture was stirred at 25 °C for 2 hours under N2 atmosphere. The reaction mixture was quenched by the addition of H 2 O (20 mL) and extracted with DCM (20 mL ×2). The combined organic layers were washed with H2O (20 mL ×1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 20% ethyl acetate / petroleum ether) to give 4- ((1r,4s)-4-(3-bromophenoxy)cyclohexyl)butan-1-ol (930 mg, 2.50 mmol, 78.98% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.21 (t, J = 8.0 Hz, 1H), 7.11 (d, J = 2.0 Hz, 1H), 7.07 (d, J = 8.0 Hz, 1H), 6.93 (dd, J = 1.6, 8.4 Hz, 1H), 4.39 - 4.23 (m, 2H), 3.43 - 3.35 (m, 2H), 2.02 (d, J = 9.6 Hz, 2H), 1.75 (d, J = 12.4 Hz, 2H), 1.44 - 1.36 (m, 2H), 1.33 - 1.26 (m, 4H), 1.21 - 1.15 (m, 3H), 1.11 - 0.97 (m, 2H) Step G. Procedure for preparation of 4-((1r,4s)-4-(3-bromophenoxy)cyclohexyl)butanal A solution of DMSO (381.99 mg, 4.89 mmol, 381.99 μL, 4.0 equiv.) in DCM (4 mL) was added dropwise to a solution of oxalyl dichloride (310.29 mg, 2.44 mmol, 213.99 μL, 2.0 equiv.) in DCM (0.5 mL) at -70 °C under N2 atmosphere. The mixture was stirred at -70 °C for 30 minutes. After which time, 4-((1r,4s)-4-(3-bromophenoxy)cyclohexyl)butan-1-ol (400.00 mg, 1.22 mmol, 1.0 equiv.) in DCM (0.5 mL) was added dropwise at -70 °C. The solution was stirred for 30 minutes at -70 °C. Then TEA (742.10 mg, 7.33 mmol, 1.02 mL, 6.0 equiv.) was added into the solution. The resulting reaction mixture was stirred at 25 °C for 1 hour under N2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with DCM (10 mL × 3). The combined organic layers were washed with brine (10 mL × 1), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give 4-((1r,4s)-4-(3- bromophenoxy)cyclohexyl)butanal (500 mg, crude) as a colorless oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 9.66 (t, J = 1.6 Hz, 1H), 7.21 (t, J = 8.0 Hz, 1H), 7.13 - 7.11 (m, 1H), 7.09 - 7.05 (m, 1H), 6.96 - 6.91 (m, 1H), 4.34 - 4.26 (m, 1H), 2.43 - 2.39 (m, 2H), 2.07 - 1.98 (m, 2H), 1.75 (d, J = 12.4 Hz, 2H), 1.58 - 1.49 (m, 2H), 1.37 - 1.27 (m, 2H), 1.26 - 1.19 (m, 3H), 1.10 - 0.99 (m, 2H) Step H. Procedure for preparation of 3-(6-(4-(4-((1r,4s)-4-(3- bromophenoxy)cyclohexyl)butyl)piperazin-1-yl)-1-methyl-1H-in dazol-3-yl)piperidine- 2,6-dione To a solution of 4-((1r,4s)-4-(3-bromophenoxy)cyclohexyl)butanal (250 mg, 628.77 μmol, 1.0 equiv.) in DCM (5 mL) was added 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl) piperidine-2, 6- dione (251.65 mg, 691.64 μmol, 1.1 equiv., HCl) at 25 °C. The mixture was stirred at 25 °C for 16 hours. NaBH(OAc)3 (399.78 mg, 1.89 mmol, 3 equiv.) was added to the mixture at 25 °C. The reaction mixture was stirred at 25 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , DCM : MeOH = 20 : 1) to give 3-(6-(4-(4-((1r,4s)-4-(3- bromophenoxy)cyclohexyl)butyl)piperazin-1-yl)-1-methyl-1H-in dazol-3-yl)piperidine-2,6- dione (237 mg, 323.88 μmol, 51.51% yield) as a yellow solid. MS (ESI) m/z: 636.3 [M+H] + . Step I. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy)phenyl)p icolinate A mixture of 3-(6-(4-(4-((1r,4s)-4-(3-bromophenoxy)cyclohexyl)butyl)piper azin-1-yl)-1- methyl-1H-indazol-3-yl)piperidine-2,6-dione (237.00 mg, 372.28 μmol, 1.0 equiv.), tert-butyl 6-[8-(1, 3-benzothiazol-2-ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-3-(4, 4, 5, 5- tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridine-2-carboxylate (250.84 mg, 409.51 μmol, 1.1 equiv.), KF (64.88 mg, 1.12 mmol, 26.16 μL, 3.0 equiv.), and [2-(2-aminophenyl) phenyl]palladium (1 + ) ;bis (1-adamantyl) -butyl-phosphane;methanesulfonate (54.22 mg, 74.46 μmol, 0.2 equiv.) in dioxane (3 mL) and H 2 O (0.3 mL) was degassed and purged with N 2 three times. The mixture was stirred at 100 °C for 16 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 6% DCM / MeOH) to give tert-butyl 6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2( 1H)-yl)-3-(3-(((1s,4r)-4-(4-(4- (3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pipera zin-1- yl)butyl)cyclohexyl)oxy)phenyl)picolinate (300 mg, 245.80 μmol, 66.03% yield) as a yellow solid. MS (ESI) m/z: 1042.6 [M+H] + . Step J. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy)phenyl)p icolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)phenyl)picolinate (300 mg, 287.83 μmol, 1.0 equiv.) in DCM (1.5 mL) and TFA (1.5 mL) was stirred at 25 °C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep–HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-6-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)phenyl)picolinic acid (58.65 mg, 56.03 μmol, 19.47% yield) as a white solid. MS (ESI) m/z: 986.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.85 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.64 (dd, J = 8.0, 12.0 Hz, 2H), 7.52 - 7.41 (m, 3H), 7.39 - 7.32 (m, 2H), 7.25 (t, J = 8.0 Hz, 1H), 6.93 (dd, J = 9.2, 16.0 Hz, 2H), 6.89 - 6.82 (m, 4H), 4.97 (s, 2H), 4.30 - 4.14 (m, 2H), 3.93 - 3.87 (m, 5H), 3.24 (s, 2H), 3.00 (t, J = 5.6 Hz, 2H), 2.69 - 2.57 (m, 7H), 2.42 (s, 2H), 2.35 - 2.25 (m, 1H), 2.20 - 2.12 (m, 1H), 2.06 (d, J = 9.2 Hz, 2H), 1.76 (d, J = 12.4 Hz, 2H), 1.53 - 1.44 (m, 2H), 1.36 - 1.17 (m, 8H), 1.09 - 0.97 (m, 2H) Example 261. Preparation of Compound 330a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1- yl)propyl)cyclohexyl)oxy)phenyl)picolinic acid Step A. Procedure for preparation of ethyl (E)-3-((1r,4r)-4-(3- bromophenoxy)cyclohexyl)acrylate A mixture of ethyl 2-diethoxyphosphorylacetate (1.19 g, 5.30 mmol, 1.05 mL, 1.0 equiv.) in THF (5 mL) was degassed and purged with N2 three times. To this mixture was slowly added LiHMDS (1 M, 6.89 mL, 1.3 equiv.). The reaction mixture was stirred at -78 °C for 1.5 hours under a N 2 atmosphere. Then (1r,4r)-4-(3-bromophenoxy)cyclohexane-1-carbaldehyde (1.5 g, 5.30 mmol, 1.0 equiv.) in THF (15 mL) was added. The resulting mixture was slowly warm to -40°C and stirred for 1.5 hours under N 2 atmosphere. The reaction mixture was quenched by the addition of saturated NH 4 Cl (15mL) and extracted with ethyl acetate (15 mL × 2). The combined organic layers were washed with brine (15 mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 15% ethyl acetate / petroleum ether) to give ethyl (E)-3-((1r,4r)-4-(3- bromophenoxy)cyclohexyl)acrylate (850 mg, 2.41 mmol, 45.42% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.25 - 7.19 (m, 1H), 7.14 (t, J = 2.0 Hz, 1H), 7.11 - 7.07 (m, 1H), 6.95 (dd, J = 1.6, 8.4 Hz, 1H), 6.84 (dd, J = 6.8, 16.0 Hz, 1H), 5.83 (dd, J = 1.2, 16.0 Hz, 1H), 4.39 - 4.26 (m, 1H), 4.16 - 4.06 (m, 2H), 2.27 - 2.17 (m, 1H), 2.07 (d, J = 10.4 Hz, 2H), 1.81 (d, J = 11.6 Hz, 2H), 1.44 - 1.30 (m, 4H), 1.21 (t, J = 7.2 Hz, 3H) Step B. Procedure for preparation of ethyl 3-((1r,4r)-4-(3- bromophenoxy)cyclohexyl)propanoate To a solution of ethyl (E)-3-((1r,4r)-4-(3-bromophenoxy)cyclohexyl)acrylate (750 mg, 2.12 mmol, 1.0 equiv.) in EtOH (5 mL) was added PtO 2 (48.21 mg, 212.31 μmol, 0.1 equiv.) under N2. The suspension was degassed under vacuum and purged with H2 several times. The mixture was stirred under H2 (15 psi) at 25°C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give ethyl 3-((1r,4r)-4-(3- bromophenoxy)cyclohexyl)propanoate (750 mg, crude) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.28 - 7.16 (m, 1H), 7.12 (t, J = 2.0 Hz, 1H), 7.08 (d, J = 8.0 Hz, 1H), 6.96 - 6.85 (m, 1H), 4.36 - 4.20 (m, 1H), 4.05 (q, J = 7.2 Hz, 2H), 2.30 (t, J = 7.6 Hz, 2H), 2.02 (d, J = 9.6 Hz, 2H), 1.75 (d, J = 12.4 Hz, 2H), 1.46 (q, J = 7.2 Hz, 2H), 1.36 - 1.22 (m, 3H), 1.20 - 1.14 (m, 3H), 1.12 - 1.02 (m, 2H) Step C. Procedure for preparation of 3-((1r,4s)-4-(3-bromophenoxy)cyclohexyl)propan- 1-ol To a solution of ethyl 3-((1r,4r)-4-(3-bromophenoxy)cyclohexyl)propanoate (750 mg, 2.11 mmol, 1.0 equiv.) in THF (8 mL) was added LiAlH 4 (80.13 mg, 2.11 mmol, 1.0 equiv.). The mixture was stirred at 0 °C for 1 hour. The reaction mixture was quenched with Na 2 SO 4 •10 H2O (200 mg) and slowly warmed to 20 °C. The mixture was filtered and washed with DCM (250 mL). The filtrate was concentrated under reduced pressure to give 3-((1r,4s)-4-(3- bromophenoxy)cyclohexyl)propan-1-ol (700 mg, crude) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.28 - 7.19 (m, 1H), 7.12 (d, J = 2.0 Hz, 1H), 7.08 (d, J = 8.0 Hz, 1H), 6.97 - 6.87 (m, 1H), 4.36 (t, J = 5.2 Hz, 1H), 4.33 - 4.25 (m, 1H), 3.37 (q, J = 6.4 Hz, 2H), 2.03 (d, J = 9.6 Hz, 2H), 1.76 (d, J = 12.0 Hz, 2H), 1.43 (d, J = 7.2 Hz, 2H), 1.37 - 1.27 (m, 2H), 1.25 - 1.15 (m, 3H), 1.11 - 0.99 (m, 2H) Step D. Procedure for preparation of 3-((1r,4r)-4-(3-bromophenoxy)cyclohexyl)propanal A solution of oxalyl dichloride (567.30 mg, 4.47 mmol, 391.24 μL, 2.0 equiv.) in DCM (10 mL) was added dropwise to a solution of DMSO (698.45 mg, 8.94 mmol, 698.45 μL, 4.0 equiv.) in DCM (5 mL) at -78 °C under N2 atmosphere. The mixture was stirred at -78°C for 90 minutes. After which time 3-((1r,4s)-4-(3-bromophenoxy)cyclohexyl)propan-1-ol (700 mg, 2.23 mmol, 1.0 equiv.) in DCM (2 mL) was added dropwise at -78 °C. The solution was stirred for 30 minutes at -78 °C. Then TEA (1.36 g, 13.41 mmol, 1.87 mL, 6 equiv.) was added into the solution. The resulting solution was stirred at 25 °C for 1 hour under a N 2 atmosphere. The reaction mixture was quenched with H2O (5mL) at 25°C and extracted with DCM (5mL × 3). The combined organic layers were dried over Na2SO4, filtered, and concentrated under reduced pressure to give 3-((1r,4r)-4-(3-bromophenoxy)cyclohexyl)propanal (600 mg, crude) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 9.68 (s, 1H), 7.28 - 7.17 (m, 1H), 7.12 (d, J = 2.0 Hz, 1H), 7.08 (d, J = 8.0 Hz, 1H), 6.96 - 6.85 (m, 1H), 2.46 - 2.43 (m, 2H), 2.03 (d, J = 12.0 Hz, 2H), 1.75 (d, J = 12.4 Hz, 2H), 1.52 - 1.39 (m, 2H), 1.32 - 1.16 (m, 4H), 1.12 - 0.99 (m, 2H) Step E. Procedure for preparation of 3-(6-(4-(3-((1r,4s)-4-(3- bromophenoxy)cyclohexyl)propyl)piperazin-1-yl)-1-methyl-1H-i ndazol-3-yl)piperidine- 2,6-dione A solution of 3-(1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidine-2,6-dio ne (210.39 mg, 642.65 μmol, 1.0 equiv.) and 3-((1r,4r)-4-(3-bromophenoxy)cyclohexyl)propanal (200 mg, 642.65 μmol, 1.0 equiv.) in DCM (3 mL) was stirred at 25 °C for 0.5 hours. Then NaBH(OAc) 3 (408.61 mg, 1.93 mmol, 3 equiv.) was added. The mixture was stirred at 25 °C for 1.5 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , DCM: MeOH = 10:1) to give 3-(6-(4-(3-((1r,4s)-4-(3- bromophenoxy)cyclohexyl)propyl)piperazin-1-yl)-1-methyl-1H-i ndazol-3-yl)piperidine-2,6- dione (190 mg, 248.60 μmol, 38.68% yield) as a yellow oil. MS (ESI) m/z: 622.4 [M+H] + . Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1-yl)propyl)cyclohexyl)oxy)phenyl) picolinate To a solution of 3-(6-(4-(3-((1r,4s)-4-(3-bromophenoxy)cyclohexyl)propyl)pipe razin-1-yl)-1- methyl-1H-indazol-3-yl)piperidine-2,6-dione (190 mg, 305.17 μmol, 1.0 equiv.) and tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-2-carboxylate (224.32 mg, 366.21 μmol, 1.2 equiv.) in dioxane (3 mL) and H2O (0.3 mL) was added [2-(2- aminophenyl)phenyl]palladium(1 + );bis(1-adamantyl)-butyl-phosphane;methanesulfonate (22.22 mg, 30.52 μmol, 0.1 equiv.) and KF (53.19 mg, 915.52 μmol, 21.45 μL, 3.0 equiv.). The mixture was stirred at 100 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 6% dichloromethane/methanol) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-( 2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)propyl)cyclohexyl)oxy) phenyl)picolinate (210 mg, 180.72 μmol, 59.22% yield) as a yellow oil. MS (ESI) m/z: 515.1 [M/2+H] + . Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1-yl)propyl)cyclohexyl)oxy)phenyl) picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-6- yl)piperazin-1-yl)propyl)cyclohexyl)oxy)phenyl)picolinate (210 mg, 204.23 μmol, 1.0 equiv.) in DCM (1 mL) was added TFA (2 mL). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep–HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-6- yl)piperazin-1-yl)propyl)cyclohexyl)oxy)phenyl)picolinic acid (86.61 mg, 85.10 μmol, 41.67% yield) as a yellow solid. MS (ESI) m/z: 972.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.36 - 12.34 (m, 1H), 10.85 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.68 - 7.60 (m, 2H), 7.54 - 7.45 (m, 2H), 7.43 (d, J = 8.4 Hz, 1H), 7.35 (q, J = 7.2 Hz, 2H), 7.25 (t, J = 7.6 Hz, 1H), 6.93 (dd, J = 9.2, 14.0 Hz, 2H), 6.89 - 6.82 (m, 4H), 4.97 (s, 2H), 4.29 - 4.18 (m, 2H), 3.90 - 3.88 (m, 5H), 3.26 - 2.24 (m, 2H), 3.02 - 3.01 (m, 2H), 2.69 - 2.61 (m, 5H), 2.55 - 2.53 (m, 2H), 2.45 - 2.43 (m, 2H), 2.34 - 2.25 (m, 1H), 2.17 - 2.15 (m, 1H), 2.07 - 2.05 (m, 2H), 1.79 - 1.77 (m, 2H), 1.53 - 1.51 (m, 2H), 1.39 - 1.13 (m, 6H), 1.11 - 0.98 (m, 2H) Example 262. Preparation of Compound 213 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(4-(1-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)amino)-2- oxoethyl)piperidin-4- yl)butoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of methyl 2-(4-(4-(2-methyl-3-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)phenoxy)butyl)piperidin-1-yl)acetate A solution of tert-butyl 4-(3-(4-bromo-3-methylphenoxy)propyl)piperidine-1-carboxylat e (2.2 g, 1 equiv., 5.3 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (2.0 g, 1.5 equiv, 8.0 mmol), potassium acetate (1.6 g, 3 equiv., 16 mmol), and PdCl2(dppf)-CH 2 Cl 2 adduct (0.22 g, 0.05 equiv., 0.26 mmol) in 1,4-dioxane (15 mL) was purged with nitrogen gas and heated to 85 ºC. After 16 h the reaction solution was concentrated, and the resulting crude material was purified by silica gel column chromatography (0-100% ethyl acetate in heptane) to provide methyl 2-(4-(4-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2- yl)phenoxy)butyl)piperidin-1-yl)acetate (2.6 g, 5.7 mmol) as a colorless oil. Step B. Procedure for preparation of tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(1-(2-methoxy-2-oxoethy l)piperidin-4-yl)butoxy)- 2-methylphenyl)picolinate A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-bromopicolinate (500 mg, 1 equiv., 884 µmol), methyl 2-(4-(4-(2-methyl-3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)butyl)piperidin- 1-yl)acetate (512 mg, 1.3 equiv., 1.15 mmol), and CataCXium PD G3 (64.4 mg, 0.1 equiv., 88.4 µmol) in 1,4-dioxane (7.5 mL) and K3PO4 (1.5 M, 2.5 mL) was heated in the microwave at 100 ºC. After 40 minutes, the reaction vessel was removed from microwave and heated to 80 ºC by conventional heating. After 12 hours, the reaction solution was filtered and concentrated to provide crude tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(4-(1-(2- methoxy-2-oxoethyl)piperidin-4-yl)butoxy)-2-methylphenyl)pic olinate, which was carried forward without further purification. Step C. Procedure for preparation of 2-(4-(4-(3-(6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyri din-3-yl)-2- methylphenoxy)butyl)piperidin-1-yl)acetic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(4-(1-(2-methoxy-2-oxoethyl)piperidin-4-yl)bu toxy)-2- methylphenyl)picolinate (711 mg, 884 µmol, 1 equiv.) in methanol (5 mL) was added an aqueous solution of NaOH (1 M, 1 mL). The reaction solution was stirred at 60 ºC. After 3 hours, the reaction solution was concentrated and purified by silica gel column chromatography to provide 2-(4-(4-(3-(6-(8-(Benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin-2(1H)- yl)-2-(tert-butoxycarbonyl)pyridine-3-yl)-2-methylphenoxy)bu tyl)piperidin-1-yl)acetic acid (278 mg, 352 µmol, 40% yield over two steps). MS (ESI) m/z: 791.2 [M+H] + . Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(1-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-7-yl)amino)-2-oxoethyl)piperidin-4-yl)butoxy)-2-meth ylphenyl)picolinate A solution of 2-(4-(4-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridine-3-yl)-2-methylphen oxy)butyl)piperidin-1-yl)acetic acid (60 mg, 1 equiv., 76 µmol), 3-(7-amino-1-methyl-1H-indazol-3-yl)piperidine-2,6-dione (24 mg, 1.2 equiv., 91 µmol), N,N,N’,N’-Tetramethylchloroformamidinium- hexafluorophosphate (26 mg, 1.2 equiv., 91 µmol), and 1-methylimidazole (22 mg, 21 µL, 3.5 equiv., 0.27 mmol) in MeCN (5 mL) was stirred at room temperature. After 16 h the reaction solution was filtered and concentrated to provide crude tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(1-(2- ((3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)amino)-2-oxoethyl)piperidin-4-yl)buto xy)-2- methylphenyl)picolinate, which was carried forward without further purification. MS (ESI) m/z: 516.0 [M+H/2] + . Step E. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(1-(2-((3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-7-yl)amino)-2-oxoethyl)piperidin-4-yl)butoxy)-2-meth ylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(4-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-meth yl-1H-indazol-7-yl)amino)-2- oxoethyl)piperidin-4-yl)butoxy)-2-methylphenyl)picolinate (78 mg, 76 µmol, 1 equiv.) in DCM (4 mL) was added TFA (1 mL). The reaction solution was stirred at room temperature. After 16 hours, the reaction solution was concentrated. The resulting residue was re-suspended in DMSO and purified by reverse-phase HPLC to provide 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(4-(1-(2- ((3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)amino)-2-oxoethyl)piperidin-4-yl)buto xy)-2-methylphenyl)picolinic acid (17 mg, 17 µmol, 23 %yield) as a white solid. MS (ESI) m/z: 974 [M+H] + . consistent with structure. Example 263. Preparation of Compound 331a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3- ((3R)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)pyrrolidin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1S,4r)-4-(3- ((3R*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)pyrrolidin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of tert-butyl (3R)-3-[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]pyrrolidine-1-carboxylate To a solution of tert-butyl (3R)-3-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6- yl]pyrrolidine-1-carboxylate (4.2 g, 7.11 mmol, 1 equiv.) in EtOH (20 mL) and THF (20 mL) was added Pd/C (2.10 g, 1.97 mmol, 10% purity, 0.278 equiv.), Pd(OH)2 (2.10 g, 14.95 mmol, 2.10 equiv.) and AcOH (426.96 mg, 7.11 mmol, 407.02 μL, 1 equiv.) under N2 atmosphere. The suspension was degassed and purged with H 2 three times. The mixture was stirred under H2 (15 Psi) at 25 °C for 16 hours. The reaction mixture was filtered, and the filter cake was washed with THF (200 mL).The filtrate was concentrated to afford tert-butyl (3R)-3-[3-(2,6- dioxo-3-piperidyl)-1-methyl-indazol-6-yl]pyrrolidine-1-carbo xylate (3.05 g, crude) as a brown solid, which was used in the next step without further purification. Step B. Procedure for preparation of 3-[1-methyl-6-[(3R)-pyrrolidin-3-yl]indazol-3- yl]piperidine-2,6-dione To a solution of tert-butyl (3R)-3-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6- yl]pyrrolidine-1-carboxylate (3.0 g, 7.27 mmol, 1 equiv.) in dioxane (10 mL) was added HCl/dioxane (4 M, 21.05 mL, 11.58 equiv.).The mixture was stirred at 25 °C for 16 hours. The reaction mixture was filtered to afford 3-[1-methyl-6-[(3R)-pyrrolidin-3-yl]indazol-3- yl]piperidine-2,6-dione (1345.01 mg, 7.26 mmol, 99.80% yield, HCl) as a yellow solid. MS (ESI) m/z: 313.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.89 (s, 1H), 9.88 - 9.57 (m, 2H), 7.68 (d, J = 8.4 Hz, 1H), 7.63 (s, 1H), 7.13 (dd, J = 1.2, 8.4 Hz, 1H), 4.36 (dd, J = 5.2, 9.6 Hz, 1H), 4.00 (s, 3H), 3.67 - 3.58 (m, 2H), 3.44 - 3.37 (m, 1H), 3.29 - 3.15 (m, 2H), 2.73 - 2.55 (m, 2H), 2.43 - 2.32 (m, 2H), 2.17 (dd, J = 5.2, 13.6 Hz, 1H), 2.08 - 1.98 (m, 1H). Step C. Procedure for preparation of 3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propanal A mixture of 3-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propan-1-ol (500 mg, 1.53 mmol, 1.0 equiv.) and DMP (972.04 mg, 2.29 mmol, 710.04 μL, 1.5 equiv.) in DCM (10 mL) was degassed and purged with N2 three times. The mixture was stirred at 35 °C for 3 hours under N 2 atmosphere. The mixture was filtered and washed with DCM (20 mL). The filtrate was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0-10% ethyl acetate/petroleum ether) to give 3-((1r,4r)-4-(3- bromo-2-methylphenoxy)cyclohexyl)propanal (530 mg, 1.63 mmol, 53.33% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ = 9.68 (s, 1H), 7.17 - 7.11 (m, 1H), 7.10 - 6.98 (m, 2H), 4.22 (m, 1H), 2.45 (m, 2H), 2.20 (s, 3H), 2.04 (m , 2H), 1.75 (m, 2H), 1.45 (q, J = 7.2 Hz, 2H), 1.36 - 1.22 (m, 3H), 1.04 (q, J = 12.0 Hz, 2H). Step D. Procedure for preparation of 3-(6-((R)-1-(3-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propyl)pyrrolidin-3-yl)-1-methyl-1H -indazol-3- yl)piperidine-2,6-dione To a solution of 3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propanal (100 mg, 307.47 μmol, 1.0 equiv.), 3-(1-methyl-6-((R)-pyrrolidin-3-yl)-1H-indazol-3-yl)piperidi ne-2,6- dione (115.25 mg, 368.96 μmol, 1.2 equiv.), and NaOAc (25.22 mg, 307.47 μmol, 1.0 equiv.) in DCM (2 mL) was added NaBH(OAc) 3 (325.82 mg, 1.54 mmol, 5.0 equiv.). The resulting mixture was stirred at 25 °C for 1 hour. After completion, the reaction mixture was poured into water (10 mL), and then extracted with ethyl acetate (10 mL × 3). The combined organic layers were washed with brine (10 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give 3-(6-((R)-1-(3-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propyl)pyrrolidin-3-yl)-1-methyl-1H -indazol-3-yl)piperidine-2,6- dione (200 mg, crude) as a yellow oil. MS (ESI) m/z: 621.4 [M+H] + . Step E. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-((3R)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)pyrrolidin-1-yl)propyl)cyclohexyl)oxy )-2- methylphenyl)picolinate A mixture of 3-(6-((R)-1-(3-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propyl)pyrrolidin-3-yl)-1-methyl-1H -indazol-3-yl)piperidine-2,6- dione (200 mg, 248.71 μmol, 1.0 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)picolinate (182.82 mg, 298.45 μmol, 1.2 equiv.), Ad 2 nBuP Pd G 3 (cataCXium® A Pd G 3 ) (36.23 mg, 49.74 μmol, 0.2 equiv.), and KF (72.25 mg, 1.24 mmol, 29.13 μL, 5.0 equiv.) in dioxane (6 mL) and H2O (0.6 mL) was degassed and purged with N2 three times. The mixture was stirred at 100 °C for 3 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , petroleum ether : ethyl acetate = 0:1 ). The product thus obtained was purified by prep-TLC (SiO2, DCM: MeOH = 10:1) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-((3R)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)pyrrolidin-1-yl)propyl)cyclohexyl)oxy )-2-methylphenyl)picolinate (120 mg, 93.24 μmol, 37.49% yield) as a yellow oil. MS (ESI) m/z: 1027.6 [M+H] + . Step F. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-((3R)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)pyrrolidin-1-yl)propyl)cyclohexyl)oxy )-2- methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-(3-((3R*)-3-(3 -(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)pyrrolidin-1-yl)propyl)cyclohexyl)oxy )-2- methylphenyl)picolinic acid] A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1s,4r)-4-(3-((3R)-3-(3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-6- yl)pyrrolidin-1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl)pic olinate (120 mg, 116.81 μmol, 1.0 equiv.) in TFA (1.5 mL) and DCM (0.5 mL) was stirred at 35 °C for 1.5 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1s,4r)-4-(3-((3R)-3-(3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-6- yl)pyrrolidin-1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl)pic olinic acid [6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1S,4r)-4-(3- ((3R*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)pyrrolidin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] (65.65 mg, 65.30 μmol, 55.90% yield) as a yellow solid. MS (ESI) m/z: 971.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.96 - 12.77 (m, 1H), 12.67 - 12.45 (m, 1H), 10.93 (d, J = 1.2 Hz, 1H), 8.08 - 7.98 (m, 1H), 7.79 (m, 1H), 7.71 (d, J = 8.8 Hz, 1H), 7.64 - 7.54 (m, 2H), 7.49 - 7.42 (m, 3H), 7.40 - 7.32 (m, 2H), 7.17 - 7.05 (m, 2H), 6.95 (m, 2H), 6.62 (d, J = 7.6 Hz, 1H), 5.05 - 4.91 (m, 2H), 4.39 - 4.31 (m, 1H), 4.26 - 4.16 (m, 1H), 4.00 (s, 3H), 3.92 (m, 2H), 3.62 - 3.54 (m, 1H), 3.27 - 3.17 (m, 4H), 3.03 (m, 2H), 2.61 (m, 4H), 2.23 - 2.15 (m, 2H), 2.13 - 2.07 (m, 2H), 1.87 (s, 3H), 1.85 - 1.79 (m, 2H), 1.74 - 1.65 (m, 2H), 1.42 - 1.23 (m, 6H), 1.17 - 1.01 (m, 3H). Example 264. Preparation of Compound 331b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(3- ((3S)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)pyrrolidin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1R,4r)-4-(3- ((3S*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)pyrrolidin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of tert-butyl 3-[3-(2,6-dibenzyloxy-3-pyridyl)-1- methyl-indazol-6-yl]pyrrolidine-1-carboxylate A solution of 6-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (15 g, 29.98 mmol, 1 equiv.), tert-butyl 3-bromopyrrolidine-1-carboxylate (9.75 g, 38.97 mmol, 1.3 equiv.), Ir[dF(CF 3 )ppy] 2 (dtbpy)(PF 6 ) (336.31 mg, 299.77 μmol, 0.01 equiv.), NiCl 2 .dtbbpy (178.96 mg, 449.65 μmol, 0.015 equiv.), TTMSS (7.45 g, 29.98 mmol, 9.25 mL, 1 equiv.), and Na2CO3 (6.35 g, 59.95 mmol, 2 equiv.) in DME (2 mL) was sealed and placed under nitrogen. The reaction was stirred and irradiated with a 10 W blue LED lamp (3 cm away) with cooling water to keep the reaction temperature at 25 °C for 14 hours. The reaction mixture was filtered, diluted with H2O (500 mL), and extracted with ethyl acetate (200 mL × 3). The combined organic layers were washed with brine (300 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~35% ethyl acetate/petroleum ether) to afford tert-butyl 3-[3-(2,6- dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-yl]pyrrolidine-1-c arboxylate (10.5 g, 17.06 mmol, 56.92% yield) as a yellow oil. MS (ESI) m/z: 591.3 [M+H] + . Step B. Procedure for preparation of tert-butyl (3R)-3-[3-(2,6-dibenzyloxy-3-pyridyl)-1- methyl-indazol-6-yl]pyrrolidine-1-carboxylate and tert-butyl (3S)-3-[3-(2,6-dibenzyloxy- 3-pyridyl)-1-methyl-indazol-6-yl]pyrrolidine-1-carboxylate The stereoisomers of tert-butyl 3-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6- yl]pyrrolidine-1-carboxylate were separated by SFC to afford tert-butyl (3S)-3-[3-(2,6- dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-yl]pyrrolidine-1-c arboxylate (3.5 g, 5.75 mmol, 32.33% yield) was obtained as a yellow oil and tert-butyl (3R)-3-[3-(2,6-dibenzyloxy-3- pyridyl)-1-methyl-indazol-6-yl]pyrrolidine-1-carboxylate (4.2 g, 6.97 mmol, 39.20% yield) as a yellow oil. MS (ESI) m/z: 591.3 [M+H] + . MS (ESI) m/z: 591.3 [M+H] + . Step C. Procedure for preparation of tert-butyl (3S)-3-[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]pyrrolidine-1-carboxylate To a solution of tert-butyl (3S)-3-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6- yl]pyrrolidine-1-carboxylate (3.5 g, 5.93 mmol, 1 equiv.) in EtOH (20 mL) and THF (20 mL) was added Pd/C (2 g, 1.88 mmol, 10% purity, 0.317 equiv.), Pd(OH)2 (2 g, 14.24 mmol, 2.40 equiv.), and AcOH (355.81 mg, 5.93 mmol, 339.19 μL, 1 equiv.) under N 2 atmosphere. The suspension was degassed and purged with H 2 three times. The mixture was stirred under H 2 (15 Psi) at 25 °C for 16 hours. The reaction mixture was filtered, and the filter cake was washed with THF (200 mL). The filtrate was concentrated under vacuum to afford tert-butyl (3S)-3- [3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]pyrrolidine -1-carboxylate (2.7 g, crude) as a brown solid, which was used in the next step without further purification. Step D. Procedure for preparation of 3-[1-methyl-6-[(3S)-pyrrolidin-3-yl]indazol-3- yl]piperidine-2,6-dione To a solution of tert-butyl (3S)-3-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6- yl]pyrrolidine-1-carboxylate (2.65 g, 6.42 mmol, 1 eq ) in dioxane (10 mL) was added HCl/dioxane (4 M, 20.78 mL, 12.94 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was filtered to afford 3-[1-methyl-6-[(3S)-pyrrolidin-3-yl]indazol-3- yl]piperidine-2,6-dione (1287.35 mg, 6.30 mmol, 98.13% yield, 2HCl) as a pink solid. MS (ESI) m/z: 313.3 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.89 (s, 1H), 9.84 - 9.64 (m, 2H), 7.68 (d, J = 8.4 Hz, 1H), 7.62 (s, 1H), 7.12 (d, J = 8.4 Hz, 1H), 4.36 (dd, J = 5.2, 9.6 Hz, 1H), 3.99 (s, 3H), 3.67 - 3.58 (m, 2H), 3.44 - 3.37 (m, 1H), 3.28 - 3.14 (m, 2H), 2.72 - 2.55 (m, 2H), 2.43 - 2.31 (m, 2H), 2.20 - 2.12 (m, 1H), 2.07 - 1.97 (m, 1H). Step E. Procedure for preparation of 3-(6-((S)-1-(3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propyl)pyrrolidin-3-yl)-1-methyl-1H -indazol-3- yl)piperidine-2,6-dione To a solution of 3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propanal (100 mg, 307.47 μmol, 1.0 equiv.) and 3-[1-methyl-6-[(3S)-pyrrolidin-3-yl]indazol-3-yl]piperidine- 2,6- dione (105.65 mg, 338.21 μmol, 1.1 equiv.) in DCM (5 mL) was added NaOAc (50.45 mg, 614.93 μmol, 2.0 equiv.) at 25 °C for 1 hour. Then sodium triacetoxyborohydride (325.82 mg, 1.54 mmol, 5.0 equiv.) was added to the mixture which was stirred at 25 °C for 1 hour. The reaction mixture was partitioned between DCM (15 mL) and water (8 mL). The organic phase was separated, dried over Na2SO4, filtered, and concentrated under reduced pressure to give 3- (6-((S)-1-(3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl) propyl)pyrrolidin-3-yl)-1- methyl-1H-indazol-3-yl)piperidine-2,6-dione (130 mg, crude) as a yellow oil. MS (ESI) m/z: 621.2 [M+H] + . Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-((3S)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)pyrrolidin-1-yl)propyl)cyclohexyl)oxy )-2- methylphenyl)picolinate A mixture of 3-(6-((S)-1-(3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propyl)pyrrolidin-3-yl)-1-methyl-1H -indazol-3-yl)piperidine-2,6- dione (130 mg, 209.14 μmol, 1.0 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3, 2-dioxaborolan-2-yl)pyridine-2- carboxylate (140.92 mg, 230.05 μmol, 1.1 equiv.), Ad2nBuP Pd G3 (30.46 mg, 41.83 μmol, 0.2 equiv.), and KF (60.75 mg, 1.05 mmol, 24.50 μL, 5.0 equiv.) in dioxane (6 mL) and H 2 O (0.6 mL) was degassed and purged with N 2 three times. The mixture was stirred at 80 °C for 12 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH = 12:1) to give tert- butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)- 4-(3-((3S)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazo l-6-yl)pyrrolidin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (100 mg, 70.09 μmol, 33.51% yield) as a yellow solid. MS (ESI) m/z: 514.5 [M/2+H] + . Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-((3S)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)pyrrolidin-1-yl)propyl)cyclohexyl)oxy )-2- methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-(3-((3S*)-3-(3 -(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)pyrrolidin-1-yl)propyl)cyclohexyl)oxy )-2- methylphenyl)picolinic acid] A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1R,4r)-4-(3-((3S)-3-(3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-6- yl)pyrrolidin-1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl)pic olinate (100 mg, 97.34 μmol, 1.0 equiv.) in TFA (3 mL) and DCM (1 mL) was stirred at 40 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by reverse-phase HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(3-((3S)-3-(3-(2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-6- yl)pyrrolidin-1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl)pic olinic acid [6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1R,4r)-4-(3- ((3S*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)pyrrolidin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] (37.80 mg, 37.28 μmol, 38.30% yield) as a white solid. MS (ESI) m/z: 971.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.87 (s, 1H), 8.15 (s, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.66 - 7.60 (m, 2H), 7.48 - 7.41 (m, 4H), 7.37 (dd, J = 8.0, 10.0 Hz, 2H), 7.09 (s, 2H), 6.93 (dd, J = 8.8, 13.2 Hz, 2H), 6.62 (d, J = 8.0 Hz, 1H), 4.97 (s, 2H), 4.38 - 4.27 (m, 1H), 4.25 - 4.12 (m, 1H), 3.96 (s, 3H), 3.94 - 3.87 (m, 2H), 3.52 - 3.45 (m, 1H), 3.16 - 3.07 (m, 1H), 3.02 (t, J = 4.8 Hz, 2H), 2.93 - 2.80 (m, 2H), 2.61 (s, 3H), 2.36 - 2.27 (m, 4H), 2.22 - 2.12 (m, 2H), 2.07 (dd, J = 2.0, 8.4 Hz, 2H), 1.87 (s, 3H), 1.82 - 1.76 (m, 2H), 1.57 - 1.48 (m, 2H), 1.40 - 1.32 (m, 2H), 1.30 - 1.22 (m, 3H), 1.12 - 1.00 (m, 2H) Example 265. Preparation of Compound 332a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- ((3R)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)pyrrolidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1S,4r)-4-(4- ((3R*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)pyrrolidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of 3-(6-((R)-1-(4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butyl)pyrrolidin-3-yl)-1-methyl-1H- indazol-3-yl)piperidine- 2,6-dione To a solution of 4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butanal (100 mg, 294.75 μmol, 1.0 equiv.) in DCM (1 mL) was added NaOAc (36.27 mg, 442.13 μmol, 1.5 equiv.) and 3-(1-methyl-6-((R)-pyrrolidin-3-yl)-1H-indazol-3-yl)piperidi ne-2,6-dione (113.10 mg, 324.23 μmol, 1.1 equiv., HCl). The mixture was stirred at 25 °C for 0.5 hours. Then, NaBH(OAc) 3 (312.35 mg, 1.47 mmol, 5.0 equiv.) was added into the mixture, and the reaction was stirred at 25 °C for 1.5 hours. The reaction mixture was diluted with DCM (100 mL) and water (100 mL). The layers were separated. The organic layers were combined, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give 3-(6-((R)-1-(4- ((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butyl)pyrroli din-3-yl)-1-methyl-1H- indazol-3-yl)piperidine-2,6-dione (150 mg, 235.99 μmol, 80.06% yield) as a yellow oil. MS (ESI) m/z: 635.4 [M+H] + Step B. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-((3R)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)pyrrolidin-1-yl)butyl)cyclohexyl)oxy) -2-methylphenyl)picolinate A mixture of 3-(6-((R)-1-(4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butyl)pyrrolidin-3-yl)-1-methyl-1H- indazol-3-yl)piperidine-2,6- dione (150 mg, 235.99 μmol, 1.0 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)picolinate (187.92 mg, 306.78 μmol, 1.3 equiv.), KF (68.55 mg, 1.18 mmol, 27.64 μL, 5 equiv.), H 2 O (0.2 mL), and [2-(2-aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-buty l- phosphane;methanesulfonate (34.37 mg, 47.20 μmol, 0.2 equiv.) in dioxane (2 mL) was degassed and purged with N2 three times. The mixture was stirred at 100 °C for 3 hours under a N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH = 15:1) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- ((3R)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)pyrrolidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (100 mg, 76.83 μmol, 32.56% yield) as a yellow solid. MS (ESI) m/z: 1041.7 [M+H] + Step C. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-((3R)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)pyrrolidin-1-yl)butyl)cyclohexyl)oxy) -2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3- (((1S,4r)-4-(4-((3R*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-6-yl)pyrrolidin- 1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4-((3R)-3-(3-(2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-6- yl)pyrrolidin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)pico linate (100 mg, 76.83 μmol, 1.0 equiv.) in DCM (1 mL) was added TFA (4.61 g, 40.39 mmol, 3 mL, 525.68 equiv.). The mixture was stirred at 35 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r) -4-(4-((3R)-3-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pyrrolidin-1-y l)butyl)cyclohexyl)oxy)-2- methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin - 2(1H)-yl)-3-(3-(((1S,4r)-4-(4-((3R*)-3-(3-(2,6-dioxopiperidi n-3-yl)-1-methyl-1H-indazol-6- yl)pyrrolidin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)pico linic acid] (47.78 mg, 47.56 μmol, 61.91% yield) as a white solid. MS (ESI) m/z: 985.7 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 13.08 - 12.48 (m, 1H), 10.88 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.70 (d, J = 8.4 Hz, 1H), 7.62 ( d, J = 7.2 Hz, 1H), 7.55 (s, 1H), 7.51 - 7.42 (m, 3H), 7.40 - 7.32 (m, 2H), 7.13 ( d, J = 8.4 Hz, 1H), 7.10 - 7.04 (m, 1H), 6.99 - 6.89 (m, 2H), 6.61 (d, J = 8.0 Hz, 1H), 4.98 (s, 2H), 4.39 - 4.32 (m, 1H), 4.24 - 4.15 (m, 1H), 3.99 (s, 3H), 3.91 ( t, J = 5.6 Hz, 2H), 3.80 - 3.57 (m, 2H), 3.23 - 3.10 (m, 4H), 3.02 ( t, J = 5.6 Hz, 2H), 2.71 - 2.65 (m, 1H), 2.64 - 2.60 (m, 1H), 2.35 - 2.30 (m, 1H), 2.23 - 2.01 (m, 5H), 1.87 (s, 3H), 1.79 - 1.77 (m, 2H), 1.64 ( s, 2H), 1.41 - 1.32 (m, 4H), 1.25 - 1.23 ( m, 4H), 1.12 - 1.00 (m, 2H) Example 266. Preparation of Compound 332b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(4- ((3S)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)pyrrolidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1R,4r)-4-(4- ((3S*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)pyrrolidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of 3-(6-((S)-1-(4-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butyl)pyrrolidin-3-yl)-1-methyl-1H- indazol-3-yl)piperidine- 2,6-dione To a solution of 4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butanal (100 mg, 294.75 μmol, 1.0 equiv.) in DCM (2 mL) was added 3-(1-methyl-6-((S)-pyrrolidin-3-yl)-1H-indazol- 3-yl)piperidine-2,6-dione (101.28 mg, 324.23 μmol, 1.1 equiv.), NaOAc (36.27 mg, 442.13 μmol, 1.5 equiv.), and NaBH(OAc) 3 (312.35 mg, 1.47 mmol, 5.0 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was diluted with H2O (20 mL) and extracted with DCM (30 mL × 2). The combined organic layers were washed with DCM (20 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give 3-(6-((S)-1-(4-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butyl)pyrrolidin-3-yl)-1-methyl-1H- indazol-3-yl)piperidine-2,6- dione (150 mg, 235.99 μmol, 80.06 % yield) as a yellow solid. MS (ESI) m/z: 635.4 [M+H] + . Step B. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(4-((3S)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)pyrrolidin-1-yl)butyl)cyclohexyl)oxy) -2-methylphenyl)picolinate A mixture of 3-(6-((S)-1-(4-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butyl)pyrrolidin-3-yl)-1-methyl-1H- indazol-3-yl)piperidine-2,6- dione (150 mg, 235.99 μmol, 1.0 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)picolinate (173.46 mg, 283.18 μmol, 1.2 equiv.), KF (27.42 mg, 471.97 μmol, 11.06 μL, 2.0 equiv.), [2- (2-aminophenyl)phenyl]palladium(1 + );bis(1-adamantyl)-butyl-phosphane;methanesulfonate (34.37 mg, 47.20 μmol, 0.2 equiv.), and H2O (1 mL) in dioxane (2 mL) was degassed and purged with N2 three times. The mixture was stirred at 100 °C for 3 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, petroleum ether / ethyl acetate = 0 / 1 to DCM / MeOH = 15 / 1 ) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1r,4r)-4-(4-((3S)-3-(3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-6- yl)pyrrolidin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)pico linate (100 mg, 96.03 μmol, 40.69 % yield) as a yellow solid. MS (ESI) m/z: 1041.7 [M+H] + Step C. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(4-((3S)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)pyrrolidin-1-yl)butyl)cyclohexyl)oxy) -2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3- (((1R,4r)-4-(4-((3S*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-6-yl)pyrrolidin- 1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(4-((3S)-3-(3-(2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-6- yl)pyrrolidin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)pico linate (100 mg, 96.03 μmol, 1.0 equiv.) in TFA (2 mL) was added DCM (2 mL). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(4-((3S)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)pyrrolidin-1-yl)butyl)cyclohexyl)oxy) -2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1R,4r)-4-(4- ((3S*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)pyrrolidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] (34.50 mg, 32.95 μmol, 34.31 % yield) as a white solid. MS (ESI) m/z: 985.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.89 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.69 (d, J = 8.4 Hz, 1H), 7.63 (d, J = 7.6 Hz, 1H), 7.54 (s, 1H), 7.50 - 7.43 (m, 3H), 7.40 - 7.33 (m, 2H), 7.14 - 7.05 (m, 2H), 6.99 - 6.89 (m, 2H), 6.65 - 6.60 (m, 1H), 4.98 (s, 2H), 4.36 (dd, J = 5.2, 9.6 Hz, 1H), 4.25 - 4.15 (m, 1H), 3.99 (s, 3H), 3.92 (t, J = 5.6 Hz, 2H), 3.65 - 3.54 (m, 2H), 3.04 - 3.02 (m, 4H), 2.70 - 2.55 (m, 4H), 2.44 - 2.30 (m, 4H), 2.25 - 2.12 (m, 2H), 2.12 - 2.04 (m, 3H), 1.80 - 1.78 (m, 2H), 1.64 - 1.56 (m, 2H), 1.38 - 1.36 (m, 4H), 1.26 - 1.24 (m, 4H), 1.12 - 1.02 (m, 2H). Example 267. Preparation of Compound 333a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4- ((4S)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 6-yl)piperazin-1- yl)pentyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1R,4r)-4- ((4S*)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)piperazin-1- yl)pentyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of 5-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)pentan-2-ol To a solution of 4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butanal (2.5 g, 7.37 mmol, 1.0 equiv.) in THF (25 mL) was added MeMgBr (3 M, 4.91 mL, 2.0 equiv.) at -70°C. The mixture was stirred at 25°C for 12 hours at N2 atmosphere. The mixture was quenched with aq. NH 4 Cl (200 mL) and extracted with EtOAc (50 mL × 3 ). The combined organic phases were washed with brine (100 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~13% ethyl acetate/petroleum ether) to give 5-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)pentan-2-ol (2.0 g, 5.07 mmol, 68.75% yield) as a brown oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.13 (d, J = 8.0 Hz, 1H), 7.06 (t, J = 8.0 Hz, 1H), 7.01 (d, J = 8.0 Hz, 1H), 4.28 (d, J = 4.0 Hz, 1H), 4.26 - 4.17 (m, 1H), 3.61 - 3.51 (m, 1H), 2.20 (s, 3H), 2.04 (d, J = 10.4 Hz, 2H), 1.76 (d, J = 12.4 Hz, 2H), 1.38 - 1.23 (m, 7H), 1.20 - 1.12 (m, 2H), 1.03 (d, J = 6.0 Hz, 5H) Step B. Procedure for preparation of 5-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)pentan-2-one To a solution of 5-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)pentan-2-ol (2.3 g, 6.47 mmol, 1.0 equiv.) in DCM (20 mL) was added DMP (5.49 g, 12.95 mmol, 4.01 mL, 2.0 equiv.) at 0 °C. The mixture was stirred at 25°C for 12 hours at N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~13% ethyl acetate/petroleum ether) to give 5- ((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)pentan-2-one (2.0 g, 5.66 mmol, 87.45% yield) as a white oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.13 (d, J = 8.0 Hz, 1H), 7.07 (t, J = 8.0 Hz, 1H), 7.01 (d, J = 8.0 Hz, 1H), 4.30 - 4.17 (m, 1H), 2.40 (t, J = 7.2 Hz, 2H), 2.21 (s, 3H), 2.09 - 1.99 (m, 5H), 1.75 (d, J = 12.4 Hz, 2H), 1.47 (td, J = 7.2, 15.1 Hz, 2H), 1.41 - 1.28 (m, 2H), 1.24 (d, J = 5.6 Hz, 1H), 1.18 - 1.09 (m, 2H), 1.09 - 0.96 (m, 2H) Step C. Procedure for preparation of tert-butyl 4-(5-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)pentan-2-yl)piperazine-1-carboxylat e A mixture of 5-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)pentan-2-on e (500 mg, 1.42 mmol, 1.0 equiv.) and tert-butyl piperazine-1-carboxylate (289.95 mg, 1.56 mmol, 1.1 equiv.) in DCM (10 mL) was stirred for 2 hours. NaBH(OAc) 3 (1.50 g, 7.08 mmol, 5.0 equiv.) was added into the mixture, and then the mixture was stirred at 25 °C for 12 hours under N2 atmosphere. The reaction mixture was quenched with H2O (50 mL) and extracted with EtOAc (50 mL × 3). The combined organic layers were washed with brine (50 mL × 1), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~20% ethyl acetate/petroleum ether) to give tert- butyl 4-(5-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)pentan-2 -yl)piperazine-1- carboxylate (0.5 g, 926.38 μmol, 65.46% yield) as a white solid. Step D. Procedure for preparation of tert-butyl 4-((S)-5-((1s,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)pentan-2-yl)piperazine-1-carboxylat e The stereoisomers of tert-butyl 4-(5-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)pentan-2-yl)piperazine-1-carboxylat e (1.0 g, 1.91 mmol, 1.0 equiv.) were separated by SFC to give tert-butyl 4-((S)-5-((1s,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)pentan-2-yl)piperazine-1-carboxylat e (490 mg, 935.93 μmol, 49.00% yield) as a yellow oil. Step E. Procedure for preparation of 1-((S)-5-((1s, 4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)pentan-2-yl)piperazine A mixture of tert-butyl 4-((S)-5-((1s,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)pent an-2- yl)piperazine-1-carboxylate (600 mg, 1.15 mmol, 1.0 equiv.) in HCl/dioxane (10 mL) was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by reverse-phase HPLC (with 0.1% formic acid) to give 1-((S)-5-((1s, 4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)pentan-2-yl)pipera zine (400 mg, 925.78 μmol, 80.78% yield) as a brown oil. MS (ESI) m/z: 423.3 [M+H] + Step F. Procedure for preparation of [3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6- yl]boronic acid To a solution of 6-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (2.0 g, 4.00 mmol, 1.0 equiv.) and triisopropyl borate (2.26 g, 11.99 mmol, 2.76 mL, 3.0 equiv.) in THF (5 mL) was added n-BuLi (2.5 M, 3.20 mL, 2.0 equiv.) at -78°C. The mixture was stirred at -78 °C for 1 hour under N 2 atmosphere. The reaction mixture was diluted with NH 4 Cl (100 mL) and extracted with EtOAc (50 mL × 3). The combined organic layers were washed with brine (50 mL × dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~2% dichloromethane: methanol) to give [3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-yl]boronic acid (1.9 g, 3.47 mmol, 86.84% yield,) as a white solid. MS (ESI) m/z: 466.3 [M+H] + Step G. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-(4-((S)-5- ((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)pentan-2-yl)p iperazin-1-yl)-1-methyl- 1H-indazole A mixture of 1-((S)-5-((1s, 4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)pentan-2- yl)piperazine (50 mg, 118.08 μmol, 1.0 equiv.), [3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl- indazol-6-yl]boronic acid (65.93 mg, 141.70 μmol, 1.2 equiv.), Cu(OAc)2 (4.29 mg, 23.62 μmol, 0.2 equiv.), TEA (35.85 mg, 354.25 μmol, 49.31 μL, 3 equiv.), and 4A MS (1 g, 118.08 μmol, 1.0 equiv.) in DCE (10 mL) was degassed and purged with O2 three times. The mixture was stirred at 25 °C for 12 hours under O2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , DCM: MeOH = 20 : 1) to give 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-(4-((S)-5-((1r,4s)- 4-(3-bromo-2-methylphenoxy)cyclohexyl)pentan-2-yl)piperazin- 1-yl)-1-methyl-1H-indazole (100 mg, 58.13 μmol, 12.31% yield) as a yellow oil. MS (ESI) m/z: 842.7 [M+H] + Step H. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-6-(4- ((S)-5-((1r,4R)-4-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2- yl)phenoxy)cyclohexyl)pentan-2-yl)piperazin-1-yl)-1H-indazol e A mixture of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-(4-((S)-5-((1r,4s)-4-(3 -bromo-2- methylphenoxy)cyclohexyl)pentan-2-yl)piperazin-1-yl)-1-methy l-1H-indazole (100 mg, 118.64 μmol, 1.0 equiv.), B 2 pin 2 (60.25 mg, 237.28 μmol, 2.0 equiv.), KOAc (34.93 mg, 355.91 μmol, 3.0 equiv.), and Pd(dppf)Cl2 (8.68 mg, 11.86 μmol, 0.1 equiv.) in DMSO (10 mL) was degassed and purged with N2 three times. The mixture was stirred at 100 °C for 12 hours under N 2 atmosphere. The reaction mixture was diluted with H 2 O (50 mL) and extracted with EtOAc (50 mL × 3). The combined organic layers were washed with brine (50 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , DCM: MeOH = 20 : 1) to give 3-(2,6-bis(benzyloxy)pyridin-3- yl)-1-methyl-6-(4-((S)-5-((1r,4R)-4-(2-methyl-3-(4,4,5,5-tet ramethyl-1,3,2-dioxaborolan-2- yl)phenoxy)cyclohexyl)pentan-2-yl)piperazin-1-yl)-1H-indazol e (70 mg, 39.33 μmol, 33.15% yield) as a yellow oil. MS (ESI) m/z: 890.8 [M+H] + Step I. Procedure for preparation of 3-(1-methyl-6-(4-((S)-5-((1r,4r)-4-(2-methyl-3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)cyclohe xyl)pentan-2-yl)piperazin- 1-yl)-1H-indazol-3-yl)piperidine-2,6-dione To a solution of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-6-(4-((S)-5-((1r ,4R)-4-(2- methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenox y)cyclohexyl)pentan-2- yl)piperazin-1-yl)-1H-indazole (70 mg, 78.65 μmol, 1.0 equiv.) in THF (5 mL) and EtOH (5 mL) was added Pd/C (8.37 mg, 7.87 μmol, 10% purity, 0.1 equiv.) and Pd(OH) 2 (5.52 mg, 7.87 μmol, 20% purity, 0.1 equiv.) under N2 atmosphere. The suspension was degassed and purged with H2 three times. The mixture was stirred under H2 (15 Psi) at 25 °C for 12 hours. The reaction mixture was then filtered and concentrated under reduced pressure to give 3-(1- methyl-6-(4-((S)-5-((1r,4r)-4-(2-methyl-3-(4,4,5,5-tetrameth yl-1,3,2-dioxaborolan-2- yl)phenoxy)cyclohexyl)pentan-2-yl)piperazin-1-yl)-1H-indazol -3-yl)piperidine-2,6-dione (80 mg, 50.58 μmol, 64.31% yield) as a yellow oil. MS (ESI) m/z: 712.7 [M+H] + Step J. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-((4S)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)pentyl)cyclohexyl)oxy) -2- methylphenyl)picolinate A mixture of 3-(1-methyl-6-(4-((S)-5-((1r,4r)-4-(2-methyl-3-(4,4,5,5-tetr amethyl-1,3,2- dioxaborolan-2-yl)phenoxy)cyclohexyl)pentan-2-yl)piperazin-1 -yl)-1H-indazol-3- yl)piperidine-2,6-dione (80 mg, 112.40 μmol, 1.0 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-bromo-pyridi ne-2-carboxylate (76.27 mg, 134.88 μmol, 1.2 equiv.), [2-(2-aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-buty l- phosphane;methanesulfonate (8.19 mg, 11.24 μmol, 0.1 equiv.), and KF (19.59 mg, 337.20 μmol, 7.90 μL, 3.0 equiv.) in dioxane (10 mL) and H2O (1 mL) was degassed and purged with N 2 three times. The mixture was stirred at 100 °C for 12 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM : MeOH= 20 : 1) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1r,4r)-4-((4S)-4- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1- yl)pentyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (30 mg, 19.62 μmol, 17.46% yield) as a yellow oil. MS (ESI) m/z: 1070.5 [M+H] + Step K. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-((4S)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)pentyl)cyclohexyl)oxy) -2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3- (((1R,4r)-4-((4S*)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-6-yl)piperazin- 1-yl)pentyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1r,4r)-4-((4S)-4-(4-(3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-6- yl)piperazin-1-yl)pentyl)cyclohexyl)oxy)-2-methylphenyl)pico linate (30 mg, 28.03 μmol, 1.0 equiv.) in TFA (2 mL) and DCM (2 mL) was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep- HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1r,4r)-4-((4S)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl -1H-indazol-6-yl)piperazin-1- yl)pentyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r) -4-((4S*)-4-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin-1-yl )pentyl)cyclohexyl)oxy)-2- methylphenyl)picolinic acid ] (14.11 mg, 13.56 μmol, 48.39% yield) as a yellow solid. MS (ESI) m/z: 1014.5 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 10.84 (s, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.4 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.51 - 7.32 (m, 6H), 7.10 - 7.01 (m, 1H), 6.96 - 6.86 (m, 3H), 6.84 - 6.79 (m, 1H), 6.62 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.28 - 4.12 (m, 2H), 3.93 - 3.86 (m, 5H), 3.19 (s, 4H), 3.02 (t, J = 5.6 Hz, 2H), 2.65 - 2.55 (m, 6H), 2.35 - 2.27 (m, 2H), 2.19 - 2.13 (m, 1H), 2.10 - 2.04 (m, 2H), 1.87 (s, 3H), 1.81 - 1.74 (m, 2H), 1.52 - 1.46 (m, 1H), 1.38 - 1.31 (m, 4H), 1.28 (s, 2H), 1.23 (s, 2H), 1.09 - 1.01 (m, 2H), 0.95 (d, J = 6.4 Hz, 3H) Example 268. Preparation of Compound 319a

6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1- yl)butyl)cyclohexyl)oxy)-2-(trifluoromethyl)phenyl)picolinic acid Step A. Procedure for preparation of methyl (1r,4r)-4-(3-bromo-2- (trifluoromethyl)phenoxy)cyclohexane-1-carboxylate A mixture of methyl (1s,4s)-4-hydroxycyclohexane-1-carboxylate (1.58 g, 9.96 mmol, 1.2 equiv.), 3-bromo-2-(trifluoromethyl)phenol (2 g, 8.30 mmol, 1.0 equiv.), and 2-(tributyl- phosphanylidene)acetonitrile (2.60 g, 10.79 mmol, 1.3 equiv.) in toluene (40 mL) was degassed and purged with N2 three times. The mixture was stirred at 120 °C for 12 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether / ethyl acetate = 100 / 1) to give methyl (1r,4r)-4-(3-bromo-2-(trifluoromethyl)phenoxy)cyclohexane- 1-carboxylate (2 g, 2.62 mmol, 31.61% yield, 50% purity) as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.49 - 7.42 (m, 1H), 7.41 - 7.34 (m, 2H), 4.60 - 4.46 (m, 1H), 3.60 (s, 3H), 2.45 - 2.40 (m, 1H), 2.06 - 2.00 (m, 2H), 1.96 - 1.89 (m, 2H), 1.58 - 1.43 (m, 4H). Step B. Procedure for preparation of ((1r,4r)-4-(3-bromo-2- (trifluoromethyl)phenoxy)cyclohexyl)methanol A mixture of methyl (1r,4r)-4-(3-bromo-2-(trifluoromethyl)phenoxy)cyclohexane-1- carboxylate (2 g, 2.62 mmol, 1.0 equiv.) in THF (40 mL) was degassed and purged with N 2 three times. To this mixture was added LiAlH4 (2.5 M, 1.05 mL, 1.0 equiv.). The reaction mixture was stirred at 0 °C for 2 hours under N2 atmosphere. The reaction mixture was quenched with aqueous Na 2 SO 4 (0.4 mL) at 0 °C. The mixture was diluted with ethyl acetate (60 mL), filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 19% ethyl acetate / petroleum ether) to give ((1r,4r)-4-(3-bromo-2-(trifluoromethyl)phenoxy)cyclohexyl)me thanol (450 mg, 1.15 mmol, 43.71% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.48 - 7.41 (m, 1H), 7.39 - 7.32 (m, 2H), 4.48 - 4.39 (m, 2H), 3.23 (t, J = 5.6 Hz, 2H), 2.05 (d, J = 9.2 Hz, 2H), 1.84 - 1.72 (m, 2H), 1.40 - 1.28 (m, 3H), 1.12 - 0.99 (m, 2H). Step C. Procedure for preparation of (1r,4r)-4-(3-bromo-2- (trifluoromethyl)phenoxy)cyclohexane-1-carbaldehyde A solution of oxalyl dichloride (323.45 mg, 2.55 mmol, 223.07 μL, 2.0 equiv.) in DCM (40 mL) was cooled to -78 °C, and DMSO (398.22 mg, 5.10 mmol, 398.22 μL, 4.0 equiv.) was added dropwise. The mixture was stirred at -78 °C for 2 hours under N2 atmosphere. Then ((1r,4r)-4-(3-bromo-2-(trifluoromethyl)phenoxy)cyclohexyl)me thanol (450 mg, 1.27 mmol, 1.0 equiv.) was added dropwise. The mixture was stirred at -78 °C for 1 hour under N2 atmosphere. TEA (773.59 mg, 7.64 mmol, 1.06 mL, 6 equiv.) was added dropwise to the mixture at -78 °C, and the mixture was warmed to 25 °C and stirred for 1 hour under N 2 atmosphere. The reaction mixture was quenched with water (40 mL) at 0 °C and extracted with ethyl acetate (50 mL × 3). The combined organic layers were washed with brine (20 mL × 3), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 5% ethyl acetate / petroleum ether) to give (1r,4r)-4-(3-bromo-2-(trifluoromethyl)phenoxy)cyclohexane-1- carbaldehyde (400 mg, 1.03 mmol, 80.46% yield) as a red oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 9.61 (s, 1H), 7.45 (t, J = 8.4 Hz, 1H), 7.39 - 7.34 (m, 2H), 4.57 - 4.48 (m, 1H), 2.42 - 2.35 (m, 1H), 2.04 - 1.90 (m, 4H), 1.55 - 1.40 (m, 4H). Step D. Procedure for preparation of 1-(((1r,4r)-4-((E)-4-(benzyloxy)but-1-en-1- yl)cyclohexyl)oxy)-3-bromo-2-(trifluoromethyl)benzene To a solution of (3-(benzyloxy)propyl)triphenylphosphonium bromide (1.12 g, 2.28 mmol, 2.0 equiv.) in THF (30 mL) was added LiHMDS (1 M, 3.42 mL, 3.0 equiv.) at 0 °C. The reaction mixture was stirred for 1 hour. Afterwards, (1r,4r)-4-(3-bromo-2- (trifluoromethyl)phenoxy)cyclohexane-1-carbaldehyde (400 mg, 1.14 mmol, 1.0 equiv.) was added, and the mixture was stirred at 0 °C for 3 hours under N2 atmosphere. The reaction mixture was quenched with aqueous NH 4 Cl (30 mL) at 0 °C and extracted with ethyl acetate (40 mL). The combined organic layers were washed with brine (20 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 3% ethyl acetate / petroleum ether) to give 1- (((1r,4r)-4-((E)-4-(benzyloxy)but-1-en-1-yl)cyclohexyl)oxy)- 3-bromo-2- (trifluoromethyl)benzene (470 mg, 875.12 μmol, 76.77% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.44 (d, J = 7.6 Hz, 1H), 7.40 - 7.24 (m, 7H), 5.52 - 5.18 (m, 2H), 4.55 - 4.36 (m, 3H), 3.47 - 3.37 (m, 2H), 2.36 - 2.27 (m, 3H), 2.03 (d, J = 9.6 Hz, 2H), 1.63 (d, J = 12.4 Hz, 2H), 1.51 - 1.34 (m, 2H), 1.31 - 1.12 (m, 2H). Step E. Procedure for preparation of 1-(((1s,4r)-4-(4-(benzyloxy)butyl)cyclohexyl)oxy)-3- bromo-2-(trifluoromethyl)benzene To a solution of 1-(((1r,4r)-4-((E)-4-(benzyloxy)but-1-en-1-yl)cyclohexyl)oxy )-3-bromo-2- (trifluoromethyl)benzene (470 mg, 972.36 μmol, 1.0 equiv.) in EtOAc (5 mL) was added Rh/Al2O3 (235.00 mg, 2.28 mmol, 2.35 equiv.) under N2 atmosphere. The suspension was degassed and purged with H2 three times. The mixture was stirred under H2 (15 Psi) at 25 °C for 12 hours. The reaction solution was filtered through the diatom layer under a moderate N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give 1-(((1s,4r)-4-(4-(benzyloxy)butyl)cyclohexyl)oxy)-3-bromo-2- (trifluoromethyl)benzene (460 mg, crude) as a colorless oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.48 - 7.41 (m, 1H), 7.38 - 7.27 (m, 7H), 4.44 (s, 3H), 3.43 - 3.39 (m, 2H), 2.03 (d, J = 9.2 Hz, 2H), 1.75 (d, J = 12.0 Hz, 2H), 1.55 - 1.49 (m, 2H), 1.36 - 1.29 (m, 4H), 1.24 - 1.14 (m, 3H), 1.10 - 0.94 (m, 2H). Step F. Procedure for preparation of 4-((1r,4s)-4-(3-bromo-2- (trifluoromethyl)phenoxy)cyclohexyl)butan-1-ol To a solution of 1-(((1s,4r)-4-(4-(benzyloxy)butyl)cyclohexyl)oxy)-3-bromo-2- (trifluoromethyl)benzene (460 mg, 947.72 μmol, 1.0 equiv.) in DCM (10 mL) was added TMSI (568.90 mg, 2.84 mmol, 387.00 μL, 3.0 equiv.) at 0 °C. The mixture was stirred at 25 °C for 2 hours under N2 atmosphere. The reaction mixture was quenched with water (10 mL) and extracted with dichloromethane (10 mL × 3). The combined organic layers were washed with brine (10 mL ×3), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 20% ethyl acetate / petroleum ether) to give compound 4-((1r,4s)-4-(3-bromo-2- (trifluoromethyl)phenoxy)cyclohexyl)butan-1-ol (220 mg, 500.94 μmol, 52.86% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.43 (d, J = 8.0 Hz, 1H), 7.39 - 7.27 (m, 2H), 4.44 (t, J = 10.4 Hz, 1H), 4.32 (t, J = 4.8 Hz, 1H), 3.43 - 3.34 (m, 2H), 2.12 - 1.93 (m, 2H), 1.76 (d, J = 12.4 Hz, 2H), 1.46 - 1.34 (m, 3H), 1.34 - 1.22 (m, 4H), 1.21 - 1.13 (m, 2H), 1.10 - 0.95 (m, 2H). Step G. Procedure for preparation of 4-((1r,4s)-4-(3-bromo-2- (trifluoromethyl)phenoxy)cyclohexyl)butanal To a solution of oxalyl dichloride (141.30 mg, 1.11 mmol, 97.45 μL, 2.0 equiv.) in DCM (30 mL) cooled to -78 °C, was added DMSO (173.95 mg, 2.23 mmol, 173.95 μL, 4.0 equiv.) dropwise. The mixture was stirred at -78 °C for 2 hours under N2 atmosphere. Then, 4-((1r,4s)- 4-(3-bromo-2-(trifluoromethyl)phenoxy)cyclohexyl)butan-1-ol (220 mg, 556.60 μmol, 1.0 equiv.) was added dropwise, and the mixture was stirred at -78 °C for 1 hour under N2 atmosphere. TEA (337.94 mg, 3.34 mmol, 464.84 μL, 6.0 equiv.) was added dropwise to the mixture at -78 °C, and the mixture was warmed to 25 °C and stirred at 25 °C for 1 hour under N 2 atmosphere. The reaction mixture was quenched with water (5 mL) and extracted with dichloromethane (10 mL × 3). The combined organic layers were washed with brine (10 mL × 3), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 12% ethyl acetate / petroleum ether) to give 4-((1r,4s)-4-(3-bromo-2-(trifluoromethyl)phenoxy)cyclohexyl) butanal (190 mg, 386.53 μmol, 69.45% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 9.66 (s, 1H), 7.49 - 7.40 (m, 1H), 7.39 - 7.31 (m, 2H), 4.52 - 4.35 (m, 1H), 2.41 (t, J = 6.8 Hz, 2H), 2.04 (d, J = 10.4 Hz, 2H), 1.76 (d, J = 12.0 Hz, 2H), 1.59 - 1.46 (m, 2H), 1.42 - 1.24 (m, 3H), 1.22 - 1.13 (m, 2H), 1.10 - 0.97 (m, 2H). Step H. Procedure for preparation of 3-(6-(4-(4-((1r,4s)-4-(3-bromo-2- (trifluoromethyl)phenoxy)cyclohexyl)butyl)piperazin-1-yl)-1- methyl-1H-indazol-3- yl)piperidine-2,6-dione A solution of 4-((1r,4s)-4-(3-bromo-2-(trifluoromethyl)phenoxy)cyclohexyl) butanal (190 mg, 483.17 μmol, 1.0 equiv.), 3-(1-methyl-6-(piperazin-1-yl)-1H-indazol-3-yl)piperidine-2, 6- dione (193.38 mg, 531.48 μmol, 1.1 eq, HCl), and NaOAc (39.64 mg, 483.17 μmol, 1.0 equiv.) in DCM (10 mL) was stirred at room temperature for 12 hours. NaBH(OAc) 3 (512.02 mg, 2.42 mmol, 5 equiv.) was then added. The mixture was stirred at 25 °C for 30 min. The reaction mixture was quenched with water (10 mL) and extracted with dichloromethane (10 mL × 3). The combined organic layers were washed with brine (10 mL × 3), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 20% ethyl acetate / petroleum ether) to give 3-(6-(4-(4-((1r,4s)- 4-(3-bromo-2-(trifluoromethyl)phenoxy)cyclohexyl)butyl)piper azin-1-yl)-1-methyl-1H- indazol-3-yl)piperidine-2,6-dione (240 mg, crude) as a white oil. MS (ESI) m/z: 706.1 [M+H] + . Step I. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy)- 2- (trifluoromethyl)phenyl)picolinate A mixture of 3-(6-(4-(4-((1r,4s)-4-(3-bromo-2- (trifluoromethyl)phenoxy)cyclohexyl)butyl)piperazin-1-yl)-1- methyl-1H-indazol-3- yl)piperidine-2,6-dione (240 mg, 340.61 μmol, 1.0 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tet ramethyl-1,3,2-dioxaborolan-2- yl)picolinate (250.37 mg, 408.73 μmol, 1.2 equiv.), KF (59.36 mg, 1.02 mmol, 23.94 μL, 3 equiv.), and Ad 2 nBuP Pd G 3 (cataCXium® A Pd G 3 ) (49.61 mg, 68.12 μmol, 0.2 equiv.) in dioxane (4 mL) and H 2 O (0.4 mL) was degassed and purged with N 2 three times. The mixture was stirred at 100 °C for 1 hour under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 20% dichloromethane / methanol) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1s,4r)-4-(4-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin -1-yl)butyl)cyclohexyl)oxy)-2- (trifluoromethyl)phenyl)picolinate (300 mg, 221.56 μmol, 65.05% yield) as a yellow solid. MS (ESI) m/z: 1110.5 [M+H] + . Step J. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy)-2- (trifluoromethyl)phenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1S,4R)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-6- yl)piperazin-1-yl)butyl)cyclohexyl)oxy)-2-(trifluoromethyl)p henyl)picolinate (300 mg, 270.20 μmol, 1.0 equiv.) in DCM (2 mL) was added TFA (3.07 g, 26.92 mmol, 2 mL, 99.65 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-6-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-(trifluoromethyl)phenyl)picolinic acid (30.62 mg, 28.20 μmol, 10.44% yield) as a brown solid. MS (ESI) m/z: 1054.5 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 13.27 - 12.29 (m, 1H), 10.85 (s, 1H), 8.14 (s, 1H), 8.03 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.52 - 7.31 (m, 7H), 7.28 - 7.22 (m, 1H), 7.02 - 6.88 (m, 2H), 6.85 (s, 1H), 6.64 (d, J = 7.6 Hz, 1H), 5.08 - 4.87 (m, 2H), 4.48 - 4.35 (m, 1H), 4.27 - 4.23 (m, 1H), 3.93 (t, J = 5.2 Hz, 2H), 3.89 (s, 3H), 3.24 (s, 6H), 3.02 (t, J = 4.8 Hz, 2H), 2.61 (d, J = 6.0 Hz, 4H), 2.43 (s, 2H), 2.23 - 2.13 (m, 2H), 2.11 - 2.04 (m, 2H), 1.83 - 1.75 (m, 2H), 1.52 - 1.44 (m, 2H), 1.38 - 1.28 (m, 4H), 1.23 (s, 2H), 1.16 - 0.99 (m, 3H). Example 269. Preparation of Compound 308b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4- ((3S)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 6-yl)piperazin-1-yl)-3- methylbutyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1R,4r)-4- ((3S*)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)piperazin-1-yl)-3- methylbutyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of (S)-4-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2-methylbutanal A mixture of (S)-4-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2-meth ylbutan-1-ol (120 mg, 337.73 μmol, 1.0 equiv.) and (1,1-diacetoxy-3-oxo-1,2-benziodoxol-1-yl) acetate (286.49 mg, 675.47 μmol, 209.27 μL, 2.0 equiv.) in DCM (5 mL) was degassed and purged with N 2 three times. The mixture was stirred at 25 °C for 3 hours under N 2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (25 mL × 3). The combined organic layers were washed with brine (15 mL × 3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give (S)-4-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2-methylbutanal (90 mg, 254.75 μmol, 75.43% yield) as a yellow oil. Step B. Procedure for preparation of 3-(6-(4-((S)-4-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2-methylbutyl)piperazin-1-yl)-1-me thyl-1H-indazol-3- yl)piperidine-2,6-dione A mixture of (S)-4-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)-2-meth ylbutanal (90 mg, 254.75 μmol, 1.0 equiv.), 3-(1-methyl-6-(piperazin-1-yl)-1H-indazol-3-yl)piperidine-2, 6- dione (100.08 mg, 305.70 μmol, 1.2 equiv.) in DCM (3 mL) was degassed and purged with N2 three times, and then the mixture was stirred at 25 °C for 15 hours under N 2 atmosphere. After 15 hours, NaBH(OAc) 3 (161.97 mg, 764.24 μmol, 3.0 equiv.) was added, and the mixture was stirred at 25°C for 30 minutes under N2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (25 mL × 3). The combined organic layers were washed with brine (15 mL) × 3, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 - 10% DCM / MeOH) to give 3-(6-(4-((S)-4-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2-methylbutyl)piperazin-1-yl)-1-me thyl-1H-indazol-3- yl)piperidine-2,6-dione (60 mg, 90.27 μmol, 35.44% yield) as a yellow solid. MS (ESI) m/z: 666.2 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 10.84 (s, 1H), 7.49 (d, J = 8.8 Hz, 1H), 7.15 - 7.12 (m, 1H), 7.07 (t, J = 8.0 Hz, 1H), 7.03 - 7.00 (m, 1H), 6.95 - 6.88 (m, 1H), 6.84 (s, 1H), 4.31 - 4.19 (m, 2H), 3.92 - 3.85 (m, 3H), 3.21 (s, 4H), 2.69 - 2.65 (m, 2H), 2.63 - 2.58 (m, 4H), 2.20 (s, 3H), 2.17 (d, J = 5.6 Hz, 2H), 2.08 - 2.02 (m, 2H), 1.83 - 1.75 (m, 2H), 1.71 - 1.60 (m, 1H), 1.50 - 1.38 (m, 2H), 1.37 - 1.29 (m, 2H), 1.26 - 1.19 (m, 4H), 1.13 - 0.98 (m, 4H), 0.88 (d, J = 6.4 Hz, 2H) Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-((3S)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-3-methylbutyl)cyclohe xyl)oxy)-2- methylphenyl)picolinate A mixture of 3-(6-(4-((S)-4-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexy l)-2- methylbutyl)piperazin-1-yl)-1-methyl-1H-indazol-3-yl)piperid ine-2,6-dione (82.94 mg, 135.40 μmol, 1.5 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)picolinate (60 mg, 90.27 μmol, 1.0 equiv.), KF (1.5 M, 180.54 μL, 3.0 equiv.), and Ad2nBuP Pd G3(cataCXium® A Pd G3 ) (6.57 mg, 9.03 μmol, 0.1 equiv.) in dioxane (2 mL) was degassed and purged with N 2 three times. The mixture was stirred at 100 °C for 2 hours under N 2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with DCM (25 mL × 3). The combined organic layers were washed with brine (15 mL × 3), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , DCM: MeOH = 10:1) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1r,4r)-4-((3S)-4- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1-yl)-3- methylbutyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (90 mg, 79.95 μmol, 88.57% yield) as a brown solid. MS (ESI) m/z: 1070.8 [M+H] + Step D. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-((3S)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-3-methylbutyl)cyclohe xyl)oxy)-2- methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-((3S*)-4-(4-(3 -(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)-3-methylbutyl)cyclohe xyl)oxy)-2- methylphenyl)picolinic acid] A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1r,4r)-4-((3S)-4-(4-(3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-6- yl)piperazin-1-yl)-3-methylbutyl)cyclohexyl)oxy)-2-methylphe nyl)picolinate (90 mg, 84.08 μmol, 1.0 equiv.) in DCM (1 mL) and TFA (0.3 mL) was degassed and purged with N2 three times. The mixture was stirred at 25 °C for 16 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep- HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1r,4r)-4-((3S)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl -1H-indazol-6-yl)piperazin-1-yl)- 3-methylbutyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r) -4-((3S*)-4-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin-1-yl )-3- methylbutyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] (27.48 mg, 26.56 μmol, 31.59% yield) as a white solid. MS (ESI) m/z: 1014.4 [M+H] + 1 H NMR (400 MHz, DMSO-d6) δ = 12.91 - 12.58 (m, 1H), 10.90 - 10.78 (m, 1H), 8.14 (d, J = 1.2 Hz, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.81 - 7.75 (m, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.51 - 7.42 (m, 4H), 7.39 - 7.33 (m, 2H), 7.10 - 7.02 (m, 1H), 6.97 - 6.88 (m, 3H), 6.84 (s, 1H), 6.64 - 6.57 (m, 1H), 4.97 (s, 2H), 4.28 - 4.14 (m, 2H), 3.92 - 3.90 (m, 2H), 3.88 (s, 3H), 3.22 - 3.20 (m, 4H), 3.03 - 3.01 (m, 2H), 2.65 - 2.57 (m, 2H), 2.37 - 2.24 (m, 2H), 2.20 - 2.14 (m, 2H), 2.14 - 2.02 (m, 4H), 1.90 - 1.85 (m, 3H), 1.81 - 1.74 (m, 2H), 1.70 - 1.63 (m, 1H), 1.47 - 1.41 (m, 1H), 1.35 (dd, J = 2.8, 1.6 Hz, 2H), 1.29 - 1.13 (m, 4H), 1.13 - 0.96 (m, 4H), 0.91 - 0.85 (m, 3H) Example 270. Preparation of Compound 296a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- (4-(3-(2,6-dioxopiperidin-3-yl)-5-fluoro-1-methyl-1H-indazol -6-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of 4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butanal To a solution of DMSO (457.88 mg, 5.86 mmol, 457.88 μL, 4 equiv.) in DCM (20 mL) was added oxalyl dichloride (371.91 mg, 2.93 mmol, 256.49 μL, 2 equiv.) in DCM (2 mL) dropwise at -78 °C under N2 atmosphere. The mixture was stirred at -78°C for 90 minutes. After which time 4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butan-1-ol (500 mg, 1.47 mmol, 1 equiv.) in DCM (2 mL) was added dropwise at -78 °C. The reaction mixture was stirred for 30 minutes at -78 °C. TEA (889.49 mg, 8.79 mmol, 1.22 mL, 6 equiv.) was then added into the solution. The solution was stirred at 25 °C for 1 hour under N2 atmosphere. The reaction mixture was quenched by addition of H 2 O (5mL) at 25 °C and extracted with DCM (5 mL × 3). The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~5% ethyl acetate / petroleum ether) to give 4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butanal (400 mg, 1.02 mmol, 69.29% yield) as a yellow oil. MS (ESI) m/z: 340.1 [M+H] +. 1 H NMR (400 MHz,CDCl3) δ = 9.77 (s, 1H), 7.17 - 7.08 (m, 1H), 6.96 (t, J = 8.0 Hz, 1H), 6.83 - 6.75 (m, 1H), 4.15 - 4.02 (m, 1H), 2.46 - 2.40 (m, 2H), 2.29 (s, 3H), 2.17 - 2.08 (m, 2H), 1.85 (d, J = 12.8 Hz, 2H), 1.66 (d, J = 7.6, 15.6 Hz, 2H), 1.50 - 1.38 (m, 3H), 1.28 - 1.21 (m, 2H), 1.09 - 0.96 (m, 2H) Step B. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(2-methyl-3-(((1s,4r)-4-(4- oxobutyl)cyclohexyl)oxy)phenyl)picolinate 4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butanal (400 mg, 1.18 mmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine-2-carb oxylate (866.64 mg, 1.41 mmol, 1.2 equiv.), KF (205.49 mg, 3.54 mmol, 82.86 μL, 3 equiv.), and [2-(2- aminophenyl)phenyl]palladium(1+)bis(1-adamantyl)-butyl-phosp hane methanesulfonate (171.73 mg, 235.80 μmol, 0.2 equiv.) were taken up into a microwave tube in dioxane (1 mL) and H2O (0.1 mL). The sealed tube was heated at 100 °C for 1 hour in a microwave reactor. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~30% ethyl acetate / petroleum ether) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)oxy)phenyl)picol inate (430 mg, 499.30 μmol, 42.35% yield) as a yellow solid. MS (ESI) m/z: 745.7 [M+H] +. Step C. Procedure for preparation of tert-butyl 4-[3-(2,6-dibenzyloxy-3-pyridyl)-5-fluoro- 1-methyl-indazol-6-yl]piperazine-1-carboxylate To a solution of 6-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-5-fluoro-1-methyl-inda zole (850 mg, 1.64 mmol, 1 equiv.) and tert-butyl piperazine-1-carboxylate (916.21 mg, 4.92 mmol, 3 equiv.) in toluene (20 mL) was added Pd2(dba)3 (150.15 mg, 163.97 μmol, 0.1 equiv.), Cs2CO3 (1.60 g, 4.92 mmol, 3 equiv.), and RuPhos (153.03 mg, 327.95 μmol, 0.2 equiv.). The mixture was stirred at 110 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~10% ethyl acetate/petroleum ether) to give tert-butyl 4-[3-(2,6-dibenzyloxy-3-pyridyl)-5-fluoro-1- methyl-indazol-6-yl]piperazine-1-carboxylate (700 mg, 1.11 mmol, 67.42% yield) as a white solid. MS (ESI) m/z: 624.5 [M+H] + . Step D. Procedure for preparation of tert-butyl 4-[3-(2,6-dioxo-3-piperidyl)-5-fluoro-1- methyl-indazol-6-yl]piperazine-1-carboxylate To a solution of tert-butyl 4-[3-(2,6-dibenzyloxy-3-pyridyl)-5-fluoro-1-methyl-indazol-6 - yl]piperazine-1-carboxylate (600 mg, 961.98 μmol, 1 equiv.) in THF (3 mL) and EtOH (3 mL) was added Pd(OH)2 (202.64 mg, 144.30 μmol, 10% purity, 0.15 equiv.), Pd/C (200 mg, 144.30 μmol, 10% purity, 0.15 equiv.), and AcOH (173.31 mg, 2.89 mmol, 165.21 μL, 3 equiv.) under a N 2 atmosphere. The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H2 (15 psi) at 25 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue to give tert-butyl 4-[3-(2,6- dioxo-3-piperidyl)-5-fluoro-1-methyl-indazol-6-yl]piperazine -1-carboxylate (400 mg, 858.39 μmol, 89.23% yield) as a yellow solid. MS (ESI) m/z: 446.1 [M+H] + . Step E. Procedure for preparation of 3-(5-fluoro-1-methyl-6-piperazin-1-yl-indazol-3- yl)piperidine-2,6-dione A solution of tert-butyl 4-[3-(2,6-dioxo-3-piperidyl)-5-fluoro-1-methyl-indazol-6- yl]piperazine-1-carboxylate (380 mg, 853.00 μmol, 1 equiv.) in HCl/dioxane (2 mL) and DCM (2 mL) was stirred at 25 °C for 5 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue to give 3-(5-fluoro-1-methyl-6-piperazin-1-yl- indazol-3-yl)piperidine-2,6-dione (300 mg, 762.66 μmol, 89.41% yield) as a white solid. MS (ESI) m/z: 345.9 [M+H] + . Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-5-fluoro- 1-methyl-1H-indazol-6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy )-2- methylphenyl)picolinate A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(2-methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)oxy)phe nyl)picolinate (100 mg, 134.24 μmol, 1 equiv.) and 3-(5-fluoro-1-methyl-6-piperazin-1-yl-indazol-3-yl)piperidin e-2,6-dione (46.36 mg, 134.24 μmol, 1 equiv.) in DCM (1.5 mL) was stirred at 25 °C for 15.5 hours. To this solution was added NaBH(OAc)3 (85.35 mg, 402.72 μmol, 3 equiv.). The resulting mixture was stirred at 25 °C for 0.5 hours. The reaction mixture was concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl )-5-fluoro-1-methyl-1H-indazol- 6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)pic olinate (100 mg, 61.16 μmol, 45.56% yield) as a yellow oil. MS (ESI) m/z: 538.0 [M/2+H] + . Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-5-fluoro- 1-methyl-1H-indazol-6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy )-2-methylphenyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-5-fl uoro-1-methyl-1H-indazol-6- yl)piperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picol inate (90 mg, 83.77 μmol, 1 equiv.) in DCM (1.5 mL) and TFA (1 mL) was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-5-fl uoro-1-methyl-1H-indazol-6- yl)piperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picol inic acid (26.86 mg, 25.88 μmol, 30.89% yield) as a white solid. MS (ESI) m/z: 1018.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.85 (s, 1H), 8.16 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.51 - 7.41 (m, 4H), 7.40 - 7.32 (m, 2H), 7.12 - 7.02 (m, 2H), 6.93 (dd, J = 8.8, 13.6 Hz, 2H), 6.61 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.27 (dd, J = 5.2, 9.6 Hz, 1H), 4.24 - 4.13 (m, 1H), 3.94 (s, 3H), 3.91 (t, J = 6.0 Hz, 2H), 3.07 (s, 3H), 3.02 (t, J = 5.6 Hz, 2H), 2.68 - 2.60 (m, 2H), 2.55 (d, J = 6.0 Hz, 4H), 2.38 - 2.31 (m, 3H), 2.13 (dd, J = 5.6, 13.6 Hz, 1H), 2.07 (t, J = 4.8 Hz, 2H), 1.87 (s, 3H), 1.78 (d, J = 10.0 Hz, 2H), 1.58 - 1.19 (m, 10H), 1.13 - 0.98 (m, 2H) Example 271. Preparation of Compound 306 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1'-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)-[1,4'-bipiper idin]-4-yl)propoxy)-2- methylphenyl)picolinic acid Step A. Preparation of 8-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-6- yl)- 1,4-dioxa-8-azaspiro[4.5]decane 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-bromo-1-methyl-1H-indaz ole (500 mg, 1 equiv, 999 μmol) and 1,4-dioxa-8-azaspiro[4.5]decane (172 mg, 153 μL, 1.2 equiv., 1.20 mmol) were suspended in 1,4-dioxane (10 mL). To this mixture (2-dicyclohexylphosphino-2',6'- diisopropoxy-1,1'-biphenyl)[2-(2'-amino-1,1'-biphenyl)]palla dium(II) methanesulfonate (83.6 mg, 0.1 equiv, 99.9 μmol), followed by sodium 2-methylpropan-2-olate (288 mg, 1.50 mL, 2.0 molar, 3 equiv, 3.00 mmol), was added dropwise. The mixture was purged with N2 and heated at 100ºC for 1 hour. The reaction was quenched with water, and concentrated to dryness. The residue was extracted with ethyl acetate and water. The organic layer was separated, washed with brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness. The residue was then purified by silica gel chromatography (0-100% ethyl acetate in heptane) to afford 8-(3-(2,6- bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-6-yl)-1,4-di oxa-8-azaspiro[4.5]decane (452 mg, 803 μmol, 80.4 %) as an orange oil. MS (ESI) m/z: 563.4 [M+H] + Step B. Preparation of 3-(1-methyl-6-(1,4-dioxa-8-azaspiro[4.5]decan-8-yl)-1H-indaz ol- 3-yl)piperidine-2,6-dione 8-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-6- yl)-1,4-dioxa-8- azaspiro[4.5]decane (450 mg, 1 equiv., 800 μmol) was dissolved in ethyl acetate (10 mL) and DMF (1mL). To this mixture was added Pd/C (426 mg, 10% Wt, 0.5 equiv., 400 μmol). The resulting mixture was stirred at 50ºC under a H2 atmosphere overnight. The mixture was filtered through celite and rinsed with ethyl acetate (3 times). The filtrate was then concentrated under reduced pressure to afford 3-(1-methyl-6-(1,4-dioxa-8-azaspiro[4.5]decan-8-yl)-1H-indaz ol- 3-yl)piperidine-2,6-dione (225 mg, 585 μmol, 73.2 %) as an off-white solid. MS (ESI) m/z: 385.1 [M+H] + Step C. Preparation of 3-(1-methyl-6-(4-oxopiperidin-1-yl)-1H-indazol-3-yl)piperidi ne- 2,6-dione 3-(1-methyl-6-(1,4-dioxa-8-azaspiro[4.5]decan-8-yl)-1H-indaz ol-3-yl)piperidine-2,6-dione (180 mg, 1 equiv., 468 μmol) was dissolved in water (2 mL) and formic acid (21.6 mg, 17.7 μL, 1 equiv., 468 μmol). The reaction was heated at 60 ~ 80ºC for 8 hours. The reaction was cooled down to room temperature and concentrated to afford 3-(1-methyl-6-(4-oxopiperidin- 1-yl)-1H-indazol-3-yl)piperidine-2,6-dione (155 mg, 455 μmol, 97.3 %) as a brown solid. MS (ESI) m/z: 341.1 [M+H] + Step D. Preparation of 2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phen ol 3-bromo-2-methylphenol (2 g, 1 equiv, 0.01 mol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2- dioxaborolane) (4 g, 1.5 equiv, 0.02 mol), potassium acetate (3 g, 3 equiv, 0.03 mol), and PdCl2(dppf)-CH2Cl2 adduct (0.4 g, 0.05 equiv, 0.5 mmol) were suspended in 1,4-dioxane (20 mL). The mixture was purged with N 2 , stirred, and heated at 85ºC overnight. The mixture was cooled down to room temperature. To this mixture was added ethyl acetate (100 mL). The resulting mixture was filtered and evaporated to dryness. The resulting residue was then purified by silica gel chromatography (0-10% MeOH in DCM) to afford 2-methyl-3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)phenol (3.2 g, 14 mmol, 100 %) as a light yellow solid. MS (ESI) m/z: 235.3 [M+H] + Step E. Preparation of tert-butyl 4-(3-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)phenoxy)propyl)piperidine-1-carboxylate 2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phen ol (300 mg, 1 equiv., 1.28 mmol) was dissolved in acetonitrile (5 mL). To this mixture was added tert-butyl 4-(3- bromopropyl)piperidine-1-carboxylate (471 mg, 1.2 equiv., 1.54 mmol), followed by K2CO3 (531 mg, 3 equiv, 3.84 mmol). The mixture was then purged with N2, stirred, and heated at 70ºC overnight. After completion, the mixture was then cooled down to room temperature, filtered, and rinsed with acetonitrile. The filtrate was then evaporated to dryness. The residue was then purified by silica gel chromatography (0-100% ethyl acetate in heptane) to afford tert- butyl 4-(3-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)phenoxy)propyl)piperidine-1-carboxylate (620 mg, 1.35 mmol, 105 %) as a colorless oil, which was carried forward without further purification. Step F. Preparation of 4-(3-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)phenoxy)propyl)piperidine, HCl tert-butyl 4-(3-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)phenoxy)propyl)piperidine-1-carboxylate (620 mg, 1 equiv., 1.35 mmol) was dissolved in HCl in dioxane (4 M, 4 mL). The mixture was then stirred at room temperature for 1 hr. The reaction was concentrated under reduced pressure to afford 4-(3-(2-methyl-3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)propyl)piperidin e, HCl (531 mg, 1.34 mmol, 99.4 %) as a white solid. MS (ESI) m/z: 361.0 [M+H] + Step G. Preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(2-methyl-3-(3-(piperidin-4- yl)propoxy)phenyl)picolinate tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- bromopicolinate (300 mg, 1 equiv., 531 μmol) was suspended in 1,4-dioxane (3 mL) and 1.5 M K3PO4 (1 mL). To this mixture was added 4-(3-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)phenoxy)propyl)piperidine, HCl (252 mg, 1.2 equiv., 637 μmol), followed by mesylate[(di(1-adamantyl)-n-butylphosphine)-2-(2'-amino-1,1' -biphenyl)]palladium(II) (38.6 mg, 0.1 equiv., 53.1 μmol). The mixture was then heated at 100ºC for 40 minutes in a microwave reactor. The mixture was extracted with ethyl acetate and water. The organic layer was then separated, washed with brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness. The crude product was then purified by reverse phase-HPLC to afford tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(2-methyl-3-(3- (piperidin-4-yl)propoxy)phenyl)picolinate (103 mg, 143 μmol, 27.0 %) as a white solid. MS (ESI) m/z: 718.5 [M+H] + Step H. Preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1'-(3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H- indazol-6-yl)-[1,4'-bipiperidin]-4-yl)propoxy)-2-methylpheny l)picolinate tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(3-(piperidin-4-yl)propoxy)phenyl)picolinate (75 mg, 1 equiv., 0.10 mmol) and 3-(1- methyl-6-(4-oxopiperidin-1-yl)-1H-indazol-3-yl)piperidine-2, 6-dione (53 mg, 1.5 equiv, 0.16 mmol) were suspended in DCM (10 mL). To this mixture was added sodium triacetoxyborohydride (66 mg, 3 equiv, 0.31 mmol). The mixture was then purged with N 2 and stirred at room temperature overnight. The reaction mixture was concentrated to dryness and purified by reverse phase-HPLC to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1'-(3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H-indazol- 6-yl)-[1,4'-bipiperidin]-4-yl)propoxy)-2-methylphenyl)picoli nate (17 mg, 16 μmol, 16 %) as a white solid. MS (ESI) m/z: 521.9 [M+2H] 2+ Step I. Preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1'-(3-(2,6-dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)-[1,4'- bipiperidin]-4-yl)propoxy)-2-methylphenyl)picolinic acid, trifluoroacetic acid tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3- (1'-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)-[ 1,4'-bipiperidin]-4-yl)propoxy)- 2-methylphenyl)picolinate (17 mg, 1 equiv, 16 μmol) was dissolved in DCM (4 mL). To this mixture was added 2 mL TFA. The mixture was then stirred at room temperature overnight. The mixture was then concentrated to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1'-(3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H-indazol- 6-yl)-[1,4'-bipiperidin]-4-yl)propoxy)-2-methylphenyl)picoli nic acid, trifluoroacetic acid (18 mg, 16 μmol, 100 %) as a white solid. MS (ESI) m/z: 986.5 [M+H] + ; 1 H NMR (400 MHz, DMSO) δ 10.78 (s, 1H), 8.90 (s, 1H), 7.96 (d, J = 7.9 Hz, 1H), 7.72 (d, J = 8.0 Hz, 1H), 7.56 (d, J = 7.5 Hz, 1H), 7.48 – 7.24 (m, 6H), 7.03 (t, J = 7.9 Hz, 1H), 6.94 – 6.78 (m, 4H), 6.57 (d, J = 7.5 Hz, 1H), 4.91 (s, 2H), 4.19 (dd, J = 9.3, 5.1 Hz, 1H), 3.94 – 3.83 (m, 5H), 3.83 (s, 3H), 3.43 (d, J = 11.5 Hz, 2H), 3.00 – 2.84 (m, 4H), 2.72 (t, J = 12.3 Hz, 2H), 2.60 – 2.45 (m, 2H), 2.23 (dd, J = 9.1, 4.8 Hz, 1H), 2.09 (dt, J = 7.6, 5.5 Hz, 1H), 2.04 (s, 1H), 2.01 (s, 1H), 1.92 – 1.82 (m, 2H), 1.83 (s, 3H), 1.70 (d, J = 9.7 Hz, 4H), 1.32 (dd, J = 23.5, 10.4 Hz, 4H). Example 272. Preparation of Compound 312 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-((1-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)azetidin- 3-yl)methyl)piperidin-4- yl)propoxy)-2-methylphenyl)picolinic acid Step A. Preparation of (1-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-6 - yl)azetidin-3-yl)methanol 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-bromo-1-methyl-1H-indaz ole (2.0 g, 1 equiv.., 4.0 mmol) and azetidin-3-ylmethanol, HCl (0.59 g, 1.2 equiv., 4.8 mmol) were suspended in 1,4- dioxane (20 mL). To this mixture was added (2-dicyclohexylphosphino-2',6'-diisopropoxy- 1,1'-biphenyl)[2-(2'-amino-1,1'-biphenyl)]palladium(II) methanesulfonate (0.33 g, 0.1 equiv., 0.40 mmol). The mixture was purged with N2.2-methylpropan-2-olate potassium (0.54 g, 4.8 mL, 1.0 M, 1.2 equiv., 4.8 mmol) was then added dropwise. The mixture was then heated at 40ºC under N 2 and stirred overnight. The mixture was cooled down to room temperature, quenched with water, filtered, and rinsed with ethyl acetate (3 x). The filtrate was evaporated to dryness and then purified by silica gel chromatography (0-100% ethyl acetate in heptane) to afford (1-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-6 -yl)azetidin-3- yl)methanol (260 mg, 513 μmol, 13 %) as an off-white solid. MS (ESI) m/z: 507.3 [M+H] + Step B. Preparation of 3-(6-(3-(hydroxymethyl)azetidin-1-yl)-1-methyl-1H-indazol-3- yl)piperidine-2,6-dione (1-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-6 -yl)azetidin-3-yl)methanol (260 mg, 1 equiv., 513 μmol) was dissolved in ethyl acetate (5 mL). To the mixture was added Pd(OH) 2 (90.1 mg, 20% Wt, 0.25 equiv., 128 μmol). The mixture was stirred at 50ºC under a H2 balloon overnight. The mixture was then cooled down to room temperature and filtered through celite. The filtrate was then concentrated and purified by reverse-phase HPLC to afford 3-(6-(3-(hydroxymethyl)azetidin-1-yl)-1-methyl-1H-indazol-3- yl)piperidine-2,6- dione (15 mg, 46 μmol, 8.9 %) as a white solid. MS (ESI) m/z: 329.2 [M+H] + Step C. Preparation of 1-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)azetidine-3-carbaldehyde 3-(6-(3-(hydroxymethyl)azetidin-1-yl)-1-methyl-1H-indazol-3- yl)piperidine-2,6-dione (30 mg, 1 equiv., 91 μmol) was dissolved in DCM (2 mL). To this mixture was added Dess-Martin periodinane (43 mg, 31 μL, 1.1 equiv., 0.10 mmol). The mixture was stirred at room temperature for ~ 2hrs. The mixture was carried onto the following step without further purification. Step D. Preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-((1-(3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)azetidin-3-yl)methyl)piperidin-4-yl)propoxy)-2- methylphenyl)picolinate tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(3-(piperidin-4-yl)propoxy)phenyl)picolinate (15 mg, 1 equiv., 21 μmol) was added to a mixture of 1-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)azet idine-3- carbaldehyde (8 mg, 1.2 equiv., 25 μmol). To this mixture was added sodium triacetoxyborohydride (22 mg, 5 equiv., 0.10 mmol). The mixture was purged with N2 and stirred at room temperature overnight. The reaction mixture was concentrated to dryness and purified by reverse phase HPLC to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-(1-((1-(3-(2,6-dioxopip eridin-3-yl)-1-methyl-1H- indazol-6-yl)azetidin-3-yl)methyl)piperidin-4-yl)propoxy)-2- methylphenyl)picolinate (4.7 mg, 4.6 μmol, 22 %) as a white solid. MS (ESI) m/z: 514.9 [M+2H] 2+ Step E. Preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1-((1-(3-(2,6-dioxopiperidin-3-yl)-1-meth yl-1H-indazol-6-yl)azetidin- 3-yl)methyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinic acid, trifluoroacetic acid tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-(1- ((1-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)az etidin-3-yl)methyl)piperidin-4- yl)propoxy)-2-methylphenyl)picolinate (4.7 mg, 1 equiv., 4.6 μmol) was dissolved in DCM (2 mL). To this mixture was added TFA (1 mL). The mixture was then stirred at room temperature for ~ 24 hours. The mixture was then concentrated under reduced pressure to afford 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-(1-((1-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)azetidin-3-yl) methyl)piperidin-4- yl)propoxy)-2-methylphenyl)picolinic acid, trifluoroacetic acid (4.9 mg, 4.5 μmol, 99 %) as an off-white solid. MS (ESI) m/z: 486.9 [M+2H] 2+ ; 1 H NMR (400 MHz, DMSO) δ 12.79 (s, 1H), 10.76 (d, J = 6.2 Hz, 1H), 7.94 (dd, J = 17.4, 8.0 Hz, 2H), 7.72 (d, J = 8.0 Hz, 1H), 7.64 (dd, J = 7.7, 1.7 Hz, 1H), 7.56 (d, J = 7.6 Hz, 1H), 7.46 – 7.24 (m, 7H), 7.17 (td, J = 7.6, 1.7 Hz, 1H), 7.04 (t, J = 7.9 Hz, 1H), 6.91 (d, J = 9.0 Hz, 1H), 6.82 (d, J = 8.2 Hz, 1H), 6.57 (d, J = 7.7 Hz, 1H), 6.30 (d, J = 9.6 Hz, 1H), 4.91 (s, 2H), 4.22 – 4.09 (m, 1H), 4.02 (t, J = 7.5 Hz, 1H), 3.94 – 3.83 (m, 4H), 3.78 (s, 2H), 3.57 (t, J = 6.7 Hz, 1H), 3.13 (s, 2H), 2.96 (s, 2H), 2.85 (d, J = 12.7 Hz, 2H), 2.55 (d, J = 6.3 Hz, 1H), 2.47 (s, 1H), 2.08 (dd, J = 13.4, 6.0 Hz, 1H), 1.83 (d, J = 4.5 Hz, 4H), 1.71 (s, 3H), 1.50 (s, 1H), 1.34 (s, 4H). Example 273. Preparation of Compound 318a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)-3, 3-dimethylpiperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-3,3-dimethylpiperazin-1-yl)propyl)cy clohexyl)oxy)-2- methylphenyl)picolinate tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(((1r,4r)-4-(3-oxopropyl)cyclohexyl)oxy)phenyl)pico linate (30 mg, 1 equiv.., 41 μmol) and 3-(6-(2,2-dimethylpiperazin-1-yl)-1-methyl-1H-indazol-3-yl)p iperidine-2,6-dione, HCl (18 mg, 1.1 equiv., 45 μmol) were suspended in DCM (2 mL). To this mixture was added sodium triacetoxyborohydride (26 mg, 18 μL, 3 equiv., 0.12 mmol). The mixture was then stirred at room temperature overnight. The mixture was then extracted with DCM and water. The organic layer was washed with brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-6-yl)-3,3- dimethylpiperazin-1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl )picolinate as a light brown solid, which was carried forward without further purification. MS (ESI) m/z: 535.9 [M+2H] 2+ . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-3,3-dimethylpiperazin-1-yl)propyl)cy clohexyl)oxy)-2- methylphenyl)picolinic acid tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-6-yl)-3,3- dimethylpiperazin-1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl )picolinate (44 mg, 1 equiv., 41 μmol) was dissolved in DCM (3 mL). To this mixture was added 1 mL TFA (1.4 g, 0.95 mL, 300 equiv., 12 mmol). The mixture was then stirred at room temperature overnight. The reaction mixture was concentrated, and then purified by reverse phase-HPLC to afford 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1s,4r)-4-(3-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)-3,3-dime thylpiperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (21 mg, 21 μmol, 50 %) as a white solid. MS (ESI) m/z: 1014.5 [M+H] + ; 1 H NMR (400 MHz, DMSO) δ 12.79 (s, 1H), 12.49 (s, 1H), 10.80 (s, 1H), 7.96 (d, J = 7.8 Hz, 1H), 7.72 (d, J = 8.1 Hz, 1H), 7.56 (d, J = 7.5 Hz, 1H), 7.46 (d, J = 8.7 Hz, 1H), 7.44 – 7.35 (m, 3H), 7.29 (dt, J = 9.4, 7.3 Hz, 2H), 7.18 (s, 1H), 7.01 (t, J = 7.9 Hz, 1H), 6.93 – 6.80 (m, 3H), 6.55 (d, J = 7.5 Hz, 1H), 4.91 (s, 2H), 4.25 (dd, J = 9.6, 5.1 Hz, 1H), 4.12 (d, J = 10.8 Hz, 1H), 3.86 (d, J = 11.1 Hz, 5H), 3.07 (s, 2H), 2.96 (t, J = 5.9 Hz, 2H), 2.55 (td, J = 10.7, 4.8 Hz, 2H), 2.47 (s, 1H), 2.24 (s, 5H), 2.10 (dd, J = 13.3, 5.6 Hz, 1H), 2.03 (s, 2H), 2.00 (s, 1H), 1.80 (s, 3H), 1.74 (d, J = 12.5 Hz, 2H), 1.43 (s, 2H), 1.20 (d, J = 6.3 Hz, 2H), 1.00 (s, 7H). Example 274. Preparation of Compound 322a

6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)-3, 3-dimethylpiperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-( 2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-3,3-dimethylpiperazin-1-yl)butyl)cyc lohexyl)oxy)-2- methylphenyl)picolinate tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)oxy)phenyl)picol inate (60 mg, 1 equiv., 81 μmol) and 3-(6-(2,2-dimethylpiperazin-1-yl)-1-methyl-1H-indazol-3-yl)p iperidine-2,6-dione, HCl (35 mg, 1.1 equiv., 89 μmol) were suspended in DCM (5 mL). To this mixture was added sodium triacetoxyborohydride (51 mg, 3 equiv., 0.24 mmol). The mixture was then stirred at room temperature overnight. The mixture was partitioned between DCM and water. The organic layer was washed with brine, dried over Na2SO4, filtered, and evaporated to dryness to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-6-yl)-3,3- dimethylpiperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl) picolinate, which was carried forward without further purification. MS (ESI) m/z: 542.9 [M+2H] 2+ . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-3,3-dimethylpiperazin-1-yl)butyl)cyc lohexyl)oxy)-2- methylphenyl)picolinic acid tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-6-yl)-3,3- dimethylpiperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl) picolinate (88 mg, 1 equiv., 81 μmol) was dissolved in DCM (4 mL). To this mixture was added TFA (1 mL). The mixture was then stirred at room temperature overnight. The mixture was then concentrated and purified by reverse phase-HPLC to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-6-yl)-3,3- dimethylpiperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl) picolinic acid (41 mg, 40 μmol, 49 %) as a white solid. MS (ESI) m/z: 1028.5 [M+H] + ; 1 H NMR (400 MHz, DMSO) δ 12.79 (s, 1H), 12.49 (s, 1H), 10.81 (s, 1H), 7.96 (d, J = 7.7 Hz, 1H), 7.72 (d, J = 8.0 Hz, 1H), 7.59 – 7.49 (m, 2H), 7.44 – 7.21 (m, 5H), 7.01 (t, J = 7.8 Hz, 1H), 6.93 – 6.81 (m, 2H), 6.55 (d, J = 7.5 Hz, 1H), 4.91 (s, 2H), 4.27 (dd, J = 9.6, 5.0 Hz, 1H), 4.14 (s, 1H), 3.88 (d, J = 23.4 Hz, 4H), 3.67 (s, 1H), 3.41 (d, J = 16.0 Hz, 2H), 3.06 (s, 3H), 2.96 (t, J = 6.2 Hz, 2H), 2.89 (s, 1H), 2.56 (d, J = 5.9 Hz, 1H), 2.10 (dd, J = 13.2, 5.6 Hz, 1H), 2.06 – 1.98 (m, 2H), 1.80 (s, 2H), 1.73 (d, J = 12.7 Hz, 2H), 1.28 (s, 5H), 1.21 (s, 2H), 1.13 (s, 2H), 1.05 (s, 3H), 0.99 (s, 2H). Example 275. Preparation of Compound 323a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)-2, 2-dimethylpiperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-2,2-dimethylpiperazin-1-yl)butyl)cyc lohexyl)oxy)-2- methylphenyl)picolinate tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)oxy)phenyl)picol inate (60 mg, 1 equiv., 81 μmol) and 3-(6-(3,3-dimethylpiperazin-1-yl)-1-methyl-1H-indazol-3-yl)p iperidine-2,6-dione, HCl (31 mg, 1.1 equiv., 89 μmol) were suspended in DCM (5 mL). To this mixture was added sodium triacetoxyborohydride (51 mg, 3 equiv., 0.24 mmol). The mixture was then stirred at room temperature overnight. The mixture was then partitioned between DCM and water. The organic layer was washed with brine, dried over Na2SO4, filtered, and evaporated to dryness to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-6-yl)-2,2- dimethylpiperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl) picolinate, which was carried forward without further purification. MS (ESI) m/z: 542.9 [M+2H] 2+ . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-2,2-dimethylpiperazin-1-yl)butyl)cyc lohexyl)oxy)-2- methylphenyl)picolinic acid tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-6-yl)-2,2- dimethylpiperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl) picolinate (88 mg, 1 equiv., 81 μmol) was dissolved in DCM (4 mL). To this mixture was added TFA (1 mL). The mixture was stirred at room temperature overnight. The mixture was then concentrated, and purified by reverse phase-HPLC to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-6-yl)-2,2- dimethylpiperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl) picolinic acid (34 mg, 33 μmol, 41 %) as a white solid. MS (ESI) m/z: 1028.5 [M+H] + ; 1 H NMR (400 MHz, DMSO) δ 10.77 (s, 1H), 7.99 – 7.92 (m, 1H), 7.72 (d, J = 8.0 Hz, 1H), 7.55 (d, J = 7.7 Hz, 1H), 7.44 – 7.34 (m, 4H), 7.34 – 7.23 (m, 2H), 7.00 (t, J = 7.9 Hz, 1H), 6.91 – 6.78 (m, 3H), 6.74 (d, J = 1.9 Hz, 1H), 6.54 (d, J = 7.4 Hz, 1H), 4.91 (s, 2H), 4.18 (dd, J = 9.1, 5.1 Hz, 1H), 3.88 – 3.79 (m, 2H), 3.82 (s, 3H), 3.11 (d, J = 7.7 Hz, 2H), 2.96 (t, J = 6.0 Hz, 2H), 2.88 (s, 2H), 2.58 (d, J = 6.6 Hz, 3H), 2.58 – 2.43 (m, 2H), 2.27 (s, 3H), 2.25 – 2.15 (m, 0H), 2.09 (dd, J = 13.2, 5.9 Hz, 1H), 2.01 (t, J = 5.9 Hz, 2H), 1.80 (s, 3H), 1.72 (d, J = 12.6 Hz, 2H), 1.36 – 1.25 (m, 2H), 1.17 (s, 2H), 0.99 (s, 6H). Example 276. Preparation of Compound 324a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- (4-(3-(2,6-dioxopiperidin-3-yl)-1,7-dimethyl-1H-indazol-6-yl )piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1,7- dimethyl-1H-indazol-6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy )-2- methylphenyl)picolinate tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)oxy)phenyl)picol inate (60 mg, 1 equiv., 81 μmol) and 3-(1,7-dimethyl-6-(piperazin-1-yl)-1H-indazol-3-yl)piperidin e-2,6-dione, HCl (33 mg, 1.1 equiv., 89 μmol) were suspended in DCM (5 mL). To this mixture was added sodium triacetoxyborohydride (51 mg, 3 equiv., 0.24 mmol). The mixture was then stirred at room temperature overnight. The mixture was then extracted DCM and water. The organic layer was washed with brine, dried over Na2SO4, filtered, and evaporated to dryness to afford to tert- butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)- 4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-1,7-dimethyl-1H-indazol -6-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate, which was carried forward without further purification. MS (ESI) m/z: 535.9 [M+2H] 2+ . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1,7- dimethyl-1H-indazol-6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy )-2-methylphenyl)picolinic acid tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-1,7-dimethyl- 1H-indazol-6-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (87 mg, 1 equiv., 81 μmol) was dissolved in DCM (4 mL). To this mixture was added TFA (1 mL). The mixture was stirred at room temperature overnight. The mixture was then concentrated, and purified by reverse phase- HPLC to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-1,7-dimethyl- 1H-indazol-6-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (49 mg, 48 μmol, 60 %) as a white solid. MS (ESI) m/z: 1014.5 [M+H] + ; 1 H NMR (400 MHz, DMSO) δ 10.78 (s, 1H), 7.99 – 7.93 (m, 1H), 7.72 (d, J = 8.0 Hz, 1H), 7.56 (d, J = 7.5 Hz, 1H), 7.44 – 7.34 (m, 4H), 7.34 – 7.23 (m, 2H), 7.00 (t, J = 7.9 Hz, 1H), 6.93 – 6.82 (m, 3H), 6.55 (d, J = 7.4 Hz, 1H), 4.91 (s, 2H), 4.21 (dd, J = 9.6, 5.1 Hz, 1H), 4.11 (s, 3H), 3.85 (t, J = 5.9 Hz, 2H), 2.96 (t, J = 5.8 Hz, 2H), 2.79 (t, J = 4.7 Hz, 4H), 2.61 – 2.43 (m, 5H), 2.33 – 2.17 (m, 3H), 2.04 (dt, J = 27.1, 5.2 Hz, 3H), 1.80 (s, 3H), 1.72 (d, J = 12.4 Hz, 2H), 1.39 (s, 1H), 1.28 (s, 1H), 1.25 (s, 4H), 1.17 (s, 2H), 0.98 (q, J = 11.9 Hz, 2H). Example 277. Preparation of Compound 325a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- (4-(3-(2,6-dioxopiperidin-3-yl)-1,5-dimethyl-1H-indazol-6-yl )piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-( 2,6-dioxopiperidin-3-yl)-1,5- dimethyl-1H-indazol-6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy )-2- methylphenyl)picolinate tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)oxy)phenyl)picol inate (60 mg, 1 equiv., 81 μmol) and 3-(1,5-dimethyl-6-(piperazin-1-yl)-1H-indazol-3-yl)piperidin e-2,6-dione, HCl (33 mg, 1.1 equiv., 89 μmol) were suspended in DCM (5 mL). To this mixture was added sodium triacetoxyborohydride (51 mg, 3 equiv., 0.24 mmol). The mixture was then stirred at room temperature overnight. The mixture was then partitioned between DCM and water. The organic layer was washed with brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness to afford to tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-1,5-dimethyl- 1H-indazol-6-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate, which was carried forward without further purification. MS (ESI) m/z: 535.9 [M+2H] 2+ . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1,5- dimethyl-1H-indazol-6-yl)piperazin-1-yl)butyl)cyclohexyl)oxy )-2-methylphenyl)picolinic acid tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-1,5-dimethyl- 1H-indazol-6-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (87 mg, 1 equiv., 81 μmol) was dissolved in DCM (4 mL). To this mixture was added TFA (1 mL). The mixture was stirred at room temperature overnight. The mixture was then concentrated, and purified by reverse phase- HPLC to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-1,5-dimethyl- 1H-indazol-6-yl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (54 mg, 53 μmol, 66 %) as a white solid. MS (ESI) m/z: 1014.5 [M+H] + ; 1 H NMR (400 MHz, DMSO) δ 10.77 (s, 1H), 7.96 (dd, J = 8.0, 1.2 Hz, 1H), 7.72 (d, J = 8.0 Hz, 1H), 7.55 (dd, J = 7.6, 1.4 Hz, 1H), 7.44 – 7.33 (m, 4H), 7.33 – 7.23 (m, 2H), 7.06 (s, 1H), 7.00 (t, J = 7.9 Hz, 1H), 6.87 (dd, J = 14.8, 8.6 Hz, 2H), 6.55 (d, J = 7.4 Hz, 1H), 4.91 (s, 2H), 4.19 (dd, J = 9.5, 5.1 Hz, 1H), 4.11 (d, J = 10.6 Hz, 1H), 3.85 (s, 3H), 3.88 – 3.81 (m, 1H), 2.96 (t, J = 6.0 Hz, 2H), 2.85 (s, 3H), 2.85 (d, J = 9.8 Hz, 1H), 2.61 – 2.45 (m, 2H), 2.50 (s, 4H), 2.35 – 2.20 (m, 3H), 2.24 (s, 3H), 2.12 – 1.98 (m, 1H), 2.02 (s, 2H), 1.80 (s, 3H), 1.72 (d, J = 12.4 Hz, 2H), 1.40 (s, 2H), 1.27 (s, 5H), 1.17 (d, J = 6.4 Hz, 3H), 1.02 (d, J = 11.8 Hz, 1H), 0.96 (d, J = 12.1 Hz, 1H). Example 278. Preparation of Compound 326a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- (4-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)ox y)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of 6-bromo-3-iodo-1-methyl-indazole To a solution of 6-bromo-3-iodo-1H-indazole (30 g, 92.90 mmol, 1 equiv.) in DMF (200 mL) was added NaH (7.43 g, 185.80 mmol, 60% purity, 2 equiv.) at 0 °C. The mixture was stirred for 1 hour. After stirring, MeI (26.37 g, 185.80 mmol, 11.57 mL, 2 equiv.) was added into the solution. The mixture was stirred at 20 °C for 1 hour. After completion, the solution was cooled to 0 °C, and then saturated NH 4 Cl solution (300 mL) was added dropwise. The solution was extracted with ethyl acetate (500 mL × 3). The combined organic layers were washed with brine (500 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by reverse-phase HPLC to give 6-bromo- 3-iodo-1-methyl-indazole (15.3 g, 45.41 mmol, 48.88% yield) as a brown solid. MS (ESI) m/z: 336.8 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 8.02 (d, J = 0.6 Hz, 1H), 7.37 - 7.28 (m, 2H), 4.04 (s, 3H) Step B. Procedure for preparation of 6-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl- indazole A mixture of 6-bromo-3-iodo-1-methyl-indazole (14.8 g, 43.92 mmol, 1 equiv.), 2,6- dibenzyloxy-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)p yridine (18.33 g, 43.92 mmol, 1 equiv.), Pd(dppf)Cl2 (3.21 g, 4.39 mmol, 0.1 equiv.), Cs2CO3 (42.93 g, 131.77 mmol, 3 equiv.), and H 2 O (10 mL) in THF (100 mL) was degassed and purged with N 2 three times. The mixture was stirred at 60 °C for 16 hours under N2 atmosphere. After completion, the reaction mixture was poured into water (200 mL) and extracted with ethyl acetate (150 mL × 3). The combined organic layers were washed with brine (200 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (petroleum ether/ethyl acetate = 100/1 to 20/1) to give 6-bromo-3-(2,6- dibenzyloxy-3-pyridyl)-1-methyl-indazole (14 g, 27.98 mmol, 63.70% yield) as a yellow solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.94 (d, J = 1.2 Hz, 1H), 7.91 (d, J = 8.0 Hz, 1H), 7.61 (d, J = 8.4 Hz, 1H), 7.51 - 7.44 (m, 2H), 7.42 - 7.23 (m, 8H), 7.10 (dd, J = 1.6, 8.8 Hz, 1H), 6.59 (d, J = 8.0 Hz, 1H), 5.44 (d, J = 8.4 Hz, 4H), 4.04 (s, 3H) Step C. Procedure for preparation of 3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6- ol A mixture of 6-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (13 g, 25.98 mmol, 1 equiv.), H 2 O (936.07 mg, 51.96 mmol, 936.07 µL, 2 equiv.), NaO t Bu (7.49 g, 77.94 mmol, 3 equiv.), [2-(2-aminophenyl)phenyl]-methylsulfonyloxy-palladium;ditert -butyl-[3,6- dimethoxy-2-(2,4,6-triisopropylphenyl)phenyl]phosphane (22.20 g, 25.98 mmol, 1 equiv.) in THF (100 mL) was degassed and purged with N 2 three times. The mixture was stirred at 80 °C for 16 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (petroleum ether/ethyl acetate = 20/1 to 1/1) to give 3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-ol (11 g, 16.77 mmol, 64.55% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 9.65 (s, 1H), 7.87 (d, J = 8.0 Hz, 1H), 7.50 - 7.44 (m, 3H), 7.43 - 7.27 (m, 8H), 6.74 (d, J = 2.0 Hz, 1H), 6.59 - 6.52 (m, 2H), 5.43 (d, J = 9.2 Hz, 4H), 3.91 (s, 3H) Step D. Procedure for preparation of tert-butyl 4-[3-(2,6-dibenzyloxy-3-pyridyl)-1- methyl-indazol-6-yl]oxypiperidine-1-carboxylate A mixture of 3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-ol (5.5 g, 12.57 mmol, 1 equiv.), tert-butyl 4-hydroxypiperidine-1-carboxylate (5.06 g, 25.14 mmol, 2 equiv.) and 2- (tributyl-λ 5 -phosphanylidene)acetonitrile (3.34 g, 13.83 mmol, 1.1 equiv.) in toluene (100 mL) was degassed and purged with N 2 three times. The mixture was stirred at 110 °C for 16 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (petroleum ether/ethyl acetate = 20/1 to 2/1) to give tert-butyl 4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6- yl]oxypiperidine-1-carboxylate (16 g, crude) as a yellow oil). 1 H NMR (400 MHz, DMSO-d6) δ = 7.89 (d, J = 8.0 Hz, 1H), 7.54 (d, J = 8.8 Hz, 1H), 7.50 - 7.45 (m, 2H), 7.43 - 7.24 (m, 8H), 7.13 (d, J = 1.6 Hz, 1H), 6.66 (dd, J = 2.0, 8.8 Hz, 1H), 6.58 (d, J = 8.0 Hz, 1H), 5.48 - 5.40 (m, 4H), 4.70 - 4.64 (m, 1H), 4.00 (s, 3H), 3.30 - 3.15 (m, 2H), 2.95 (s, 3H), 2.03 - 1.92 (m, 2H), 1.60 - 1.53 (m, 2H), 1.39 (s, 9H) Step E. Procedure for preparation of tert-butyl 4-[3-(2,6-dioxo-3-piperidyl)-1-methyl- indazol-6-yl]oxypiperidine-1-carboxylate A mixture of tert-butyl 4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6- yl]oxypiperidine-1-carboxylate (16 g, 25.78 mmol, 1 equiv.), Pd/C (3 g, 25.78 mmol, 10% purity, 1 equiv.), AcOH (1.55 g, 25.78 mmol, 1.48 mL, 1 equiv.), Pd(OH)2 (3 g, 2.14 mmol, 10% purity, 0.0829 equiv.), and EtOH (100 mL) in THF (100 mL) was degassed and purged with H 2 (50 Psi) three times. The mixture was stirred at 80 °C for 16 hours under H 2 (50 Psi) atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by reversed-phase HPLC (with 0.1% TFA) to give tert- butyl 4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]oxypiperi dine-1-carboxylate (3.8 g, 8.59 mmol, 33.32% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.87 (s, 1H), 7.57 (d, J = 8.8 Hz, 1H), 7.13 (d, J = 1.6 Hz, 1H), 6.77 (dd, J = 1.6, 8.8 Hz, 1H), 4.74 - 4.65 (m, 1H), 4.30 (dd, J = 5.2, 9.6 Hz, 1H), 3.93 (s, 3H), 3.74 - 3.63 (m, 2H), 3.29 - 3.16 (m, 2H), 2.71 - 2.56 (m, 2H), 2.39 - 2.27 (m, 1H), 2.21 - 2.12 (m, 1H), 2.02 - 1.92 (m, 2H), 1.64 - 1.52 (m, 2H), 1.42 (s, 9H) Step F. Procedure for preparation of 3-[1-methyl-6-(4-piperidyloxy)indazol-3- yl]piperidine-2,6-dione To a solution of tert-butyl 4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]oxypiperi dine- 1-carboxylate (3.8 g, 8.59 mmol, 1 equiv.) in HCl/dioxane (30 mL) DCM (10 mL) was stirred at 25 °C for 1 hour. The reaction mixture was filtered to give a filter cake. The filter cake was triturated with MeCN at 25 °C for 10 minutes to give 3-[1-methyl-6-(4-piperidyloxy)indazol- 3-yl]piperidine-2,6-dione (2.20 g, 5.82 mmol, 67.74% yield) as a white solid. MS (ESI) m/z: 343.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.87 (s, 1H), 9.39 (s, 2H), 7.59 (d, J = 8.8 Hz, 1H), 7.21 (d, J = 1.6 Hz, 1H), 6.79 (dd, J = 2.0, 8.8 Hz, 1H), 6.02 (s, 4H), 4.88 - 4.73 (m, 1H), 4.31 (dd, J = 5.2, 9.6 Hz, 1H), 3.97 - 3.91 (m, 3H), 3.22 (s, 2H), 3.12 - 3.01 (m, 2H), 2.72 - 2.54 (m, 2H), 2.35 - 2.26 (m, 1H), 2.22 - 2.11 (m, 3H), 1.97 - 1.87 (m, 2H). Step G. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-((3-(2,6 -dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)oxy)piperidin-1-yl)butyl)cyclohexyl)o xy)-2- methylphenyl)picolinate tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)oxy)phenyl)picol inate (60 mg, 1 equiv., 81 μmol) and 3-[1-methyl-6-(4-piperidyloxy)indazol-3-yl]piperidine-2,6-di one, HCl (34 mg, 1.1 equiv., 89 μmol) were suspended in DCM (5 mL). To this mixture was added sodium triacetoxyborohydride (51 mg, 3 equiv., 0.24 mmol). The mixture was then stirred at room temperature overnight. The mixture was then partitioned between DCM and water. The organic layer was washed with brine, dried over Na2SO4, filtered, and evaporated to dryness to afford to tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(4-(4-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)oxy)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate, which was carried forward without further purification. MS (ESI) m/z: 536.4 [M+2H] 2+ . Step H. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-((3-(2,6 -dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)oxy)piperidin-1-yl)butyl)cyclohexyl)o xy)-2- methylphenyl)picolinic acid tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(4-(4-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)oxy)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (87 mg, 1 equiv., 81 μmol) was dissolved in DCM (4 mL). To the mixture was added TFA (1 mL). The mixture was stirred at room temperature overnight. The mixture was then concentrated, and purified by reverse phase- HPLC to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- (3-(((1s,4r)-4-(4-(4-((3-(2,6-dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)oxy)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (46 mg, 45 μmol, 56 %) as a white solid. MS (ESI) m/z: 1015.5 [M+H] + ; 1 H NMR (400 MHz, DMSO) δ 10.78 (s, 1H), 7.96 (d, J = 8.1 Hz, 1H), 7.72 (d, J = 8.0 Hz, 1H), 7.56 (d, J = 7.4 Hz, 1H), 7.48 (d, J = 8.8 Hz, 1H), 7.44 – 7.34 (m, 3H), 7.34 – 7.23 (m, 2H), 7.04 – 6.96 (m, 2H), 6.86 (dd, J = 13.8, 8.6 Hz, 2H), 6.67 (dd, J = 8.8, 2.1 Hz, 1H), 6.55 (d, J = 7.5 Hz, 1H), 4.91 (s, 2H), 4.45 (s, 1H), 4.22 (dd, J = 9.4, 5.1 Hz, 1H), 4.12 (s, 1H), 3.85 (s, 4H), 3.88 – 3.81 (m, 1H), 2.96 (t, J = 6.0 Hz, 2H), 2.66 (s, 2H), 2.60 – 2.43 (m, 2H), 2.28 (d, J = 7.7 Hz, 1H), 2.22 (s, 1H), 2.09 (dd, J = 13.2, 5.8 Hz, 1H), 2.00 (q, J = 4.4 Hz, 2H), 1.80 (s, 3H), 1.71 (d, J = 12.4 Hz, 2H), 1.60 (d, J = 10.1 Hz, 3H), 1.35 (d, J = 7.1 Hz, 2H), 1.22 (s, 8H), 1.16 (s, 1H), 0.97 (q, J = 12.3 Hz, 2H). Example 279. Preparation of Compound 327a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- (4-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)ox y)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of 7-bromo-3-iodo-1-methyl-indazole To a solution of 7-bromo-3-iodo-1H-indazole (20 g, 61.93 mmol, 1.0 equiv.) in THF (50 mL) was added t-BuOK (13.90 g, 123.87 mmol, 2.0 equiv.) at 0 °C for 1 hour. Then CH3I (26.37 g, 185.80 mmol, 11.57 mL, 3.0 equiv.) was added to the mixture at 0 °C. The mixture was stirred at 20 °C for 2 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by reverse-phase HPLC to give 7-bromo-3-iodo-1-methyl-indazole (15 g, 42.29 mmol, 54.63 % yield) as a yellow solid. MS (ESI) m/z: 336.6 [M+H] + Step B. Procedure for preparation of 7-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl- indazole A mixture of 2,6-dibenzyloxy-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)pyridine (17.34 g, 41.55 mmol, 1.0 equiv.), 7-bromo-3-iodo-1-methyl-indazole (14 g, 41.55 mmol, 1.0 equiv.), [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II ) (6.79 g, 8.31 mmol, 0.2 equiv.), and Cs 2 CO 3 (40.61 g, 124.65 mmol, 3.0 equiv.) in THF (200 mL) and H 2 O (40 mL) was degassed and purged with N2 three times. The mixture was stirred at 85 °C for 1 hour under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (petroleum ether / ethyl acetate = 1/0 to 10/1) to give 7-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (15.1 g, 28.06 mmol, 63.04 % yield) as a white solid. MS (ESI) m/z: 502.3 [M+H] + Step C. Procedure for preparation of 3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-7- ol A solution of 7-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (15 g, 29.98 mmol, 1.0 equiv.) in THF (300 mL) was added NaOt-Bu (8.64 g, 89.93 mmol, 3.0 equiv.), [(2-di-tert- butylphosphino-3,6-dimethoxy-2′,4′,6′-triisopropyl-1,1 ′-biphenyl)-2-(2′-amino-1,1′- biphenyl)]palladium(II) methanesulfonate (2.56 g, 3.00 mmol, 0.1 equiv.), and H2O (1.08 g, 59.95 mmol, 1.08 mL, 2.0 equiv.) was stirred at 60 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (petroleum ether / ethyl acetate = 1 / 0 to 4 / 1) to give 3-(2,6-dibenzyloxy-3- pyridyl)-1-methyl-indazol-7-ol (5.6 g, 11.52 mmol, 38.43 % yield) as a brown oil. MS (ESI) m/z: 438.6 [M+H] + Step D. Procedure for preparation of tert-butyl 4-((3-(2,6-bis(benzyloxy)pyridin-3-yl)-1- methyl-1H-indazol-7-yl)oxy)piperidine-1-carboxylate A solution of 3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-7-ol (2.8 g, 6.40 mmol, 1.0 equiv.) in toluene (100 mL) was added 2-(tributyl-λ 5 -phosphanylidene) acetonitrile (3.09 g, 12.80 mmol, 2.0 equiv.) and tert-butyl 4-hydroxypiperidine-1-carboxylate (1.55 g, 7.68 mmol, 1.2 equiv.). The reaction mixture was stirred at 110 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (petroleum ether / ethyl acetate = 20 / 1 to 5 / 1) to give tert-butyl 4-((3-(2,6- bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-7-yl)oxy)pip eridine-1-carboxylate (4.5 g, 6.52 mmol, 50.97 % yield) as a yellow solid. MS (ESI) m/z: 621.6 [M+H] + Step E. Procedure for preparation of tert-butyl 4-((3-(2,6-dioxopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)oxy)piperidine-1-carboxylate To a solution of tert-butyl 4-((3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-7 - yl)oxy)piperidine-1-carboxylate (4.5 g, 7.25 mmol, 1.0 equiv.) in EtOH (50 mL) and THF (50 mL) were added AcOH (870.66 mg, 14.50 mmol, 829.20 µL, 2.0 equiv.), Pd(OH)2 (3 g, 4.27 mmol, 20 % purity, 0.589 equiv.), and Pd/C (3 g, 3.62 mmol, 10 % purity, 0.5 equiv.) under N2 atmosphere. The suspension was degassed and purged with H 2 three times. The mixture was stirred under H2 (50 Psi) at 50 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product was purified by reverse-phase HPLC to give tert-butyl 4-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)oxy )piperidine-1- carboxylate (2.58 g, 5.71 mmol, 78.81 % yield) as a white solid. MS (ESI) m/z: 443.0 [M+H] + Step F. Procedure for preparation of 3-(1-methyl-7-(piperidin-4-yloxy)-1H-indazol-3- yl)piperidine-2,6-dione, HCl To a solution of tert-butyl 4-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7- yl)oxy)piperidine-1-carboxylate (2.58 g, 5.82 mmol, 1.0 eq) in DCM (20 mL) was added HCl/dioxane (4 M, 1.46 mL, 1.0 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give 3-(1-methyl-7- (piperidin-4-yloxy)-1H-indazol-3-yl)piperidine-2,6-dione, HCl (1.01 g, 2.49 mmol, 42.74 % yield) as a yellow solid which was used into next steps without further purification. MS (ESI) m/z: 343.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.87 (s, 1H), 9.54 (s, 1H), 9.32 (s, 1H), 7.24 (d, J = 7.6 Hz, 1H), 7.02 - 6.90 (m, 2H), 4.92 - 4.83 (m, 1H), 4.33 (dd, J = 5.2, 9.8 Hz, 1H), 4.19 (s, 3H), 3.20 (s, 2H), 3.12 (s, 2H), 2.72 - 2.55 (m, 2H), 2.34 - 2.13 (m, 4H), 2.07 - 1.98 (m, 2H) Step G. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-((3-(2,6 -dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)oxy)piperidin-1-yl)butyl)cyclohexyl)o xy)-2- methylphenyl)picolinate tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)oxy)phenyl)picol inate (60 mg, 1 equiv., 81 μmol) and 3-(1-methyl-7-(piperidin-4-yloxy)-1H-indazol-3-yl)piperidine -2,6-dione, HCl (34 mg, 1.1 equiv., 89 μmol) were suspended in DCM (5 mL). To this mixture was added sodium triacetoxyborohydride (51 mg, 3 equiv., 0.24 mmol). The mixture was then stirred at room temperature overnight. The mixture was then partitioned between DCM and water. The organic layer was washed with brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness to afford to tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(4-(4-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H- indazol-7-yl)oxy)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate, which was carried forward without further purification. MS (ESI) m/z: 536.3 [M+2H] 2+ . Step H. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-((3-(2,6 -dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)oxy)piperidin-1-yl)butyl)cyclohexyl)o xy)-2- methylphenyl)picolinic acid tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(4-(4-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H- indazol-7-yl)oxy)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (87 mg, 1 equiv., 81 μmol) was dissolved in DCM (4 mL). To this mixture was added TFA (1 mL). The mixture was stirred at room temperature overnight. The mixture was then concentrated, and purified by reverse phase- HPLC to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(4-(4-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H- indazol-7-yl)oxy)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (16 mg, 16 μmol, 19 %) as a white solid. MS (ESI) m/z: 1015.4 [M+H] + ; 1 H NMR (400 MHz, DMSO d6 ) δ 10.80 (s, 1H), 7.96 (d, J = 8.0 Hz, 1H), 7.72 (d, J = 8.1 Hz, 1H), 7.55 (d, J = 7.5 Hz, 1H), 7.44 – 7.34 (m, 3H), 7.34 – 7.23 (m, 2H), 7.14 (d, J = 8.0 Hz, 1H), 7.00 (t, J = 7.9 Hz, 1H), 6.95 – 6.78 (m, 4H), 6.54 (d, J = 7.5 Hz, 1H), 4.91 (s, 2H), 4.59 (s, 1H), 4.24 (dd, J = 9.8, 5.1 Hz, 1H), 4.13 (s, 3H), 3.85 (t, J = 5.9 Hz, 2H), 2.96 (t, J = 6.0 Hz, 2H), 2.60 – 2.45 (m, 1H), 2.13 – 1.98 (m, 1H), 1.99 (s, 3H), 1.80 (s, 3H), 1.71 (d, J = 12.8 Hz, 2H), 1.39 (s, 2H), 1.23 (s, 7H), 1.16 (s, 1H), 0.98 (q, J = 11.8 Hz, 2H). Example 280. Preparation of Compound 301a

6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3- ((3R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)-3-(trifluoromethyl)piperazin- 1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1s,4r)-4-(3-((3R)-4-(3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-6-yl)-3- (trifluoromethyl)piperazin-1-yl)propyl)cyclohexyl)oxy)-2-met hylphenyl)picolinate To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2-methyl-3-(((1r,4r)-4-(3-oxopropyl)cyclohexyl) oxy)phenyl)picolinate (100 mg, 1 equiv., 137 μmol) and 3-(1-methyl-6-((R)-2-(trifluoromethyl)piperazin-1-yl)-1H-ind azol-3- yl)piperidine-2,6-dione (59.5 mg, 1.1 equiv., 150 μmol) in DCM (2.00 mL) was added sodium triacetoxyborohydride (87.0 mg, 3 equiv., 410 μmol). The reaction mixture was stirred at room temperature for 18 h. The reaction mixture was diluted with DCM (75 mL), water (10 mL), and brine (10 mL). The organic layer was separated, dried over anhydrous sodium sulfate, and concentrated to provide crude tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-((3R)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-3-(trifluoromethyl)piperazin-1-yl)pr opyl)cyclohexyl)oxy)-2- methylphenyl)picolinate (122 mg, 110 μmol, 80 %yield), which was carried forward without further purification. MS (ESI) m/z: 1110.6 [M+H] + . Step B. 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(3-((3R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl -1H-indazol-6-yl)-3- (trifluoromethyl)piperazin-1-yl)propyl)cyclohexyl)oxy)-2-met hylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3-((3R)-4-(3-(2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-6- yl)-3-(trifluoromethyl)piperazin-1-yl)propyl)cyclohexyl)oxy) -2-methylphenyl)picolinate (122 mg, 1 equiv., 110 μmol) in DCM (10.00 mL) was added trifluoroacetic acid (5 mL). The reaction solution was stirred at room temperature for 48 h. The reaction solution was then concentrated and purified by reverse-phase HPLC to provide 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r) -4-(3-((3R)-4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)-3-(trifluorom ethyl)piperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (64 mg, 61 μmol, 55 % yield). MS (ESI) m/z: 1054.4 [M+H] + Example 281. Preparation of Compound 301b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(3- ((3S)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)-3-(trifluoromethyl)piperazin- 1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1r,4r)-4-(3-((3S)-4-(3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-6-yl)-3- (trifluoromethyl)piperazin-1-yl)propyl)cyclohexyl)oxy)-2-met hylphenyl)picolinate To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2-methyl-3-(((1r,4r)-4-(3-oxopropyl)cyclohexyl) oxy)phenyl)picolinate (50 mg, 1 equiv., 68 μmol) and 3-(1-methyl-6-((S)-2-(trifluoromethyl)piperazin-1-yl)-1H-ind azol-3- yl)piperidine-2,6-dione (30 mg, 1.1 equiv., 75 μmol) in DCM (2.00 mL) was added sodium triacetoxyborohydride (43 mg, 3 equiv., 0.21 mmol). The reaction solution was stirred at room temperature for 72 h. The reaction solution was diluted with DCM (75 mL) and water (10 mL). The organic layer was separated, washed with brine (10 mL), dried over anhydrous sodium sulfate and concentrated to provide crude tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-((3S)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-3-(trifluoromethyl)piperazin-1-yl)pr opyl)cyclohexyl)oxy)-2- methylphenyl)picolinate (84 mg), which was carried forward without further purification. MS (ESI): m/z 1110.6 [M+H] + . Step B. 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1r,4r)-4-(3-((3S)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl -1H-indazol-6-yl)-3- (trifluoromethyl)piperazin-1-yl)propyl)cyclohexyl)oxy)-2-met hylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(3-((3S)-4-(3-(2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-6-yl)- 3-(trifluoromethyl)piperazin-1-yl)propyl)cyclohexyl)oxy)-2-m ethylphenyl)picolinate (84 mg, 1 equiv., 76 μmol) DCM (10.00 mL) was added trifluoroacetic acid (4.3 g, 2.9 mL, 500 equiv., 38 mmol). The reaction solution was stirred at room temperature for 18 h. The reaction solution was concentrated and purified by reverse-phase HPLC to provide 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r) -4-(3-((3S)-4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)-3-(trifluorom ethyl)piperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (20 mg, 19 μmol, 25% yield). MS (ESI): m/z 1054.6 [M+H] + Example 282. Preparation of Compound 339a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- ((3R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)-3- (trifluoromethyl)piperazin-1-yl)butyl)cyclohexyl)oxy)-2-meth ylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl (3R)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1- methyl-indazol-6-yl]-3-(trifluoromethyl)piperazine-1-carboxy late A mixture of 6-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (1.97 g, 3.93 mmol, 2.0 equiv.), tert-butyl (3R)-3-(trifluoromethyl)piperazine-1-carboxylate (500 mg, 1.97 mmol, 1.0 equiv.), Pd2(dba)3 (180.08 mg, 196.66 μmol, 0.1 equiv.), NaOtBu (377.99 mg, 3.93 mmol, 2.0 equiv.), and tritert-butylphosphane (795.74 mg, 393.32 μmol, 923.14 μL, 10% purity, 0.2 equiv.) in toluene (25 mL) was degassed and purged with N 2 three times. The mixture was stirred at 100 °C for 16 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~30% ethyl acetate/petroleum ether) to afford tert-butyl (3R)-4-[3-(2,6- dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-yl]-3-(trifluorome thyl)piperazine-1-carboxylate (1.4 g, 1.91 mmol, 97.21% yield) as a yellow oil. MS (ESI) m/z: 674.3 [M+H] + . Step B. Procedure for preparation of tert-butyl (3R)-4-[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]-3-(trifluoromethyl)piperazine-1-carboxy late To a solution of tert-butyl (3R)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-yl] -3- (trifluoromethyl)piperazine-1-carboxylate (1.4 g, 2.08 mmol, 1.0 equiv.) in THF (10 mL) and EtOH (10 mL) was added Pd/C (653.33 mg, 613.92 μmol, 10% purity, 0.295 equiv.) and Pd(OH)2 (653.33 mg, 4.65 mmol, 2.24 equiv.) under N2 atmosphere. The suspension was degassed and purged with H 2 three times. The mixture was stirred under H 2 (15 Psi) at 40 °C for 16 hours. The reaction mixture was filtered and washed with THF (100 mL). The filtrate was concentrated to afford tert-butyl (3R)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6- yl]-3-(trifluoromethyl)piperazine-1-carboxylate (1.3 g, crude) as a brown solid, which was used into the next step without further purification. MS (ESI) m/z: 496.3 [M+H] + . Step C. Procedure for preparation of 3-[1-methyl-6-[(2R)-2-(trifluoromethyl)piperazin-1- yl]indazol-3-yl]piperidine-2,6-dione A solution of tert-butyl (3R)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]-3- (trifluoromethyl)piperazine-1-carboxylate (1.3 g, 2.62 mmol, 1.0 equiv.) in HCl/dioxane (20 mL) was stirred at 25 °C for 1 hour. The reaction mixture was filtered to afford 3-[1-methyl-6- [(2R)-2-(trifluoromethyl)piperazin-1-yl]indazol-3-yl]piperid ine-2,6-dione (900 mg, crude) as a white solid, which was used into next step without further purification. MS (ESI) m/z: 396.1 [M+H] + . Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-((3R)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-3-(trifluoromethyl)piperazin-1-yl)bu tyl)cyclohexyl)oxy)-2- methylphenyl)picolinate To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[2-methyl-3-[4-(4-oxobutyl)cyclohexoxy]phenyl]pyridi ne-2-carboxylate (200 mg, 268.48 μmol, 1.0 equiv.) and 3-[1-methyl-6-[(2R)-2-(trifluoromethyl)piperazin-1-yl]indazo l-3- yl]piperidine-2,6-dione (159.23 mg, 402.72 μmol, 1.5 equiv.) in DCM (5 mL), was added NaBH(OAc) 3 (170.70 mg, 805.44 μmol, 3.0 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was diluted with H 2 O (20 mL) and extracted with ethyl acetate (20 mL × 3). The combined organic layers were washed with brine (30 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to afford tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r) -4-(4-((3R)-4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)-3-(trifluorom ethyl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (300 mg, crude) as a yellow solid, which was used into next step without further purification. MS (ESI) m/z: 1124.6 [M+H] + . Step E. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-((3R)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-3-(trifluoromethyl)piperazin-1-yl)bu tyl)cyclohexyl)oxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4-((3R)-4-(3-(2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-6- yl)-3-(trifluoromethyl)piperazin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinate (300 mg, 266.83 μmol, 1.0 equiv.) in DCM (1 mL) was added TFA (4.60 g, 40.39 mmol, 3.00 mL, 151.36 equiv.). The mixture was stirred at 30 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(4-((3R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl -1H-indazol-6-yl)-3- (trifluoromethyl)piperazin-1-yl)butyl)cyclohexyl)oxy)-2-meth ylphenyl)picolinic acid (118.19 mg, 109.27 μmol, 40.95% yield) as a white solid. MS (ESI) m/z: 1068.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.86 (s, 1H), 12.64 - 12.52 (m, 1H), 10.85 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.54 - 7.41 (m, 4H), 7.40 - 7.32 (m, 2H), 7.11 - 6.88 (m, 5H), 6.61 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.34 - 4.15 (m, 2H), 3.95 - 3.87 (m, 5H), 3.49 - 3.39 (m, 2H), 3.23 - 3.16 (m, 1H), 3.02 (t, J = 6.0 Hz, 2H), 2.99 - 2.90 (m, 1H), 2.71 - 2.53 (m, 3H), 2.44 - 2.21 (m, 4H), 2.20 - 1.98 (m, 4H), 1.93 - 1.86 (m, 3H), 1.78 (d, J = 12.0 Hz, 2H), 1.50 - 1.20 (m, 9H), 1.05 (d, J = 12.4 Hz, 2H) Example 283. Preparation of Compound 340a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1-yl)-3- methylbutyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of 1-(4-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1-yl)-3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propan-1-one A mixture of 3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propanoic acid (800 mg, 2.34 mmol, 1.0 equiv.), 3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-6-piperazin-1-yl-inda zole (1.42 g, 2.81 mmol, 1.2 equiv.), EDCI (674.14 mg, 3.52 mmol, 1.5 equiv.) in pyridine (10 mL) was stirred at 40 °C for 2 hours. The mixture was then concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 70% ethyl acetate / petroleum ether) to give 1-(4-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H- indazol-6-yl)piperazin-1-yl)-3-((1r,4r)-4-(3-bromo-2-methylp henoxy)cyclohexyl)propan-1- one (2.7 g, crude) as a yellow oil. MS (ESI) m/z: 828.4 [M+H] + . 1 H NMR (400 MHz, DMSO- d6) δ = 7.89 (d, J = 8.0 Hz, 1H), 7.53 (d, J = 9.2 Hz, 1H), 7.48 - 7.28 (m, 10H), 7.15 - 7.00 (m, 3H), 6.89 - 6.82 (m, 2H), 6.56 (d, J = 8.0 Hz, 1H), 5.76 - 5.74 (m, 1H), 5.46 - 5.40 (m, 4H), 3.97 (s, 3H), 3.66 - 3.58 (m, 4H), 3.24 - 3.15 (m, 4H), 2.21 (s, 2H), 1.99 (s, 3H), 1.50 - 1.26 (m, 6H), 1.22 - 1.13 (m, 4H), 1.11 - 0.98 (m, 2H) Step B. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-(4-(4-((1r,4s)- 4-(3-bromo-2-methylphenoxy)cyclohexyl)-2-methylbutan-2-yl)pi perazin-1-yl)-1-methyl- 1H-indazole A solution of 1-(4-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol -6-yl)piperazin-1- yl)-3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propan- 1-one (2.7 g, 3.26 mmol, 1.0 equiv.), and ZrCl4 (835.05 mg, 3.58 mmol, 298.23 μL, 1.1 equiv.) in THF (30 mL) was degassed and purged with N 2 three times. The mixture was stirred at -10 °C for 1 hour under N2 atmosphere. Then MeMgBr (3 M, 7.60 mL, 7.0 equiv.) was added dropwise, and the mixture was warmed to 25 °C. The resulting mixture was stirred for 1 hour. After completion, the reaction mixture was poured into saturated aq. NH 4 Cl (50 mL), and then extracted with ethyl acetate (50 mL × 3). The combined organic layers were washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 10% DCM / MeOH) to give 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-(4-(4-((1r,4s)-4-(3-bro mo-2- methylphenoxy)cyclohexyl)-2-methylbutan-2-yl)piperazin-1-yl) -1-methyl-1H-indazole (1.3 g, 1.54 mmol, 47.34% yield) as a yellow oil. MS (ESI) m/z: 844.4 [M+H] + . Step C. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-6-(4-(2- methyl-4-((1r,4s)-4-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-d ioxaborolan-2- yl)phenoxy)cyclohexyl)butan-2-yl)piperazin-1-yl)-1H-indazole A mixture of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-(4-(4-((1r,4s)-4-(3-bro mo-2- methylphenoxy)cyclohexyl)-2-methylbutan-2-yl)piperazin-1-yl) -1-methyl-1H-indazole (1.2 g, 1.42 mmol, 1.0 equiv.), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (1.08 g, 4.27 mmol, 3.0 equiv.), KOAc (209.58 mg, 2.14 mmol, 1.5 equiv.), and Pd(dppf)Cl 2 (312.51 mg, 427.10 μmol, 0.3 equiv.) in dioxane (15 mL) was degassed and purged with N2 three times. The mixture was stirred at 90 °C for 16 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 10% ethyl acetate / petroleum ether) to give 3-(2,6- bis(benzyloxy)pyridin-3-yl)-1-methyl-6-(4-(2-methyl-4-((1r,4 s)-4-(2-methyl-3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)cyclohexyl)butan -2-yl)piperazin-1-yl)-1H- indazole (1.6 g, crude) as a yellow oil. MS (ESI) m/z: 890.8 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 7.95 - 7.92 (m, 1H), 7.91 - 7.86 (m, 1H), 7.48 - 7.45 (m, 2H), 7.42 - 7.28 (m, 8H), 7.20 - 7.16 (m, 1H), 7.11 - 7.06 (m, 1H), 6.85 - 6.80 (m, 1H), 6.57 (d, J = 8.0 Hz, 1H), 5.76 - 5.74 (m, 2H), 5.47 - 5.40(m, 4H), 4.24 - 4.13 (m, 1H), 3.97 (s, 3H), 3.94 - 3.92 (m, 1H), 3.63 - 3.54 (m, 1H), 3.16 (s, 3H), 2.72 - 2.56 (m, 3H), 2.30 (s, 2H), 2.13 - 2.00 (m, 2H), 1.91 (s, 2H), 1.87 - 1.73 (m, 2H), 1.48 - 1.32 (m, 4H), 1.30 - 1.28 (m, 8H), 1.17 - 1.15 (m, 11H), 1.08 - 1.06 (m, 4H) Step D. Procedure for preparation of 3-(1-methyl-6-(4-(2-methyl-4-((1r,4s)-4-(2-methyl- 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)cyclo hexyl)butan-2- yl)piperazin-1-yl)-1H-indazol-3-yl)piperidine-2,6-dione To a solution of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-6-(4-(2-methyl-4 -((1r,4s)-4-(2- methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenox y)cyclohexyl)butan-2- yl)piperazin-1-yl)-1H-indazole (800 mg, 898.91 μmol, 1.0 equiv.) in EtOH (10 mL) was added Pd/C (400 mg, 375.87 μmol, 10% purity, 0.418 equiv.) and Pd(OH) 2 (400 mg, 569.66 μmol, 20% purity, 0.634 equiv.). The mixture was stirred at 50 °C for 16 hours. The combined organic layers were filtered and concentrated under reduced pressure to give 3-(1-methyl-6-(4-(2- methyl-4-((1r,4s)-4-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-d ioxaborolan-2- yl)phenoxy)cyclohexyl)butan-2-yl)piperazin-1-yl)-1H-indazol- 3-yl)piperidine-2,6-dione (300 mg, 421.50 μmol, 46.89% yield) as a yellow oil. MS (ESI) m/z: 712.5 [M+H] + . Step E. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1-yl)-3-methylbutyl)cyclohexyl)oxy )-2- methylphenyl)picolinate A mixture of 3-(1-methyl-6-(4-(2-methyl-4-((1r,4s)-4-(2-methyl-3-(4,4,5,5 -tetramethyl-1,3,2- dioxaborolan-2-yl)phenoxy)cyclohexyl)butan-2-yl)piperazin-1- yl)-1H-indazol-3- yl)piperidine-2,6-dione (271.87 mg, 381.98 μmol, 1.2 equiv.), tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-bromo-pyridine-2- carboxylate (180 mg, 318.31 μmol, 1 equiv.), KF (1.5 M, 636.63 μL, 3.0 equiv.), and [2-(2- aminophenyl)phenyl]palladium(1 + )bis(1-adamantyl)-butyl-phosphane methanesulfonate (46.36 mg, 63.66 μmol, 0.2 equiv.) in dioxane (0.5 mL) was degassed and purged with N 2 three times. The mixture was stirred at 100 °C for 1 hour under N 2 atmosphere. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 100% petroleum ether / ethyl acetate) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-6-yl)piperazin-1-yl)-3- methylbutyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (110 mg, 102.77 μmol, 32.29% yield) as a yellow solid. MS (ESI) m/z: 1070.6 [M+H] + . Step F. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1-yl)-3-methylbutyl)cyclohexyl)oxy )-2-methylphenyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)piperazin-1- yl)-3-methylbutyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (100 mg, 93.43 μmol, 1.0 equiv.) in TFA (1 mL) and DCM (1 mL) was stirred at 25 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-6- yl)piperazin-1-yl)-3-methylbutyl)cyclohexyl)oxy)-2-methylphe nyl)picolinic acid (28.38 mg, 27.07 μmol, 28.98% yield) as a white solid. MS (ESI) m/z: 1014.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.06 - 12.47 (m, 2H), 10.97 - 10.73 (m, 1H), 8.06 - 8.01 (m, 1H), 7.79 (d, J = 7.6 Hz, 1H), 7.65 - 7.59 (m, 1H), 7.50 - 7.34 (m, 6H), 7.10 - 7.04 (m, 1H), 6.98 - 6.91 (m, 3H), 6.88 - 6.80 (m, 1H), 6.61 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.32 - 4.14 (m, 2H), 3.95 - 3.87 (m, 5H), 3.31 - 3.29 (m, 7H), 3.05 - 3.00 (m, 2H), 2.68 - 2.65 (m, 2H), 2.35 - 2.28 (m, 2H), 2.19 - 2.04 (m, 4H), 1.87 (s, 3H), 1.84 - 1.77 (m, 2H), 1.49 - 1.22 (m, 8H), 1.12 - 1.01 (m, 6H) Example 284. Preparation of Compound 341a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s, 4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pip erazin-1-yl)-3- methylbutyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of 3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propanoic acid A mixture of ethyl 3-((1r, 4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propanoate (3 g, 8.12 mmol, 1.0 equiv.) and LiOH•H 2 O (1.70 g, 40.62 mmol, 5.0 equiv.) in THF (30 mL) and H 2 O (10 mL) was stirred at 25 °C for 16 hours. The reaction mixture was diluted with H2O (5 mL) and concentrated as a turbid liquid. Then the pH of the turbid liquid was adjusted to pH ~ 3 with 1 N HCl (5 mL). The resulting solution was extracted with DCM / MeOH (20:1) and the combined organic layers were filtered and concentrated to afford 3-((1r,4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propanoic acid (3 g, crude) as a yellow oil which was used in the next step without further purification. Step B. Procedure for preparation of 1-(4-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl- 1H-indazol-7-yl)piperazin-1-yl)-3-((1r, 4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propan-1-one A mixture of 3-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propanoic acid (1.60 g, 4.68 mmol, 1.1 equiv.), 3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-7-piperazin-1-yl-inda zole (2.15 g, 4.25 mmol, 1.0 equiv.), EDCI (1.22 g, 6.38 mmol, 1.5 equiv.) in pyridine (5 mL) was stirred at 25 °C for 2 hours. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 60% ethyl acetate / petroleum ether) to give 1-(4-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)-3-((1r, 4r)-4-(3-bromo-2- methylphenoxy)cyclohexyl)propan-1-one (1.8 g, 2.17 mmol, 51.07% yield) as a white solid. MS (ESI) m/z: 830.4 [M+H] + . Step C. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-7-(4-(4-((1r, 4s)- 4-(3-bromo-2-methylphenoxy)cyclohexyl)-2-methylbutan-2-yl)pi perazin-1-yl)-1-methyl- 1H-indazole A mixture of 1-(4-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol -7-yl)piperazin-1- yl)-3-((1r, 4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)propan-1-one (1.8 g, 2.17 mmol, 1.0 equiv.) and ZrCl4 (556.70 mg, 2.39 mmol, 198.82 μL, 1.1 equiv.) in THF (20 mL) was degassed and purged with N 2 three times. The mixture was stirred at -10 °C for 1 hour under N 2 atmosphere, followed by the addition of MeMgBr (3 M, 5.07 mL, 7 equiv.). The reaction mixture was stirred at 25 °C for 1 hour under N2 atmosphere. After completion, the reaction mixture was poured into saturated aq. NH 4 Cl (50 mL), and extracted with ethyl acetate 150 mL (50 mL × 3). The combined organic layers were washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 10% DCM / MeOH) to give 3-(2,6-bis(benzyloxy)pyridin-3-yl)-7-(4-(4-((1r, 4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2-methylbutan-2-yl)piperazin-1-yl) -1-methyl-1H-indazole (1.2 g, 1.42 mmol, 65.55% yield) as a yellow solid. MS (ESI) m/z: 844.4 [M+H] + . Step D. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-7-(4-(2- methyl-4-((1r,4s)-4-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-d ioxaborolan-2- yl)phenoxy)cyclohexyl)butan-2-yl)piperazin-1-yl)-1H-indazole A mixture of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-7-(4-(4-((1r, 4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)-2-methylbutan-2-yl)piperazin-1-yl) -1-methyl-1H-indazole (1.2 g, 1.42 mmol, 1.0 equiv.), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (1.08 g, 4.27 mmol, 3.0 equiv.), KOAc (209.58 mg, 2.14 mmol, 1.5 equiv.), Pd(dppf)Cl2 (312.51 mg, 427.10 μmol, 0.3 equiv.) in dioxane (12 mL) was degassed and purged with N2 three times. The mixture was stirred at 90 °C for 2 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 10% ethyl acetate / petroleum ether) to give 3-(2,6- bis(benzyloxy)pyridin-3-yl)-1-methyl-7-(4-(2-methyl-4-((1r,4 s)-4-(2-methyl-3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)cyclohexyl)butan -2-yl)piperazin-1-yl)-1H- indazole (1.6 g, crude) as a yellow oil. MS (ESI) m/z: 890.8 [M+H] + . Step E. Procedure for preparation of 3-(1-methyl-7-(4-(2-methyl-4-((1r,4s)-4-(2-methyl- 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)cyclo hexyl)butan-2- yl)piperazin-1-yl)-1H-indazol-3-yl)piperidine-2,6-dione A solution of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-7-(4-(2-methyl-4 -((1r,4s)-4-(2- methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenox y)cyclohexyl)butan-2- yl)piperazin-1-yl)-1H-indazole (800 mg, 898.91 μmol, 1 equiv.), Pd/C (400 mg, 375.87 μmol, 10% purity, 0.418 equiv.), and Pd(OH)2 (400 mg, 569.66 μmol, 20% purity, 0.634 equiv.) in EtOH (10 mL) was degassed and purged with N 2 three times. The mixture was stirred at 50 °C for 16 hours under N2 atmosphere. The combined organic layers were filtered and concentrated under reduced pressure to give 3-(1-methyl-7-(4-(2-methyl-4-((1r,4s)-4-(2-methyl-3-(4,4,5,5 - tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)cyclohexyl)butan -2-yl)piperazin-1-yl)-1H- indazol-3-yl)piperidine-2,6-dione (350 mg, 491.75 μmol, 54.71% yield) was obtained as a yellow oil which was used in the next step without further purification. MS (ESI) m/z: 712.5 [M+H] + . Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)piperazin-1-yl)-3-methylbutyl)cyclohexyl)oxy )-2- methylphenyl)picolinate A mixture of 3-(1-methyl-7-(4-(2-methyl-4-((1r,4s)-4-(2-methyl-3-(4,4,5,5 -tetramethyl-1,3,2- dioxaborolan-2-yl)phenoxy)cyclohexyl)butan-2-yl)piperazin-1- yl)-1H-indazol-3- yl)piperidine-2,6-dione (151.04 mg, 212.21 μmol, 1.2 equiv.), tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-bromo-pyridine-2- carboxylate (100 mg, 176.84 μmol, 1.0 equiv.), KF (1.5 M, 353.68 μL, 3.0 equiv.), and [2-(2- aminophenyl)phenyl]palladium(1 + ) bis(1-adamantyl)-butyl-phosphane methanesulfonate (25.76 mg, 35.37 μmol, 0.2 equiv.) in dioxane (2 mL) was degassed and purged with N2 three times. The mixture was stirred at 100 °C for 1 hour under N 2 atmosphere in a microwave reactor. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 100% ethyl acetate / petroleum ether) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)piperazin-1-yl)-3-methylbutyl)cyclohexyl)oxy )-2-methylphenyl)picolinate (65 mg, 60.73 μmol, 34.34% yield) as a yellow solid. MS (ESI) m/z: 1070.6 [M+H] + . Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s, 4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)-3-methylbutyl)cyclohe xyl)oxy)-2- methylphenyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-7-yl)piperazin-1- yl)-3-methylbutyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (60 mg, 56.06 μmol, 1.0 equiv.) in TFA (1 mL) and DCM (1 mL) was stirred at 25 °C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)- 3-(3-(((1s, 4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -7-yl)piperazin-1- yl)-3-methylbutyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (24.74 mg, 23.73 μmol, 42.34% yield) as a white solid. MS (ESI) m/z: 1014.6 [M+H] + . 1 H NMR (400 MHz, DMSO- d6) δ = 12.98 - 12.77 (m, 1H), 12.69 - 12.33 (m, 1H), 10.95 - 10.81 (m, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 7.6 Hz, 1H), 7.62 (d, J = 8.0 Hz, 1H), 7.49 - 7.32 (m, 6H), 7.10 - 6.90 (m, 5H), 6.61 (d, J = 7.6 Hz, 1H), 5.06 - 4.90 (m, 2H), 4.37 - 4.31 (m, 1H), 4.25 (s, 3H), 3.98 - 3.84 (m, 2H), 3.29 - 3.27 (m, 6H), 3.05 - 3.01 (m, 2H), 2.70 - 2.59 (m, 4H), 2.36 - 2.30 (m, 2H), 2.23 - 2.03 (m, 4H), 1.89 - 1.79 (m, 5H), 1.41 - 1.23 (m, 8H), 1.16 - 0.97 (m, 6H) Example 285. Preparation of Compound 342a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pip erazin-1-yl)-4- methylpentyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of 1-(4-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl- 1H-indazol-7-yl)piperazin-1-yl)-4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butan-1-one To a solution of 4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butanoic acid (380 mg, 1.07 mmol, 1 equiv.) in DMF (10 mL) was added HATU (488.04 mg, 1.28 mmol, 1.2 equiv.) and TEA (324.70 mg, 3.21 mmol, 446.63 μL, 3 equiv.). Afterwards, 3-(2,6- bis(benzyloxy)pyridin-3-yl)-1-methyl-7-(piperazin-1-yl)-1H-i ndazole (648.97 mg, 1.28 mmol, 1.2 equiv.) was added to the mixture. The mixture was stirred at 40 °C for 12 hours. The reaction mixture was quenched by addition H 2 O 20 mL. The reaction mixture was filtered, and the filter cake was washed with 10 mL of water and diluted in DCM (30 mL). The combined organic layer was dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~60% ethyl acetate/petroleum ether) to give 1-(4-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H- indazol-7-yl)piperazin-1-yl)-4-((1r,4s)-4-(3-bromo-2-methylp henoxy)cyclohexyl)butan-1- one (900 mg, crude) as a yellow solid. MS (ESI) m/z: 844.4 [M+H] + . Step B. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-7-(4-(5-((1r,4s)- 4-(3-bromo-2-methylphenoxy)cyclohexyl)-2-methylpentan-2-yl)p iperazin-1-yl)-1- methyl-1H-indazole A mixture of 1-(4-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol -7-yl)piperazin-1- yl)-4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butan-1 -one (600 mg, 711.86 μmol, 1 equiv.), and ZrCl4 (199.07 mg, 854.23 μmol, 71.10 μL, 1.2 equiv.) in THF (20 mL) was degassed and purged with N 2 three times. MeMgBr (2.5 M, 1.99 mL, 7 equiv.) was then added dropwise at -10 °C. The mixture was stirred at -10 °C for 15 min under N 2 atmosphere. After completion, the reaction mixture was poured into saturated aq. NH4Cl (40 mL), and extracted was ethyl acetate (40 mL). The combined organic layers were washed with brine (40 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~60% ethyl acetate/petroleum ether) to give 3-(2,6-bis(benzyloxy)pyridin-3-yl)-7-(4-(5-((1r,4s)-4-(3- bromo-2-methylphenoxy)cyclohexyl)-2-methylpentan-2-yl)pipera zin-1-yl)-1-methyl-1H- indazole (160 mg, 179.24 μmol, 25.18% yield) as a yellow oil. MS (ESI) m/z: 858.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 7.85 (d, J = 8.0 Hz, 1H), 7.50 - 7.43 (m, 2H), 7.43 - 7.24 (m, 9H), 7.16 - 7.11 (m, 1H), 7.09 - 7.04 (m, 1H), 7.01 (d, J = 7.8 Hz, 2H), 6.95 - 6.90 (m, 1H), 6.58 (d, J = 8.2 Hz, 1H), 5.76 (s, 1H), 5.42 (d, J = 5.6 Hz, 4H), 4.32 (s, 3H), 4.27 - 4.19 (m, 1H), 2.98 - 2.70 (m, 4H), 2.20 (s, 3H), 2.08 - 2.02 (m, 2H), 1.82 - 1.74 (m, 2H), 1.43 - 1.26 (m, 8H), 1.17 (t, J = 8.0 Hz, 4H), 1.07 - 0.99 (m, 8H). Step C. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-7-(4-(2- methyl-5-((1r,4s)-4-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-d ioxaborolan-2- yl)phenoxy)cyclohexyl)pentan-2-yl)piperazin-1-yl)-1H-indazol e A mixture of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-7-(4-(5-((1r,4s)-4-(3-bro mo-2- methylphenoxy)cyclohexyl)-2-methylpentan-2-yl)piperazin-1-yl )-1-methyl-1H-indazole (160 mg, 186.71 μmol, 1 equiv.), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (71.12 mg, 280.07 μmol, 1.5 equiv.), KOAc (91.62 mg, 933.57 μmol, 5 equiv.), and Pd(dppf)Cl 2 (27.32 mg, 37.34 μmol, 0.2 equiv.) in dioxane (3 mL) was degassed and purged with N2 three times. The mixture was stirred at 90 °C for 12 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~60% ethyl acetate/petroleum ether) to give 3- (2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-7-(4-(2-methyl-5-( (1r,4s)-4-(2-methyl-3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)cyclohexyl)penta n-2-yl)piperazin-1-yl)-1H- indazole (150 mg, 152.66 μmol, 81.76% yield) as a yellow oil. MS (ESI) m/z: 904.6 [M+H] + . Step D. Procedure for preparation of 3-(1-methyl-7-(4-(2-methyl-5-((1r,4s)-4-(2-methyl- 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)cyclo hexyl)pentan-2- yl)piperazin-1-yl)-1H-indazol-3-yl)piperidine-2,6-dione A mixture of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-7-(4-(2-methyl-5 -((1r,4s)-4-(2- methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenox y)cyclohexyl)pentan-2- yl)piperazin-1-yl)-1H-indazole (150 mg, 152.66 μmol, 1 equiv.), Pd/C (90 mg, 84.57 μmol, 10% purity, 0.554 equiv.), and Pd(OH)2 (90 mg, 128.17 μmol, 20% purity, 0.84 equiv.) in EtOH (4 mL) and THF (4 mL) was degassed and purged with H 2 three times. The mixture was stirred at 40 °C for 12 hours under H2 atmosphere. The mixture was filtered and washed with THF (50 mL), DCM (50 mL), and MeOH (20 mL). The filtrate was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , petroleum ether : ethyl acetate = 0:1) to give 3-(1-methyl-7-(4-(2-methyl-5-((1r,4s)-4-(2-methyl-3-(4,4,5,5 - tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)cyclohexyl)penta n-2-yl)piperazin-1-yl)-1H- indazol-3-yl)piperidine-2,6-dione (110 mg, 137.92 μmol, 90.35% yield) as a yellow solid. MS (ESI) m/z: 726.5 [M+H] + . Step E. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)piperazin-1-yl)-4-methylpentyl)cyclohexyl)ox y)-2- methylphenyl)picolinate A mixture of 3-(1-methyl-7-(4-(2-methyl-5-((1r,4s)-4-(2-methyl-3-(4,4,5,5 -tetramethyl-1,3,2- dioxaborolan-2-yl)phenoxy)cyclohexyl)pentan-2-yl)piperazin-1 -yl)-1H-indazol-3- yl)piperidine-2,6-dione (110 mg, 151.56 μmol, 1 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-bromopicolin ate (102.85 mg, 181.88 μmol, 1.2 equiv.) in dioxane (2 mL) and H 2 O (0.2 mL) was added KF (26.42 mg, 454.69 μmol, 10.65 μL, 3 equiv.) and Ad2nBuP Pd G3(cataCXium® A Pd G3 ) (22.08 mg, 30.31 μmol, 0.2 equiv.). After addition, the mixture was degassed and purged with N2 three times. The mixture was stirred at 100 °C for 2 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~40% ethyl acetate/petroleum ether) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1s,4r)-4-(4-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperazin -1-yl)-4- methylpentyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (83 mg, 62.00 μmol, 40.91% yield) as a yellow solid. MS (ESI) m/z: 1084.7 [M+H] + . Step F. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)-4-methylpentyl)cycloh exyl)oxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-7- yl)piperazin-1-yl)-4-methylpentyl)cyclohexyl)oxy)-2-methylph enyl)picolinate (83 mg, 76.54 μmol, 1 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.46 mmol, 1 mL, 175.88 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-7-yl)piperazin-1-yl)-4- methylpentyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (39.52 mg, 37.37 μmol, 48.83% yield) as a yellow solid. MS (ESI) m/z: 1028.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 13.17 - 12.27 (m, 2H), 10.89 (s, 1H), 8.13 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 7.8 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.52 - 7.28 (m, 6H), 7.13 - 7.00 (m, 3H), 6.94 (dd, J = 8.8, 17.6 Hz, 2H), 6.61 (d, J = 7.4 Hz, 1H), 4.98 (s, 2H), 4.34 (dd, J = 5.6, 10.0 Hz, 1H), 4.24 (s, 3H), 4.22 - 4.15 (m, 1H), 3.91 (t, J = 5.6 Hz, 2H), 3.02 (t, J = 5.2 Hz, 2H), 2.72 - 2.53 (m, 6H), 2.43 - 2.23 (m, 2H), 2.20 - 2.13 (m, 1H), 2.12 - 2.02 (m, 2H), 1.87 (s, 3H), 1.80 (d, J = 12.8 Hz, 2H), 1.65 - 0.80 (m, 20H). Example 286. Preparation of Compound 343a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((2R,4R)-1- (2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)am ino)-2-oxoethyl)-2- methylpiperidin-4-yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of (2R,4R)-4-(3-(3-bromo-2-methylphenoxy)propyl)-2- methylpiperidine A mixture of tert-butyl (2R,4R)-4-(3-(3-bromo-2-methylphenoxy)propyl)-2-methylpiperi dine- 1-carboxylate (300 mg, 703.59 μmol, 1.0 equiv.) in HCl / dioxane (5 mL) was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give (2R,4R)-4- (3-(3-bromo-2-methylphenoxy)propyl)-2-methylpiperidine (230 mg, 624.69 μmol, 88.79% yield, HCl) as a white solid. MS (ESI) m/z: 326.1 [M+H] + . Step B. Procedure for preparation of ethyl 2-((2R,4R)-4-(3-(3-bromo-2- methylphenoxy)propyl)-2-methylpiperidin-1-yl)acetate A mixture of (2R,4R)-4-(3-(3-bromo-2-methylphenoxy)propyl)-2-methylpiperi dine (230 mg, 704.94 μmol, 1.0 equiv.), ethyl 2-bromoacetate (117.72 mg, 704.94 μmol, 78.02 μL, 1.0 equiv.), and K2CO3 (292.28 mg, 2.11 mmol, 3.0 equiv.) in DMF (5 mL) was stirred at 40 °C for 0.5 hours. The reaction mixture was partitioned between H2O (5 mL) and ethyl acetate (10 mL×3). The organic phase was separated, washed with brine (15 mL × 2), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give ethyl 2-((2R,4R)-4-(3-(3-bromo-2- methylphenoxy)propyl)-2-methylpiperidin-1-yl)acetate (300 mg, crude) as a yellow solid. MS (ESI) m/z: 414.2 [M+H] + . Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((2R,4R)-1-(2-ethoxy-2- oxoethyl)-2- methylpiperidin-4-yl)propoxy)-2-methylphenyl)picolinate A mixture of ethyl 2-((2R,4R)-4-(3-(3-bromo-2-methylphenoxy)propyl)-2-methylpip eridin-1- yl)acetate (300 mg, 727.52 μmol, 1.0 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tetramethyl-1,3, 2-dioxaborolan-2-yl)picolinate (490.20 mg, 800.27 μmol, 1.1 equiv.), Ad 2 nBuP Pd G 3 (105.97 mg, 145.50 μmol, 0.2 equiv.), and KF (126.80 mg, 2.18 mmol, 51.13 μL, 3.0 equiv.) in dioxane (5 mL) and H2O (0.5 mL) was degassed and purged with N2 three times. The mixture was stirred at 100 °C for 16 hours under N 2 atmosphere. The reaction mixture was partitioned between ethyl acetate (5 mL × 3) and H2O (5 mL). The organic phase was separated, washed with brine (8 mL × 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~58% ethyl acetate / petroleum ether) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3- ((2R,4R)-1-(2-ethoxy-2-oxoethyl)-2-methylpiperidin-4-yl)prop oxy)-2- methylphenyl)picolinate (412 mg, 453.84 μmol, 62.38% yield) as a yellow solid. MS (ESI) m/z: 818.8 [M+H] + . Step D. Procedure for preparation of 2-((2R,4R)-4-(3-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)propyl)-2-methylpiperidin-1-yl)acetic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-((2R,4R)-1-(2-ethoxy-2-oxoethyl)-2-methylpiperid in-4-yl)propoxy)-2- methylphenyl)picolinate (410 mg, 501.20 μmol, 1.0 equiv.) LiOH.H 2 O (84.13 mg, 2.00 mmol, 4 equiv.) in THF (4 mL) and H2O (1 mL) was stirred at 25 °C for 19 hours. The reaction mixture was partitioned between DCM (10 mL × 3) and H2O (10 mL). The organic phase was separated, washed with brine (20 mL × 2), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give 2-((2R,4R)-4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4 - dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)propyl)-2-methylpiperidin-1-yl)acetic acid (398 mg, crude) as a yellow solid. MS (ESI) m/z: 790.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.80 (dd, J = 1.6, 6.8 Hz, 1H), 7.61 (d, J = 7.6 Hz, 1H), 7.47 - 7.36 (m, 2H), 7.27 - 7.22 (m, 1H), 7.18 - 7.14 (m, 3H), 7.10 (t, J = 7.6 Hz, 1H), 6.97 (t, J = 7.2 Hz, 1H), 6.91 (d, J = 8.8 Hz, 1H), 6.59 (d, J = 7.6 Hz, 1H), 5.15 (s, 2H), 4.02 - 3.81 (m, 4H), 3.00 - 2.95 (m, 2H), 2.84 (d, J = 10.8 Hz, 1H), 2.30 (s, 2H), 2.15 - 2.03 (m, 2H), 1.88 (s, 3H), 1.71 (d, J = 6.8 Hz, 2H), 1.62 (s, 1H), 1.53 (d, J = 12.0 Hz, 2H), 1.31 (s, 2H), 1.23 (d, J = 2.4 Hz, 2H), 1.06 (s, 9H), 0.99 (d, J = 6.0 Hz, 3H) Step E. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((2R,4R)-1-(2-((3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)-2-methylpiperidin- 4-yl)propoxy)-2- methylphenyl)picolinate A mixture of 2-((2R,4R)-4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4 - dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)propyl)-2-methylpiperidin-1-yl)acetic acid (200 mg, 253.17 μmol, 1.0 equiv.) and EDCI (72.80 mg, 379.76 μmol, 1.5 equiv.) in pyridine (2 mL) was added 3-(6-amino-1- methyl-1H-indazol-3-yl)piperidine-2,6-dione (78.47 mg, 303.81 μmol, 1.2 equiv.). The reaction mixture was stirred at 40 °C for 3 hours. The reaction mixture was concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((2R,4R)-1-(2-((3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)-2-methylpiperidin-4-yl)pr opoxy)-2- methylphenyl)picolinate (220 mg, crude) as a yellow solid. MS (ESI) m/z: 1030.7 [M+H] + Step F. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((2R,4R)-1-(2-((3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)-2-methylpiperidin- 4-yl)propoxy)-2- methylphenyl)picolinic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-((2R,4R)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)amino)-2- oxoethyl)-2-methylpiperidin-4-yl)propoxy)-2-methylphenyl)pic olinate (220 mg, 213.54 μmol, 1.0 equiv.) in TFA (3 mL) and DCM (1 mL) was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(3-((2R,4R)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)amino)-2- oxoethyl)-2-methylpiperidin-4-yl)propoxy)-2-methylphenyl)pic olinic acid (84.23 mg, 84.58 μmol, 39.61% yield) as a yellow solid. MS (ESI) m/z: 974.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.93 - 12.76 (m, 1H), 10.89 (s, 1H), 8.07 - 7.99 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.69 - 7.59 (m, 2H), 7.49 - 7.43 (m, 3H), 7.40 - 7.32 (m, 2H), 7.24 - 7.16 (m, 1H), 7.10 (t, J = 7.6 Hz, 1H), 6.97 (d, J = 8.8 Hz, 1H), 6.88 (d, J = 8.0 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.33 (dd, J = 4.8, 9.6 Hz, 1H), 3.96 (t, J = 5.6 Hz, 2H), 3.93 (s, 3H), 3.03 (t, J = 5.6 Hz, 2H), 2.69 - 2.59 (m, 4H), 2.39 - 2.32 (m, 2H), 2.20 - 2.14 (m, 1H), 1.90 (s, 3H), 1.83 - 1.68 (m, 4H), 1.40 (d, J = 2.8 Hz, 3H), 1.21 - 0.93 (m, 4H). Example 287. Preparation of Compound 343b 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[(2R,4S)-1- [2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]- 2-oxo-ethyl]-2-methyl-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl (2R,4E)-4-(3-benzyloxypropylidene)-2- methyl-piperidine-1-carboxylate A mixture of 3-benzyloxypropyl(triphenyl)phosphonium bromide (10.14 g, 20.63 mmol, 1.1 equiv.) in THF (40 mL) was added LiHMDS (1 M, 28.13 mL, 1.5 equiv.) at 0 °C under N 2 atmosphere for 1 hour. To this mixture was added tert-butyl (2R)-2-methyl-4-oxo-piperidine- 1-carboxylate (4.0 g, 18.76 mmol, 1.0 equiv.) at 0 °C under N2 atmosphere. The mixture was stirred at 25 °C for 11 hours under N2 atmosphere. The reaction mixture was quenched by the addition saturated NH 4 Cl (100 mL) and then extracted with ethyl acetate (100 mL× 3). The combined organic layers were washed with brine (90 mL × 3), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (petroleum ether/ethyl acetate = 100/1 to 50/1) to give tert-butyl (2R,4E)-4- (3-benzyloxypropylidene)-2-methyl-piperidine-1-carboxylate (3.6 g, 7.92 mmol, 42.23% yield) as a yellow oil. MS (ESI) m/z: 246.4 [M+H] + . Step B. Procedure for preparation of tert-butyl (2R)-4-(3-hydroxypropyl)-2-methyl- piperidine-1-carboxylate To a solution of tert-butyl (2R,4E)-4-(3-benzyloxypropylidene)-2-methyl-piperidine-1- carboxylate (3.6 g, 10.42 mmol, 1.0 equiv.) in EtOH (50 mL) was added Pd/C (1.11 g, 1.04 mmol, 10% purity, 0.1 equiv.) and Pd(OH)2 (1.10 g, 1.56 mmol, 20% purity, 0.15 equiv.) under N 2 atmosphere. The suspension was degassed and purged with H 2 three times. The mixture was stirred under H2 (15 Psi) at 40 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give tert-butyl (2R)-4-(3-hydroxypropyl)-2-methyl- piperidine-1-carboxylate (2.4 g, 9.33 mmol, 89.49% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 4.37 - 4.31 (m, 2H), 3.81 - 3.74 (m, 1H), 2.81 - 2.68 (m, 1H), 1.56 - 1.50 (m, 3H), 1.44 - 1.37 (m, 12H), 1.30 - 1.16 (m, 4H), 1.12 - 1.05 (m, 3H) Step C. Procedure for preparation of tert-butyl (2R)-4-[3-(3-bromo-2-methyl- phenoxy)propyl]-2-methyl-piperidine-1-carboxylate To a solution of tert-butyl (2R)-4-(3-hydroxypropyl)-2-methyl-piperidine-1-carboxylate (2.4 g, 9.33 mmol, 1.0 equiv.) in toluene (25 mL) was added 2-(tributyl- phosphanylidene)acetonitrile (3.38 g, 13.99 mmol, 1.5 equiv.) and 3-bromo-2-methyl-phenol (2.09 g, 11.19 mmol, 1.2 equiv.). The mixture was stirred at 120 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (petroleum ether/ethyl acetate=100/1 to 10/1) and reverse-phase HPLC to give tert-butyl (2R)-4-[3-(3-bromo-2-methyl-phenoxy)propyl]-2-methyl-piperid ine- 1-carboxylate (1.7 g, 3.93 mmol, 42.11% yield) as a yellow oil. MS (ESI) m/z: 326.0 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ = 7.15 (d, J = 8.0 Hz, 1H), 7.00 (t, J = 8.0 Hz, 1H), 6.76 (d, J = 8.0 Hz, 1H), 3.94 (t, J = 6.4 Hz, 3H), 2.88 - 2.82 (m, 1H), 2.32 (s, 3H), 1.85 - 1.81 (m, 2H), 1.60 (s, 4H), 1.47 (s, 9H), 1.42 - 1.35 (m, 2H), 1.34 - 1.27 (m, 1H), 1.18 (d, J = 6.4 Hz, 1H), 1.14 (d, J = 7.2 Hz, 3H) Step D. Procedure for preparation of tert-butyl (2R,4S)-4-[3-(3-bromo-2-methyl- phenoxy)propyl]-2-methyl-piperidine-1-carboxylate and tert-butyl (2R,4R)-4-[3-(3- bromo-2-methyl-phenoxy)propyl]-2-methyl-piperidine-1-carboxy late The stereoisomers of tert-butyl (2R)-4-[3-(3-bromo-2-methyl-phenoxy)propyl]-2-methyl- piperidine-1-carboxylate were separated by prep-HPLC (column: DAICEL CHIRALCEL OD(250mm × 30 mm,10 um);mobile phase: [CO 2 -i-PrOH(0.1%NH 3 H 2 O)];B%:20%, isocratic elution mode ) to give tert-butyl (2R,4S)-4-[3-(3-bromo-2-methyl-phenoxy)propyl]-2-methyl- piperidine-1-carboxylate (1 g, 2.35 mmol, 58.82% yield) as a yellow oil and tert-butyl (2R,4R)- 4-[3-(3-bromo-2-methyl-phenoxy)propyl]-2-methyl-piperidine-1 -carboxylate (300 mg, 703.59 μmol, 17.65% yield) as a yellow oil. 1 H NMR (400 MHz, CDCl3) δ = 7.07 (d, J = 8.0 Hz, 1H), 6.91 (t, J = 8.0 Hz, 1H), 6.68 (d, J = 8.0 Hz, 1H), 4.35 (s, 1H), 3.93 - 3.80 (m, 3H), 2.82 - 2.72 (m, 1H), 2.24 (s, 3H), 1.78 - 1.70 (m, 2H), 1.65 - 1.56 (m, 2H), 1.49 (d, J = 13.8 Hz, 1H), 1.39 (s, 9H), 1.34 - 1.27 (m, 2H), 1.27 - 1.19 (m, 1H), 1.06 (d, J = 7.2 Hz, 3H), 1.03 - 0.95 (m, 1H). 1 H NMR (400 MHz, CDCl 3 ) δ = 7.07 (d, J = 8.0 Hz, 1H), 6.91 (t, J = 8.0 Hz, 1H), 6.68 (d, J = 8.0 Hz, 1H), 3.86 (t, J = 6.4 Hz, 2H), 3.84 - 3.78 (m, 1H), 3.71 - 3.60 (m, 1H), 3.05 - 2.92 (m, 1H), 2.24 (s, 3H), 1.90 - 1.82 (m, 1H), 1.76 - 1.69 (m, 3H), 1.55 - 1.49 (m, 1H), 1.45 - 1.40 (m, 2H), 1.39 (s, 9H), 1.13 - 1.07 (m, 5H). Step E. Procedure for preparation of (2R,4S)-4-[3-(3-bromo-2-methyl-phenoxy)propyl]- 2-methyl-piperidine To a solution of tert-butyl (2R,4S)-4-[3-(3-bromo-2-methyl-phenoxy)propyl]-2-methyl- piperidine-1-carboxylate (400.00 mg, 938.12 μmol, 1.0 equiv.) in DCM (2 mL) was added HCl/dioxane (4 M, 4 mL, 17.06 equiv.). The mixture was stirred at 25 °C for 30 min. The reaction mixture was concentrated under reduced pressure to give (2R,4S)-4-[3-(3-bromo-2- methyl-phenoxy)propyl]-2-methyl-piperidine (300 mg, 919.48 μmol, 98.01% yield) as a white solid. MS (ESI) m/z: 326.1 [M+H] + . Step F. Procedure for preparation of ethyl 2-[(2R,4S)-4-[3-(3-bromo-2-methyl- phenoxy)propyl]-2-methyl-1-piperidyl]acetate To a solution of (2R,4S)-4-[3-(3-bromo-2-methyl-phenoxy)propyl]-2-methyl-pipe ridine (280.00 mg, 858.18 μmol, 1.0 equiv.) in acetone (3 mL) was added TEA (260.52 mg, 2.57 mmol, 358.34 μL, 3.0 equiv.) and ethyl 2-bromoacetate (143.32 mg, 858.18 μmol, 94.97 μL, 1.0 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (petroleum ether/ethyl acetate = 3/1 to 1/1) to give ethyl 2-[(2R,4S)-4-[3-(3- bromo-2-methyl-phenoxy)propyl]-2-methyl-1-piperidyl]acetate (260 mg, 624.21 μmol, 72.74% yield) as a yellow oil. MS (ESI) m/z: 412.3 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ = 7.14 (d, J = 8.0 Hz, 1H), 6.99 (t, J = 8.0 Hz, 1H), 6.76 (d, J = 8.0 Hz, 1H), 4.24 - 4.14 (m, 2H), 3.94 (t, J = 6.4 Hz, 2H), 3.40 - 3.24 (m, 2H), 3.11 (s, 1H), 2.74 (d, J = 3.6 Hz, 1H), 2.63 - 2.53 (m, 1H), 2.32 (s, 3H), 1.85 - 1.78 (m, 2H), 1.67 - 1.61 (m, 2H), 1.59 (d, J = 4.0 Hz, 2H), 1.49 - 1.40 (m, 3H), 1.28 (t, J = 7.2 Hz, 3H), 1.03 (d, J = 6.4 Hz, 3H) Step G. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(2R,4S)-1-(2-ethox y-2-oxo-ethyl)-2-methyl-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate To a solution of ethyl 2-[(2R,4S)-4-[3-(3-bromo-2-methyl-phenoxy)propyl]-2-methyl-1 - piperidyl]acetate (130.00 mg, 315.26 μmol, 1.0 equiv.) in dioxane (4 mL) and H2O (1 mL) was added KF (54.95 mg, 945.78 μmol, 22.16 μL, 3.0 equiv.), [2-(2- aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-butyl-phos phane;methanesulfonate (22.96 mg, 31.53 μmol, 0.1 equiv.), and tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)pyridine-2- carboxylate (212.42 mg, 346.78 μmol, 1.1 equiv.). The mixture was stirred at 100 °C for 1 hour. The reaction mixture was then concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (petroleum ether/ethyl acetate = 5/1 to 1/1) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[(2R,4S)-1-(2-ethoxy-2-oxo-ethyl)-2-methyl-4-pip eridyl]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (250 mg, 287.27 μmol, 91.12% yield) as a yellow oil. MS (ESI) m/z: 818.7 [M+H] + . Step H. Procedure for preparation of 2-[(2R,4S)-4-[3-[3-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-2- methyl-phenoxy]propyl]-2-methyl-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[3-[(2R,4S)-1-(2-ethoxy-2-oxo-ethyl)-2-methyl-4-p iperidyl]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (240.00 mg, 293.39 μmol, 1.0 equiv.) in THF (4 mL) and H2O (1 mL) was added LiOH•H 2 O (36.93 mg, 880.16 μmol, 3.0 equiv.). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was acidified to pH = 5 with 1M HCl and extracted with DCM (30 mL × 3). The combined organic layers were washed with brine (30 mL × 1), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give 2-[(2R,4S)-4-[3-[3-[6- [8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinol in-2-yl]-2-tert- butoxycarbonyl-3-pyridyl]-2-methyl-phenoxy]propyl]-2-methyl- 1-piperidyl]acetic acid (220 mg, 278.49 μmol, 94.92% yield) as a yellow solid. MS (ESI) m/z: 790.4 [M+H] + . Step I. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(2R,4S)-1-[2-[[3-(2,6- dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-2-methyl-4-piperidyl]propox y]-2-methyl- phenyl]pyridine-2-carboxylate To a solution of 2-[(2R,4S)-4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3, 4-dihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]propyl]-2-methyl-1- piperidyl]acetic acid (200 mg, 253.17 μmol, 1 equiv.) in pyridine (2 mL) was added EDCI (72.80 mg, 379.76 μmol, 1.5 equiv.) and 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6- dione (71.93 mg, 278.49 μmol, 1.2 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction was diluted with water (300 mL) and extracted with DCM (100 mL × 2). The combined organic layers were washed with brine (100 mL × 1), dried over Na2SO4, filtered, and concentrated under reduced pressure to give tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(2R,4 S)-1-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]-2-methy l-4-piperidyl]propoxy]-2- methyl-phenyl]pyridine-2-carboxylate (200 mg, 194.13 μmol, 76.68% yield) as a yellow solid. MS (ESI) m/z: 1030.6 [M+H] + . Step J. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[(2R,4S)-1-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-2-methyl-4-piperidyl]propoxy]-2-met hyl-phenyl]pyridine-2- carboxylic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[3-[(2R,4S)-1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-me thyl-indazol-6-yl]amino]-2-oxo- ethyl]-2-methyl-4-piperidyl]propoxy]-2-methyl-phenyl]pyridin e-2-carboxylate (200 mg, 194.13 μmol, 1.0 equiv.) in DCM (1 mL) was added TFA (3.07 g, 26.92 mmol, 2 mL, 138.69 equiv.). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was then concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3- [(2R,4S)-1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6 -yl]amino]-2-oxo-ethyl]-2- methyl-4-piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carbo xylic acid (37.85 mg, 36.17 μmol, 18.63% yield) as a white solid. MS (ESI) m/z: 974.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 13.00 - 12.54 (m, 1H), 10.86 (s, 1H), 10.01 - 9.89 (m, 1H), 8.06 - 7.99 (m, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.66 - 7.60 (m, 2H), 7.49 - 7.42 (m, 3H), 7.40 - 7.31 (m, 2H), 7.20 (d, J = 8.8 Hz, 1H), 7.09 (t, J = 8.0 Hz, 1H), 6.96 (d, J = 8.8 Hz, 1H), 6.88 (d, J = 8.4 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.36 - 4.29 (m, 1H), 3.96 (t, J = 6.4 Hz, 2H), 3.94 - 3.89 (m, 5H), 3.02 (t, J = 5.6 Hz, 2H), 2.76 - 2.52 (m, 7H), 2.37 - 2.30 (m, 1H), 2.21 - 2.14 (m, 1H), 1.90 (s, 3H), 1.79 - 1.65 (m, 4H), 1.58 (s, 2H), 1.46 - 1.38 (m, 2H), 1.34 (d, J = 8.0 Hz, 1H), 1.07 (d, J = 5.2 Hz, 3H) Example 288. Preparation of Compound 343c 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((2S,4R)-1- (2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)am ino)-2-oxoethyl)-2- methylpiperidin-4-yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of (2S,4R)-4-(3-(3-bromo-2-methylphenoxy)propyl)-2- methylpiperidine To a solution of tert-butyl (2S,4R)-4-(3-(3-bromo-2-methylphenoxy)propyl)-2- methylpiperidine-1-carboxylate (320.00 mg, 750.49 μmol, 1.0 equiv.) in DCM (3 mL) was added HCl/dioxane (4 M, 187.62 μL, 1.0 equiv.). The mixture was stirred at 25 °C for 0.5 hour. The reaction mixture was concentrated under reduced pressure to give (2S,4R)-4-(3-(3-bromo- 2-methylphenoxy)propyl)-2-methylpiperidine (200 mg, 612.99 μmol, 81.68 % yield) as a white solid, which was used in the next step without further purification. MS (ESI) m/z: 326.0 [M+H] + . Step B. Procedure for preparation of ethyl 2-((2S,4R)-4-(3-(3-bromo-2- methylphenoxy)propyl)-2-methylpiperidin-1-yl)acetate To a solution of (2S,4R)-4-(3-(3-bromo-2-methylphenoxy)propyl)-2-methylpiperi dine (150.00 mg, 459.74 μmol, 1.0 equiv.) in DMF (2 mL) was added K 2 CO 3 (190.62 mg, 1.38 mmol, 3.0 equiv.) and ethyl 2-bromoacetate (76.78 mg, 459.74 μmol, 50.88 μL, 1.0 equiv.). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was diluted with H2O (100 mL) and extracted with ethyl acetate (50 mL × 3). The combined organic layers were washed with DCM (50 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give ethyl 2-((2S,4R)-4-(3-(3- bromo-2-methylphenoxy)propyl)-2-methylpiperidin-1-yl)acetate (150 mg, 363.76 μmol, 79.12 % yield) as a yellow oil, which was used in the next step without further purification. MS (ESI) m/z: 413.1 [M+H] + . Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((2S,4R)-1-(2-ethoxy-2- oxoethyl)-2- methylpiperidin-4-yl)propoxy)-2-methylphenyl)picolinate A mixture of ethyl 2-((2S,4R)-4-(3-(3-bromo-2-methylphenoxy)propyl)-2-methylpip eridin-1- yl)acetate (150.00 mg, 363.76 μmol, 1.0 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(4,4,5,5-tet ramethyl-1,3,2-dioxaborolan-2- yl)picolinate (245.10 mg, 400.14 μmol, 1.1 equiv.), KF (63.40 mg, 1.09 mmol, 25.56 μL, 3.0 equiv.), [2-(2-aminophenyl)phenyl]palladium(1+) bis(1-adamantyl)-butyl-phosphane methanesulfonate (26.49 mg, 36.38 μmol, 0.1 equiv.), and H2O (0.5 mL) in dioxane (2 mL) was degassed and purged with N 2 three times. The mixture was stirred at 100 °C for 1 hour under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was diluted with H2O (100 mL) and extracted with ethyl acetate (50 mL × 3). The combined organic layers were washed with ethyl acetate (50 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (petroleum ether / ethyl acetate = 8 / 1 to 2 / 1) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((2S,4R)-1- (2-ethoxy-2-oxoethyl)-2-methylpiperidin-4-yl)propoxy)-2-meth ylphenyl)picolinate (290 mg, 319.06 μmol, 87.71 % yield) as a yellow solid. MS (ESI) m/z: 818.4 [M+H] + . Step D. Procedure for preparation of 2-((2S,4R)-4-(3-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)propyl)-2-methylpiperidin-1-yl)acetic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((2S,4R)-1-(2-ethoxy-2-oxoethyl)-2-methylp iperidin-4-yl)propoxy)-2- methylphenyl)picolinate (290.00 mg, 354.51 μmol, 1.0 equiv.) in THF (3 mL) was added LiOH•H2O (44.63 mg, 1.06 mmol, 3.0 equiv.) and H2O (1 mL). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was diluted with H2O (100 mL) and extracted with DCM (50 mL × 3). The combined organic layers were washed with DCM (50 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give 2-((2S,4R)-4-(3-(3-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-2- methylphenoxy)propyl)-2-methylpiperidin-1-yl)acetic acid (260 mg, 329.12 μmol, 92.84 % yield) as a yellow oil which was used in the next step without further purification. MS (ESI) m/z: 790.5 [M+H] + . Step E. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((2S,4R)-1-(2-((3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)-2-methylpiperidin- 4-yl)propoxy)-2- methylphenyl)picolinate To a solution of 2-((2S,4R)-4-(3-(3-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4 - dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-2- methylphenoxy)propyl)-2-methylpiperidin-1-yl)acetic acid (150 mg, 189.88 μmol, 1.0 equiv.) in pyridine (2 mL) was added EDCI (47.32 mg, 246.84 μmol, 1.3 equiv.) and 3-(6-amino-1- methyl-1H-indazol-3-yl)piperidine-2,6-dione (58.85 mg, 227.85 μmol, 1.2 equiv.). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was diluted with H 2 O (60 mL) and extracted with DCM (50 mL × 2). The combined organic layers were washed with DCM (30 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(3-((2S,4R)-1-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2- oxoethyl)-2-methylpiperidin- 4-yl)propoxy)-2-methylphenyl)picolinate (150 mg, 145.60 μmol, 76.68 % yield) as a yellow oil, which was used in the next step without further purification. MS (ESI) m/z: 1030.8 [M+H] + . Step F. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((2S,4R)-1-(2-((3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)-2-methylpiperidin- 4-yl)propoxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(3-((2S,4R)-1-(2-((3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-6- yl)amino)-2-oxoethyl)-2-methylpiperidin-4-yl)propoxy)-2-meth ylphenyl)picolinate (150 mg, 145.60 μmol, 1.0 equiv.) in DCM (1 mL) was added TFA (3 mL). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(3-((2S,4R)-1-(2-((3-( 2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)-2-methylpiperidin- 4-yl)propoxy)-2- methylphenyl)picolinic acid (61.41 mg, 57.56 μmol, 39.53 % yield) as a yellow solid. MS (ESI) m/z: 974.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.95 – 12.79 (m, 1H), 12.73 – 12.45 (m, 1H), 10.89 (s, 1H), 8.14 (s, 1H), 8.06 – 8.01 (m, 2H), 7.79 (d, J = 8.4 Hz, 1H), 7.65 (dd, J = 8.4, 12.8 Hz, 2H), 7.50 – 7.44 (m, 3H), 7.37 (td, J = 7.6, 11.4 Hz, 2H), 7.20 (d, J = 8.8 Hz, 1H), 7.13 – 7.08 (m, 1H), 6.98 (d, J = 8.8 Hz, 1H), 6.92 – 6.87 (m, 1H), 6.64 (d, J = 7.6 Hz, 1H), 4.99 (s, 2H), 4.34 (dd, J = 5.2, 9.6 Hz, 1H), 3.98 (t, J = 6.0 Hz, 2H), 3.94 – 3.90 (m, 5H), 3.03 (t, J = 5.6 Hz, 2H), 2.66 – 2.56 (m, 8H), 2.18 (dd, J = 5.6, 13.2 Hz, 1H), 1.91 (s, 3H), 1.81 – 1.65 (m, 5H), 1.52 – 1.35 (m, 4H), 1.26 – 1.02 (m, 3H) 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[(2S,4S)-1- [2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]- 2-oxo-ethyl]-2-methyl-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl (2S,4E)-4-(3-benzyloxypropylidene)-2- methyl-piperidine-1-carboxylate To a solution of 3-benzyloxypropyl(triphenyl)phosphoniumbromide (11.06 g, 22.51 mmol, 1.2 equiv.) in THF (100 mL) was added LiHMDS (1 M, 37.51 mL, 2.0 equiv.) at 0°C. The mixture was stirred for 2 hours. To this solution was added tert-butyl (2S)-2-methyl-4-oxo-piperidine- 1-carboxylate (4 g, 18.76 mmol, 1.0 equiv.) in THF (20 mL). The resulting mixture was stirred at 25 °C for 10 hours. The reaction mixture was quenched by addition saturated NH4Cl (50 mL) under 0 °C. The mixture was extracted with ethyl acetate (100 mL), and the combined organic layers were washed with brine (50 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~3% ethyl acetate/petroleum ether) to give tert-butyl (2S,4E)-4-(3- benzyloxypropylidene)-2-methyl-piperidine-1-carboxylate (1.44 g, 4.08 mmol, 21.78% yield) as a yellow oil. MS (ESI) m/z: 246.3 [M+H] + . Step B. Procedure for preparation of tert-butyl (2S)-4-(3-hydroxypropyl)-2-methyl- piperidine-1-carboxylate A mixture of tert-butyl (2S,4E)-4-(3-benzyloxypropylidene)-2-methyl-piperidine-1- carboxylate (1.44 g, 4.17 mmol, 1 equiv.), Pd/C (887.16 mg, 833.64 μmol, 10% purity, 0.2 equiv.), and Pd(OH) 2 (585.36 mg, 833.64 μmol, 20% purity, 0.2 equiv.) in MeOH (20 mL) was degassed and purged with H 2 three times. The mixture was stirred at 40 °C for 12 hours under H2 atmosphere. The mixture was filtered and washed with MeOH (150 mL). The resulting filtrate was concentrated under reduced pressure to give a residue. The filter cake was immersed in MeOH and concentrated under vacuum to give tert-butyl (2S)-4-(3- hydroxypropyl)-2-methyl-piperidine-1-carboxylate (1 g, crude) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 4.40 - 4.26 (m, 1H), 3.85 - 3.67 (m, 1H), 3.37 (q, J = 6.0 Hz, 2H), 3.32 (s, 1H), 2.91 - 2.63 (m, 1H), 1.67 - 1.42 (m, 4H), 1.39 (s, 9H), 1.32 - 1.22 (m, 1H), 1.20 - 1.03 (m, 6H), 0.95 - 0.78 (m, 1H) Step C. Procedure for preparation of tert-butyl (2S)-4-[3-(3-bromo-2-methyl- phenoxy)propyl]-2-methyl-piperidine-1-carboxylate A mixture of tert-butyl (2S)-4-(3-hydroxypropyl)-2-methyl-piperidine-1-carboxylate (1 g, 3.89 mmol, 1 equiv.), 3-bromo-2-methyl-phenol (872.06 mg, 4.66 mmol, 1.2 equiv.), and 2- (tributyl-phosphanylidene)acetonitrile (1.41 g, 5.83 mmol, 1.5 equiv.) in toluene (20 mL) was degassed and purged with N2 three times. The mixture was stirred at 120 °C for 12 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~2% ethyl acetate/petroleum ether) and SFC to give tert-butyl (2S)-4-[3-(3-bromo-2-methyl- phenoxy)propyl]-2-methyl-piperidine-1-carboxylate (1.4 g, 3.15 mmol, 81.09% yield) as a yellow oil. MS (ESI) m/z: 362.2 [M-100+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 7.18 - 7.13 (m, 1H), 7.12 - 7.06 (m, 1H), 6.97 (d, J = 8.0 Hz, 1H), 4.38 - 4.19 (m, 1H), 3.98 (t, J = 6.4 Hz, 2H), 3.81 (d, J = 9.2 Hz, 1H), 2.92 - 2.70 (m, 1H), 2.24 (s, 3H), 1.79 - 1.60 (m, 4H), 1.59 - 1.49 (m, 1H), 1.39 (s, 9H), 1.36 - 1.27 (m, 2H), 1.24 - 1.15 (m, 1H), 1.13 - 1.04 (m, 3H), 1.01 - 0.63 (m, 1H) Step D. Procedure for preparation of tert-butyl (2S,4S)-4-(3-(3-bromo-2- methylphenoxy)propyl)-2-methylpiperidine-1-carboxylate and tert-butyl (2S,4R)-4-(3-(3- bromo-2-methylphenoxy)propyl)-2-methylpiperidine-1-carboxyla te The tert-butyl (2S)-4-[3-(3-bromo-2-methyl-phenoxy)propyl]-2-methyl-piperid ine-1- carboxylate as a stereoisomeric mixture was separated by SFC to give tert-butyl (2S,4S)-4-(3- (3-bromo-2-methylphenoxy)propyl)-2-methylpiperidine-1-carbox ylate (320 mg, 750.49 μmol, 43.84 % yield) as a yellow oil and tert-butyl (2S,4R)-4-(3-(3-bromo-2-methylphenoxy)propyl)- 2-methylpiperidine-1-carboxylate (170 mg, 398.70 μmol, 23.29 % yield) as a yellow oil. 1 H NMR (400 MHz, CDCl 3 ) δ = 7.06 (d, J = 8.0 Hz, 1H), 6.91 (t, J = 8.0 Hz, 1H), 6.67 (d, J = 8.4 Hz, 1H), 3.92 - 3.78 (m, 3H), 3.65 (dd, J = 7.2, 14.0 Hz, 1H), 3.04 - 2.93 (m, 1H), 2.24 (s, 3H), 1.92 - 1.80 (m, 1H), 1.79 - 1.64 (m, 3H), 1.49 (s, 2H), 1.44 - 1.41 (m, 1H), 1.39 (s, 9H), 1.13 - 1.06 (m, 5H) 1 H NMR (400 MHz, CDCl3) δ = 7.07 (d, J = 8.0 Hz, 1H), 6.91 (t, J = 8.0 Hz, 1H), 6.68 (d, J = 8.0 Hz, 1H), 3.86 (t, J = 6.4 Hz, 3H), 2.77 (t, J = 13.2 Hz, 1H), 2.24 (s, 3H), 1.79 - 1.71 (m, 2H), 1.66 - 1.46 (m, 4H), 1.39 (s, 9H), 1.35 - 1.17 (m, 4H), 1.06 (d, J = 6.8 Hz, 3H) Step E. Procedure for preparation of (2S,4S)-4-[3-(3-bromo-2-methyl-phenoxy)propyl]- 2-methyl-piperidine A solution of tert-butyl (2S,4S)-4-(3-(3-bromo-2-methylphenoxy)propyl)-2-methylpiperi dine- 1-carboxylate (190 mg, 445.61 μmol, 1 equiv.) in HCl/dioxane (3 mL) and DCM (1 mL) was stirred at 25 °C for 1.5 hours. The reaction mixture was concentrated under reduced pressure to give (2S,4S)-4-[3-(3-bromo-2-methyl-phenoxy)propyl]-2-methyl-pipe ridine (145 mg, 444.42 μmol, 99.73% yield) as a white solid which was used in the next step without further purification. MS (ESI) m/z: 362.0 [M+H] + . Step F. Procedure for preparation of ethyl 2-[(2S,4S)-4-[3-(3-bromo-2-methyl- phenoxy)propyl]-2-methyl-1-piperidyl]acetate To a solution of (2S,4S)-4-[3-(3-bromo-2-methyl-phenoxy)propyl]-2-methyl-pipe ridine (145 mg, 444.42 μmol, 1 equiv.) and ethyl 2-bromoacetate (74.22 mg, 444.42 μmol, 49.18 μL, 1 equiv.) in DMF (2 mL) was added K2CO3 (184.27 mg, 1.33 mmol, 3 equiv.). The mixture was stirred at 40 °C for 1.5 hours. The reaction mixture was partitioned between ethyl acetate (20 mL) and water (10 mL). The organic phase was separated, washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give ethyl 2-[(2S,4S)-4-[3- (3-bromo-2-methyl-phenoxy)propyl]-2-methyl-1-piperidyl]aceta te (190 mg, crude) as a yellow oil. MS (ESI) m/z: 412.1 [M+H] + . Step G. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(2S,4S)-1-(2-ethox y-2-oxo-ethyl)-2-methyl-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate A mixture of ethyl 2-[(2S,4S)-4-[3-(3-bromo-2-methyl-phenoxy)propyl]-2-methyl-1 - piperidyl]acetate (150 mg, 363.76 μmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tet ramethyl-1,3,2-dioxaborolan-2- yl)pyridine-2-carboxylate (267.38 mg, 436.51 μmol, 1.2 equiv.), KF (63.40 mg, 1.09 mmol, 25.56 μL, 3.0 equiv.), and Ad2nBuP Pd G 3 (cataCXium® A Pd G 3 ) (52.98 mg, 72.75 μmol, 0.2 equiv.) in dioxane (5 mL) and H 2 O (0.5 mL) was degassed and purged with N 2 three times. The mixture was stirred at 100 °C for 1 hour under N2 atmosphere. The reaction mixture was partitioned between ethyl acetate (30 mL) and water (20 mL). The organic phase was separated, washed with brine (20 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~45% ethyl acetate/petroleum ether) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(2S,4S)-1-(2-ethoxy-2- oxo-ethyl)-2-methyl-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate (260 mg, 295.59 μmol, 81.26% yield) as a yellow oil. MS (ESI) m/z: 818.3 [M+H] + . Step H. Procedure for preparation of 2-[(2S,4S)-4-[3-[3-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-2- methyl-phenoxy]propyl]-2-methyl-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[3-[(2S,4S)-1-(2-ethoxy-2-oxo-ethyl)-2-methyl-4-p iperidyl]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (130 mg, 158.92 μmol, 1.0 equiv.) in THF (3 mL) and H2O (1 mL) was added LiOH•H 2 O (20.01 mg, 476.75 μmol, 3.0 equiv.). The mixture was stirred at 40 °C for 6 hours. The reaction mixture was acidified to pH = 3-4 with citric acid and partitioned between DCM (20 mL) and water (15 mL). The organic phase was separated, washed with brine (15 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give 2-[(2S,4S)-4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3, 4-dihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]propyl]-2-methyl-1- piperidyl]acetic acid (100 mg, crude) as a yellow solid. MS (ESI) m/z: 790.4 [M+H] + . Step I. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[(2S,4S)-1-[2-[[3-( 2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]amino]-2-oxo-ethyl]-2-methyl-4-piperidyl ]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate To a solution of 2-[(2S,4S)-4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3, 4-dihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]propyl]-2-methyl-1- piperidyl]acetic acid (100 mg, 126.59 μmol, 1 equiv.) and 3-(6-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (39.23 mg, 151.90 μmol, 1.2 equiv.) in pyridine (1 mL) was added EDCI (36.40 mg, 189.88 μmol, 1.5 equiv.). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was partitioned between DCM (30 mL) and water (20 mL). The organic phase was separated, washed with brine (20 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[3-[3-[(2S,4S)-1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-2-methyl-4-piperidyl]propoxy]-2-methy l-phenyl]pyridine-2- carboxylate (100 mg, crude) as a yellow oil. MS (ESI) m/z: 516.1 [M/2+H] + . Step J. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[(2S,4S)-1-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-2-methyl-4-piperidyl]propoxy]-2-met hyl-phenyl]pyridine-2- carboxylic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[(2S,4S)-1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-meth yl-indazol-6-yl]amino]-2-oxo- ethyl]-2-methyl-4-piperidyl]propoxy]-2-methyl-phenyl]pyridin e-2-carboxylate (130 mg, 126.18 μmol, 1 equiv.) in TFA (3 mL) and DCM (1 mL) was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by reverse-phase HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[(2S,4S)-1-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-2-methyl-4-piperidyl]propoxy]-2-methy l-phenyl]pyridine-2- carboxylic acid (40.67 mg, 37.77 μmol, 29.93% yield) as a yellow solid. MS (ESI) m/z: 487.9 [M/2+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.96 - 12.51 (m, 2H), 10.88 (s, 1H), 9.89 - 9.71 (m, 1H), 8.18 - 8.09 (m, 1H), 8.03 (s, 2H), 7.78 (s, 1H), 7.63 (d, J = 8.0 Hz, 2H), 7.45 (d, J = 8.0 Hz, 3H), 7.36 (d, J = 11.2 Hz, 2H), 7.26 - 7.19 (m, 1H), 7.13 - 7.06 (m, 1H), 6.97 (d, J = 8.8 Hz, 1H), 6.88 (d, J = 8.4 Hz, 1H), 6.63 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.37 - 4.27 (m, 1H), 3.98 - 3.94 (m, 2H), 3.92 (s, 5H), 3.19 - 3.06 (m, 1H), 3.03 (d, J = 5.6 Hz, 2H), 2.99 - 2.87 (m, 1H), 2.68 - 2.60 (m, 2H), 2.41 - 2.30 (m, 3H), 2.20 - 2.14 (m, 1H), 1.90 (s, 3H), 1.83 - 1.60 (m, 5H), 1.39 (s, 3H), 1.30 - 1.21 (m, 1H), 1.06 (d, J = 4.0 Hz, 4H) Example 290. Preparation of Compound 344a 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4-[3-[(2S,4S)-1- [2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]- 2-oxo-ethyl]-2-methyl-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of (2S,4S)-4-[3-(4-bromo-3-methyl-phenoxy)propyl]- 2-methyl-piperidine A mixture of tert-butyl (2S,4S)-4-[3-(4-bromo-3-methyl-phenoxy)propyl]-2-methyl- piperidine-1-carboxylate (230 mg, 539.42 μmol, 1.0 equiv.) in HCl/dioxane (10 mL) was degassed and purged with N2 three times. The mixture was stirred at 25 °C for 2 hours under N2 atmosphere. The combined organic layers were filtered and concentrated under reduced pressure to give (2S,4S)-4-[3-(4-bromo-3-methyl-phenoxy)propyl]-2-methyl-pipe ridine (55 mg, crude) as a white solid, which was used in the next step without further purification. MS (ESI) m/z: 327.8 [M+H] + Step B. Procedure for preparation of ethyl 2-[(2S,4S)-4-[3-(4-bromo-3-methyl- phenoxy)propyl]-2-methyl-1-piperidyl]acetate A mixture of (2S,4S)-4-[3-(4-bromo-3-methyl-phenoxy)propyl]-2-methyl-pipe ridine (230 mg, 704.94 μmol, 1.0 equiv.), ethyl 2-bromoacetate (123.61 mg, 740.18 μmol, 81.92 μL, 1.05 equiv.), and TEA (214.00 mg, 2.11 mmol, 294.36 μL, 3.0 equiv.) in acetone (5 mL) was degassed and purged with N2 three times. The mixture was stirred at 30 °C for 10 hours under N2 atmosphere. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~100% ethyl acetate/petroleum ether) to give ethyl 2-[(2S,4S)-4-[3-(4-bromo-3-methyl- phenoxy)propyl]-2-methyl-1-piperidyl]acetate (230 mg, 557.77 μmol, 79.12% yield) as a white solid. MS (ESI) m/z: 412.2 [M+H] + Step C. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[3-[(2S,4S)-1-(2-ethox y-2-oxo-ethyl)-2-methyl-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine- 2-carboxylate (392.16 mg, 640.22 μmol, 1.2 equiv.), ethyl 2-[(2S,4S)-4-[3-(4-bromo-3-methyl-phenoxy)propyl]-2-methyl-1 - piperidyl]acetate (250 mg, 606.27 μmol, 1 equiv.), Ad2nBuP Pd G3 (77.71 mg, 106.70 μmol, 0.2 equiv.), and K2CO3 (1.5 M, 1.07 mL, 3 equiv.) in dioxane (5 mL) was degassed and purged with N 2 three times. The mixture was stirred at 100 °C for 1 hour under N 2 atmosphere in a microwave reactor. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~55% ethyl acetate/petroleum ether) to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[4-[3-[(2S,4S)-1-(2-ethoxy-2-oxo-ethy l)-2-methyl-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate (360 mg, 440.08 μmol, 82.49% yield) as a yellow solid. MS (ESI) m/z: 818.7 [M+H] + . Step D. Procedure for preparation of 2-[(2S,4S)-4-[3-[4-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-3- methyl-phenoxy]propyl]-2-methyl-1-piperidyl]acetic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[4-[3-[(2S,4S)-1-(2-ethoxy-2-oxo-ethyl)-2-methyl-4-pip eridyl]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (360 mg, 440.08 μmol, 1 equiv.), and LiOH•H2O (55.40 mg, 1.32 mmol, 3.0 equiv.) in H 2 O (1.5 mL) and THF (3 mL) was degassed and purged with N 2 three times. The mixture was stirred at 25 °C for 10 hours under N2 atmosphere. The combined organic layers were filtered and concentrated under reduced pressure to give 2-[(2S,4S)-4-[3- [4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-iso quinolin-2-yl]-2-tert- butoxycarbonyl-3-pyridyl]-3-methyl-phenoxy]propyl]-2-methyl- 1-piperidyl]acetic acid (350 mg, crude) as a yellow solid, which was used in the next step without further purification. MS (ESI) m/z: 790.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.03 - 12.67 (m, 1H), 8.01 (d, J = 8.0 Hz, 1H), 7.80 - 7.74 (m, 1H), 7.60 (d, J = 7.6 Hz, 1H), 7.49 - 7.30 (m, 6H), 7.02 - 6.89 (m, 1H), 6.86 (d, J = 8.4 Hz, 1H), 6.81 (d, J = 2.4 Hz, 1H), 6.71 (dd, J = 2.4, 8.4 Hz, 1H), 4.96 (s, 2H), 4.05 - 3.92 (m, 4H), 3.89 - 3.79 (m, 4H), 3.60 (dd, J = 2.8, 4.0, 6.4 Hz, 2H), 3.48 (d, J = 12.4 Hz, 2H), 3.33 (dd, J = 6.4, 9.2 Hz, 2H), 3.13 - 2.96 (m, 4H), 2.35 - 2.18 (m, 2H), 2.00 (s, 3H), 1.85 - 1.62 (m, 9H), 1.23 (d, J = 6.4 Hz, 3H) Step E. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(3-((2S,4S)-1-(2-((3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)-2-methylpiperidin- 4-yl)propoxy)-2- methylphenyl)picolinate A mixture of 2-[(2S,4S)-4-[3-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3, 4-dihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-3-methyl- phenoxy]propyl]-2-methyl-1- piperidyl]acetic acid (60 mg, 78.54 μmol, 1.0 equiv.), EDCI (22.58 mg, 117.81 μmol, 1.5 equiv.), and 3-(6-amino-1-methyl-1H-indazol-3-yl)piperidine-2,6-dione (24.34 mg, 94.25 μmol, 1.2 equiv.) in pyridine (2 mL) was degassed and purged with N 2 three times. The mixture was stirred at 25 °C for 2 hours under N 2 atmosphere. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH = 10:1) to give tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(4-(3-((2S,4 S)-1-(2-((3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2-oxoet hyl)-2-methylpiperidin-4- yl)propoxy)-2-methylphenyl)picolinate (30 mg, 29.87 μmol, 38.04% yield) as a yellow solid. MS (ESI) m/z: 1030.6 [M+H] + . Step F. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[4-[3-[(2S,4S)-1-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-2-methyl-4-piperidyl]propoxy]-2-met hyl-phenyl]pyridine-2- carboxylic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4-(3-((2S,4S)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)amino)-2- oxoethyl)-2-methylpiperidin-4-yl)propoxy)-2-methylphenyl)pic olinate (120 mg, 116.48 μmol, 1.0 equiv.) in TFA (1 mL) and DCM (1 mL) was degassed and purged with N 2 three times. The mixture was stirred at 25 °C for 16 hours under N2 atmosphere. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H- isoquinolin-2-yl]-3-[4-[3-[(2S,4S)-1-[2-[[3-(2,6-dioxo-3-pip eridyl)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-2-methyl-4-piperidyl]propoxy]-2-methy l-phenyl]pyridine-2- carboxylic acid (46.66 mg, 47.44 μmol, 40.73% yield) as a white solid. MS (ESI) m/z: 974.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.02 - 12.29 (m, 2H), 10.88 (s, 1H), 9.89 - 9.73 (m, 1H), 8.06 - 7.99 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.66 - 7.59 (m, 2H), 7.51 - 7.42 (m, 3H), 7.40 - 7.32 (m, 2H), 7.23 (dd, J = 1.2, 8.8 Hz, 1H), 6.93 (dd, J = 8.8, 14.8 Hz, 2H), 6.79 (d, J = 2.4 Hz, 1H), 6.71 (dd, J = 2.4, 8.4 Hz, 1H), 4.97 (s, 2H), 4.33 (dd, J = 5.2, 9.6 Hz, 1H), 3.97 - 3.89 (m, 7H), 3.10 - 2.89 (m, 4H), 2.72 - 2.54 (m, 3H), 2.35 (dt, J = 4.8, 9.2 Hz, 2H), 2.21 - 2.13 (m, 1H), 2.03 (s, 3H), 1.76 - 1.62 (m, 4H), 1.49 - 1.17 (m, 5H), 1.11 - 0.99 (m, 4H) Example 291. Preparation of Compound 344b 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4-[3-[(2R,4R)-1- [2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]- 2-oxo-ethyl]-2-methyl-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of (2R,4R)-4-[3-(4-bromo-3-methyl-phenoxy)propyl]- 2-methyl-piperidine A mixture of tert-butyl (2R,4R)-4-[3-(4-bromo-3-methyl-phenoxy)propyl]-2-methyl- piperidine-1-carboxylate (300.00 mg, 703.59 μmol, 1 equiv.) in DCM (5 mL) and HCl/dioxane (5 mL) was degassed and purged with N 2 three times. The mixture was stirred at 25 °C for 1 hour under N 2 atmosphere. The reaction mixture was then filtered and concentrated under reduced pressure to give (2R,4R)-4-[3-(4-bromo-3-methyl-phenoxy)propyl]-2-methyl- piperidine (229 mg, 701.87 μmol, 99.76% yield) as a white solid. MS (ESI) m/z: 328.0 [M+H] + Step B. Procedure for preparation of ethyl 2-[(2R,4R)-4-[3-(4-bromo-3-methyl- phenoxy)propyl]-2-methyl-1-piperidyl]acetate A mixture of (2R,4R)-4-[3-(4-bromo-3-methyl-phenoxy)propyl]-2-methyl-pipe ridine (229.00 mg, 701.87 μmol, 1 equiv.), ethyl 2-bromoacetate (105.49 mg, 631.68 μmol, 69.91 μL, 0.9 equiv.), K 2 CO 3 (291.01 mg, 2.11 mmol, 3 equiv.), and KI (11.65 mg, 70.19 μmol, 0.1 equiv.) in DMF (3 mL) was degassed and purged with N 2 three times. The mixture was stirred at 40 °C for 1 hour under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 30% ethyl acetate/petroleum ether) to give ethyl 2-[(2R,4R)-4-[3-(4-bromo-3-methyl- phenoxy)propyl]-2-methyl-1-piperidyl]acetate (280 mg, 679.02 μmol, 96.74% yield) as a white solid. MS (ESI) m/z: 412.2 [M+H] + Step C. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[3-[(2R,4R)-1-(2-ethox y-2-oxo-ethyl)-2-methyl-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate A mixture of ethyl 2-[(2R,4R)-4-[3-(4-bromo-3-methyl-phenoxy)propyl]-2-methyl-1 - piperidyl]acetate (190.00 mg, 460.76 μmol, 1 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tet ramethyl-1,3,2-dioxaborolan-2- yl)pyridine-2-carboxylate (338.69 mg, 552.92 μmol, 1.2 equiv.), Ad 2 nBuP Pd G 3 (67.11 mg, 92.15 μmol, 0.2 equiv.), and KF (40.15 mg, 691.14 μmol, 16.19 μL, 1.5 equiv.) in dioxane (2 mL) and H2O (0.2 mL) was degassed and purged with N2 three times. The mixture was stirred at 100 °C for 2 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 50% ethyl acetate / petroleum ether) to give tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[4-[3-[(2R,4R)-1-(2- ethoxy-2-oxo-ethyl)-2-methyl-4-piperidyl]propoxy]-2-methyl-p henyl]pyridine-2-carboxylate (310 mg, 378.96 μmol, 82.25% yield) as a white solid. MS (ESI) m/z: 818.4 [M+H] + Step D. Procedure for preparation of 2-[(2R,4R)-4-[3-[4-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-3- methyl-phenoxy]propyl]-2-methyl-1-piperidyl]acetic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[4-[3-[(2R,4R)-1-(2-ethoxy-2-oxo-ethyl)-2-methyl-4-pip eridyl]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (310.00 mg, 378.96 μmol, 1 equiv.) and LiOH•H2O (79.51 mg, 1.89 mmol, 5 equiv.) in THF (4 mL) and H 2 O (2 mL) was degassed and purged with N 2 three times. The mixture was stirred at 25 °C for 12 hours under N2 atmosphere. The reaction mixture was diluted with H2O (5 mL) and extracted with DCM (4 mL × 3). The combined organic layers were washed with H 2 O (4 mL × 3), filtered, and concentrated under reduced pressure to give 2-[(2R,4R)-4-[3-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3, 4-dihydro-1H-isoquinolin- 2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-3-methyl-phenoxy]prop yl]-2-methyl-1- piperidyl]acetic acid (260 mg, 329.12 μmol, 86.85% yield) as a yellow solid. MS (ESI) m/z: 790.3 [M+H] + Step E. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[3-[(2R,4R)-1-[2-[[3-( 2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]amino]-2-oxo-ethyl]-2-methyl-4-piperidyl ]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate A mixture of 2-[(2R,4R)-4-[3-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3, 4-dihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-3-methyl- phenoxy]propyl]-2-methyl-1- piperidyl]acetic acid (260.00 mg, 329.12 μmol, 1 equiv.), 3-(6-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (93.50 mg, 362.03 μmol, 1.1 equiv.), and EDCI (75.71 mg, 394.95 μmol, 1.2 equiv.) in pyridine (3 mL) was degassed and purged with N 2 three times. The mixture was stirred at 40 °C for 1 hour under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 60% ethyl acetate / petroleum ether) to give tert-butyl 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[4-[3-[(2R,4R)-1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-2-methyl-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate (300 mg, 291.19 μmol, 88.48% yield) as a yellow solid. MS (ESI) m/z: 1031.2 [M+H] + . Step F. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[4-[3-[(2R,4R)-1-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-2-methyl-4-piperidyl]propoxy]-2-met hyl-phenyl]pyridine-2- carboxylic acid A mixture of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[4-[3-[(2R,4R)-1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-meth yl-indazol-6-yl]amino]-2-oxo- ethyl]-2-methyl-4-piperidyl]propoxy]-2-methyl-phenyl]pyridin e-2-carboxylate (150 mg, 145.60 μmol, 1 equiv.) and TFA (767.50 mg, 6.73 mmol, 0.5 mL, 46.23 equiv.) in DCM (1.5 mL) was degassed and purged with N2 three times. The mixture was stirred at 40 °C for 140 min under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -3-[4-[3-[(2R,4R)-1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-2-methyl-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid (32 mg, 25.95 μmol, 17.82% yield) as a yellow solid. MS (ESI) m/z: 974.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 13.19 - 12.33 (m, 2H), 10.88 (s, 1H), 9.82 (s, 1H), 8.14 (s, 1H), 8.07 - 7.98 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.66 - 7.60 (m, 2H), 7.51 - 7.41 (m, 3H), 7.40 - 7.32 (m, 2H), 7.23 (d, J = 8.8 Hz, 1H), 6.93 (d, J = 8.8 Hz, 2H), 6.79 (d, J = 2.4 Hz, 1H), 6.70 (d, J = 8.4 Hz, 1H), 4.97 (s, 2H), 4.33 (d, J = 9.6 Hz, 1H), 3.94 - 3.90 (m, 5H), 3.46 - 3.41 (m, 2H), 3.17 (s, 1H), 3.04 - 3.01 (m, 2H), 2.94 (s, 2H), 2.78 (s, 2H), 2.45 - 2.38 (m, 2H), 2.22 - 2.13 (m, 1H), 2.03 (s, 3H), 1.96 (s, 2H), 1.78 - 1.70 (m, 2H), 1.68 - 1.65 (m, 2H), 1.36 - 1.34 (m, 2H), 1.09 - 1.00 (m, 5H). Example 292. Preparation of Compound 344c 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4-[3-[(2R,4S)-1- [2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]- 2-oxo-ethyl]-2-methyl-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl (2R)-4-[3-(4-bromo-3-methyl- phenoxy)propyl]-2-methyl-piperidine-1-carboxylate To a solution of tert-butyl (2R)-4-(3-hydroxypropyl)-2-methyl-piperidine-1-carboxylate (1.1 g, 4.27 mmol, 1.0 equiv.) in toluene (20 mL) was added 4-bromo-3-methyl-phenol (879.33 mg, 4.70 mmol, 1.1 equiv.) and 2-(tributyl-phosphanylidene) acetonitrile (1.55 g, 6.41 mmol, 1.5 equiv.) at 25 °C. The reaction mixture was degassed and purged with N2 three times, and then stirred at 120 °C for 16 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The crude product was purified by reversed-phase HPLC (with 0.1% formic acid) to give tert-butyl (2R)-4-[3-(4-bromo-3-methyl- phenoxy)propyl]-2-methyl-piperidine-1-carboxylate (1.9 g, 4.18 mmol, 48.95% yield) as a colorless oil. MS (ESI) m/z: 448.2 [M+Na] + . Step B. Procedure for preparation of tert-butyl (2R,4S)-4-[3-(4-bromo-3-methyl- phenoxy)propyl]-2-methyl-piperidine-1-carboxylate and tert-butyl (2R,4R) -4-[3-(4- bromo-3-methyl-phenoxy)propyl]-2-methyl-piperidine-1-carboxy late The stereoisomers of tert-butyl (2R)-4-[3-(4-bromo-3-methyl-phenoxy)propyl]-2-methyl- piperidine-1-carboxylate were separated by SFC to give tert-butyl (2R,4R) -4-[3-(4-bromo-3- methyl-phenoxy)propyl]-2-methyl-piperidine-1-carboxylate (310 mg, 727.04 μmol, 16.32% yield) and tert-butyl (2R,4S)-4-[3-(4-bromo-3-methyl-phenoxy)propyl]-2-methyl-pipe ridine- 1-carboxylate (1.15 g, 2.70 mmol, 60.53% yield) as colorless oils. 1 H NMR (400 MHz, DMSO- d 6 ) δ = 7.42 (d, J = 8.4 Hz, 1H), 6.94 (d, J = 2.8 Hz, 1H), 6.70 (dd, J = 3.2, 8.8 Hz, 1H), 3.92 (t, J = 6.4 Hz, 2H), 3.81 - 3.70 (m, 1H), 3.58 - 3.43 (m, 1H), 3.08 - 2.98 (m, 1H), 2.29 (s, 3H), 1.83 - 1.63 (m, 4H), 1.54 - 1.45 (m, 1H), 1.44 - 1.33 (m, 11H), 1.19 - 1.04 (m, 5H). 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.42 (d, J = 8.8 Hz, 1H), 6.94 (d, J = 2.8 Hz, 1H), 6.71 (dd, J = 3.2, 8.8 Hz, 1H), 4.26 (s, 1H), 3.92 (t, J = 6.4 Hz, 2H), 3.80 (d, J = 11.2 Hz, 1H), 2.93 - 2.66 (m, 1H), 2.29 (s, 3H), 1.77 - 1.58 (m, 4H), 1.57 - 1.50 (m, 1H), 1.38 (s, 9H), 1.31 - 1.21 (m, 2H), 1.20 - 1.10 (m, 1H), 1.06 (d, J = 6.8 Hz, 3H), 0.95 - 0.80 (m, 1H). Step C. Procedure for preparation of (2R,4S)-4-[3-(4-bromo-3-methyl-phenoxy)propyl]- 2-methyl-piperidine To a solution of tert-butyl (2R,4S)-4-[3-(4-bromo-3-methyl-phenoxy)propyl]-2-methyl- piperidine-1-carboxylate (500.00 mg, 1.17 mmol, 1.0 equiv.) in DCM (1.5 mL) was added HCl/dioxane (4 M, 2.50 mL, 8.53 equiv.) at 25 °C. The reaction was stirred at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give (2R,4S)-4-[3-(4-bromo- 3-methyl-phenoxy)propyl]-2-methyl-piperidine (460 mg, crude) as a white solid. MS (ESI) m/z: 326.1 [M+H] + . Step D. Procedure for preparation of ethyl 2-[(2R, 4S)-4-[3-(4-bromo-3-methyl- phenoxy)propyl]-2-methyl-1-piperidyl]acetate To a solution of (2R,4S)-4-[3-(4-bromo-3-methyl-phenoxy)propyl]-2-methyl-pipe ridine (450.00 mg, 1.24 mmol, 1 equiv., HCl) in acetone (4 mL) was added ethyl 2-bromoacetate (207.18 mg, 1.24 mmol, 137.30 μL, 1.0 equiv.) and TEA (251.07 mg, 2.48 mmol, 345.35 μL, 2.0 equiv.) at 25 °C. The reaction was stirred at 25 °C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 50% ethyl acetate/petroleum ether) to give ethyl 2-[(2R, 4S)-4-[3-(4-bromo-3-methyl-phenoxy)propyl]-2-methyl-1-piperi dyl]acetate (300 mg, 727.52 μmol, 58.64% yield) as a colorless oil. MS (ESI) m/z: 412.1 [M+H] + . Step E. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[3-[(2R,4S)-1-(2-ethox y-2-oxo-ethyl)-2-methyl-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate A mixture of ethyl 2-[(2R, 4S)-4-[3-(4-bromo-3-methyl-phenoxy)propyl]-2-methyl-1- piperidyl]acetate (290.00 mg, 703.27 μmol, 1.0 equiv.), tert-butyl 6-[8-(1, 3-benzothiazol-2- ylcarbamoyl) -3, 4-dihydro-1H-isoquinolin-2-yl]-3-(4, 4, 5, 5-tetramethyl-1, 3, 2- dioxaborolan-2-yl) pyridine-2-carboxylate (473.86 mg, 773.60 μmol, 1.1 equiv.), KF (122.58 mg, 2.11 mmol, 49.43 μL, 3.0 equiv.), and [2-(2-aminophenyl)phenyl]palladium(1+)bis(1- adamantyl)-butyl-phosphanemethanesulfonate (102.43 mg, 140.65 μmol, 0.2 equiv.) in dioxane (3 mL) and H2O (0.3 mL) was degassed and purged with N2 three times. The mixture was stirred at 100 °C for 16 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 6% MeOH/DCM) to give tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[3-[(2R,4 S)-1-(2-ethoxy-2-oxo-ethyl)-2- methyl-4-piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carbo xylate (400 mg, 327.62 μmol, 46.58% yield) as a yellow solid. MS (ESI) m/z: 818.5 [M+H] + . Step F. Procedure for preparation of 2-[(2R,4S)-4-[3-[4-[6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxyc arbonyl-3-pyridyl]-3- methyl-phenoxy]propyl]-2-methyl-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[4-[3-[(2R,4S)-1-(2-ethoxy-2-oxo-ethyl)-2-methyl-4-p iperidyl]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (390 mg, 319.43 μmol, 1.0 equiv.) in THF (4 mL) and H2O (0.4 mL) was added LiOH•H 2 O (40.21 mg, 958.28 μmol, 3.0 equiv.) at 25 °C. The reaction was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was treated with water (30 mL), and its pH was adjusted to ~ 5 by progressively adding diluted HCl. The mixture was extracted with DCM/MeOH (10/1) (20 mL × 5). The combined organic layers were washed with brine (20 mL × 1), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give 2-[(2R,4S)-4-[3-[4-[6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl] -2-tert-butoxycarbonyl-3- pyridyl]-3-methyl-phenoxy]propyl]-2-methyl-1-piperidyl]aceti c acid (250 mg, 233.23 μmol, 73.02% yield) as a yellow solid. MS (ESI) m/z: 790.5 [M+H] + . Step G. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[3-[(2R,4S)-1-[2-[[3-( 2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]amino]-2-oxo-ethyl]-2-methyl-4-piperidyl ]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate To a solution of 2-[(2R,4S)-4-[3-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3, 4-dihydro-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-3-methyl- phenoxy]propyl]-2-methyl-1- piperidyl]acetic acid (250.00 mg, 316.46 μmol, 1.0 equiv.) in pyridine (3 mL) was added 3-(6- amino-1-methyl-indazol-3-yl) piperidine-2,6-dione (98.08 mg, 379.76 μmol, 1.2 equiv.) and EDCI (78.87 mg, 411.40 μmol, 1.3 equiv.) at 25 °C. The reaction was stirred at 25 °C for 16 hours. The mixture was diluted with water (50 mL) and filtered. The organic layer was concentrated to afford a brown residue. The residue was suspended in MeOH (30 mL) and stirred at 15 °C for 30 min. The resulting solid was collected by filtration and dried in vacuo to give tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4- [3-[(2R,4S)-1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazo l-6-yl]amino]-2-oxo-ethyl]-2- methyl-4-piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carbo xylate (235 mg, 164.23 μmol, 51.90% yield) as a yellow solid. MS (ESI) m/z: 1030.6 [M+H] + . Step H. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[4-[3-[(2R,4S)-1-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-2-methyl-4-piperidyl]propoxy]-2-met hyl-phenyl]pyridine-2- carboxylic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[4-[3-[(2R,4S)-1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-meth yl-indazol-6-yl]amino]-2-oxo- ethyl]-2-methyl-4-piperidyl]propoxy]-2-methyl-phenyl]pyridin e-2-carboxylate (225 mg, 218.40 μmol, 1.0 equiv.) in DCM (1 mL) and TFA (1 mL) was stirred at 40 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[4-[3-[(2R,4S)-1-[2-[[3-(2,6- dioxo-3-piperidyl)-1-methyl- indazol-6-yl]amino]-2-oxo-ethyl]-2-methyl-4-piperidyl]propox y]-2-methyl-phenyl]pyridine- 2-carboxylic acid (49.42 mg, 48.00 μmol, 21.98% yield) as a yellow solid. MS (ESI) m/z: 974.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.03 - 12.23 (m, 2H), 10.88 (s, 1H), 10.09 - 9.67 (m, 1H), 8.08 - 7.99 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.64 (t, J = 8.8 Hz, 2H), 7.51 - 7.41 (m, 3H), 7.41 - 7.32 (m, 2H), 7.21 (d, J = 8.8 Hz, 1H), 6.95 (d, J = 8.8 Hz, 1H), 6.92 (d, J = 8.4 Hz, 1H), 6.79 (s, 1H), 6.71 (d, J = 8.0 Hz, 1H), 4.97 (s, 2H), 4.33 (dd, J = 4.8, 9.6 Hz, 1H), 3.97 - 3.89 (m, 7H), 3.02 (t, J = 5.2 Hz, 2H), 2.75 - 2.57 (m, 4H), 2.42 - 2.28 (m, 2H), 2.17 (dd, J = 5.2, 12.8 Hz, 1H), 2.03 (s, 3H), 1.79 - 1.53 (m, 7H), 1.46 - 1.27 (m, 4H), 1.13 - 1.00 (m, 3H). Example 293. Preparation of Compound 344d 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(3-((2S,4R)-1- (2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)am ino)-2-oxoethyl)-2- methylpiperidin-4-yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl (S,E)-4-(3-(benzyloxy)propylidene)-2- methylpiperidine-1-carboxylate To a solution of 3-benzyloxypropyl(triphenyl)phosphoniumbromide (17.28 g, 35.17 mmol, 1.5 equiv.) in THF (50 mL) was added LiHMDS (1 M, 46.89 mL, 2.0 equiv.) at 0°C. The reaction mixture was stirred for 1 hour under N2 atmosphere, then tert-butyl (2S)-2-methyl-4-oxo- piperidine-1-carboxylate (5 g, 23.44 mmol, 1.0 equiv.) in THF (50 mL) was added dropwise at 0°C. The resulting mixture was stirred at 25°C for 15 hours under N 2 atmosphere. The reaction mixture was quenched by addition of saturated NH4Cl 50 mL at 0°C, and then extracted with ethyl acetate (50 mL × 3). The combined organic layers were washed with H2O (100 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 6% ethyl acetate / petroleum ether) to give tert-butyl (S,E)-4-(3-(benzyloxy)propylidene)-2-methylpiperidine-1-carb oxylate (3.4 g, 9.84 mmol, 41.98% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.43 - 7.22 (m, 5H), 5.42 - 5.20 (m, 1H), 4.47- 4.40 (m, 2H), 4.37 - 4.19 (m, 1H), 4.05 - 3.77 (m, 1H), 3.43 - 3.38 (m, 2H), 3.35 - 3.30 (m, 1H), 2.83 - 2.64 (m, 1H), 2.44 - 1.74 (m, 6H), 1.45 - 1.36 (m, 9H), 1.09 - 1.07 (m, 1H), 0.97 - 0.92 (m, 2H) Step B. Procedure for preparation of tert-butyl (2S)-4-(3-hydroxypropyl)-2- methylpiperidine-1-carboxylate A mixture of tert-butyl (S,E)-4-(3-(benzyloxy)propylidene)-2-methylpiperidine-1-carb oxylate (3.4 g, 9.84 mmol, 1.0 equiv.), Pd / C (1.5 g, 1.41 mmol, 10% purity, 0.143 equiv.), and Pd(OH)2 (1.5 g, 2.14 mmol, 20% purity, 0.217 equiv.) in EtOH (34 mL) was degassed and purged with N2 three times. The mixture was stirred at 60 °C for 16 hours under N2 atmosphere. The combined organic layers were filtered and concentrated under reduced pressure to give tert-butyl (2S)-4-(3-hydroxypropyl)-2-methylpiperidine-1-carboxylate (2.4 g, crude) as a yellow oil, which was used in the next step without further purification. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 4.42 - 4.17 (m, 2H), 3.86 - 3.71 (m, 1H), 2.90 - 2.64 (m, 1H), 1.70 - 1.47 (m, 3H), 1.43 - 1.37 (m, 13H), 1.30 - 1.11 (m, 4H), 1.10 - 1.04 (m, 3H). Step C. Procedure for preparation of tert-butyl (2S)-4-(3-(4-bromo-3- methylphenoxy)propyl)-2-methylpiperidine-1-carboxylate A mixture of tert-butyl (2S)-4-(3-hydroxypropyl)-2-methylpiperidine-1-carboxylate (2.3 g, 8.94 mmol, 1.1 equiv.), 4-bromo-3-methyl-phenol (1.52 g, 8.12 mmol, 1.0 equiv.), 2-(tributyl- phosphanylidene)acetonitrile (2.94 g, 12.19 mmol, 1.5 equiv.) in toluene (23 mL) was degassed and purged with N2 three times. The mixture was stirred at 120 °C for 16 hours under N2 atmosphere. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 3% ethyl acetate / petroleum ether) to give tert-butyl (2S)-4-(3-(4-bromo-3- methylphenoxy)propyl)-2-methylpiperidine-1-carboxylate (1.95 g, 4.37 mmol, 53.79% yield) as a white solid. Step D. Procedure for preparation of tert-butyl (2S,4S)-4-(3-(4-bromo-3- methylphenoxy)propyl)-2-methylpiperidine-1-carboxylate and tert-butyl (2S,4R)-4-(3-(4- bromo-3-methylphenoxy)propyl)-2-methylpiperidine-1-carboxyla te The stereoisomers of tert-butyl (2S)-4-(3-(4-bromo-3-methylphenoxy)propyl)-2- methylpiperidine-1-carboxylate were separated by prep-HPLC (column: DAICEL CHIRALPAK IG (250 mm × 30 mm, 10 um); mobile phase: [CO 2 - I – PrOH (0.1% NH 3 H 2 O)]; B%: 15%, isocratic elution mode and column: DAICEL CHIRALPAK IG (250 mm × 30 mm, 10 um); mobile phase: [CO2 - CAN / I - PrOH(0.1% NH3H2O)]; B%: 20%, isocratic elution mode) to give tert-butyl (2S,4S)-4-(3-(4-bromo-3-methylphenoxy)propyl)-2-methylpiperi dine- 1-carboxylate (293 mg, 687.17 μmol, 15.42% yield) and tert-butyl (2S,4R)-4-(3-(4-bromo-3- methylphenoxy)propyl)-2-methylpiperidine-1-carboxylate (650 mg, 1.52 mmol, 34.21% yield) as a colorless oil. MS (ESI) m/z: 327.8 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ =7.42 (d, J = 8.8 Hz, 1H), 6.94 (d, J = 2.8 Hz, 1H), 6.70 (dd, J = 2.8, 8.8 Hz, 1H), 3.92 (t, J = 6.4 Hz, 2H), 3.81 - 3.72 (m, 1H), 3.56 - 3.51 (m, 1H), 3.07-3.00 (m, 1H), 2.29 (s, 3H), 1.77 - 1.67 (m, 4H), 1.52 - 1.45 (m, 1H), 1.38 (s, 11H), 1.16 - 1.09 (m, 5H). 1 H NMR (400 MHz, DMSO-d6) δ = 7.42 (d, J = 8.8 Hz, 1H), 6.94 (d, J = 2.8 Hz, 1H), 6.71 (dd, J = 2.8, 8.8 Hz, 1H), 4.27 (s, 1H), 3.92 (t, J = 6.4 Hz, 2H), 3.80 (d, J = 12.0 Hz, 1H), 2.90 - 2.70 (m, 1H), 2.29 (s, 3H), 1.75 - 1.62 (m, 4H), 1.58 - 1.52 (m, 1H), 1.40 - 1.37(m, 9H), 1.31 - 1.24 (m, 2H), 1.19-1.12 (m, 1H), 1.06 (d, J = 6.8 Hz, 3H), 0.95 - 0.84 (m, 1H) Step E. Procedure for preparation of (2S,4R)-4-(3-(4-bromo-3-methylphenoxy)propyl)-2- methylpiperidine A solution of tert-butyl (2S,4R)-4-(3-(4-bromo-3-methylphenoxy)propyl)-2-methylpiperi dine- 1-carboxylate (360.00 mg, 844.30 μmol, 1.0 equiv.) in HCl/dioxane (4 mL) was stirred at 25 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give (2S,4R)-4-(3-(4-bromo-3-methylphenoxy)propyl)-2-methylpiperi dine (300 mg, crude) as a white solid, which was used in the next step without further purification. MS (ESI) m/z: 327.8 [M+H] + . Step F. Procedure for preparation of ethyl 2-((2S,4R)-4-(3-(4-bromo-3- methylphenoxy)propyl)-2-methylpiperidin-1-yl)acetate A mixture of (2S,4R)-4-(3-(4-bromo-3-methylphenoxy)propyl)-2-methylpiperi dine (270.00 mg, 744.35 μmol, 1.0 equiv., HCl), ethyl 2-bromoacetate (124.31 mg, 744.35 μmol, 82.38 μL, 1.0 equiv.), and TEA (150.64 mg, 1.49 mmol, 207.21 μL, 2.0 equiv.) in acetone (3 mL) was stirred at 25 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 100% ethyl acetate / petroleum ether) to give ethyl 2-((2S,4R)-4-(3-(4-bromo-3- methylphenoxy)propyl)-2-methylpiperidin-1-yl)acetate (300 mg, 727.52 μmol, 97.74% yield) as a white solid. MS (ESI) m/z: 414.0 [M+H] + . Step G. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(3-((2S,4R)-1-(2-ethoxy-2- oxoethyl)-2- methylpiperidin-4-yl)propoxy)-2-methylphenyl)picolinate A mixture of ethyl 2-((2S,4R)-4-(3-(4-bromo-3-methylphenoxy)propyl)-2-methylpip eridin-1- yl)acetate (300 mg, 727.52 μmol, 1.0 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tet ramethyl-1,3,2-dioxaborolan-2- yl)pyridine-2-carboxylate (534.77 mg, 873.02 μmol, 1.2 equiv.), KF (1.5 M, 1.46 mL, 3.0 equiv.), and [2-(2-aminophenyl)phenyl]palladium(1 + )bis(1-adamantyl)-butyl-phosphane methanesulfonate (52.98 mg, 72.75 μmol, 0.1 equiv.) in dioxane (4 mL) was degassed and purged with N2 three times. The mixture was stirred at 100 °C for 1 hour under N2 atmosphere in a microwave reactor. The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0 ~ 70% ethyl acetate / petroleum ether) to give tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(4-(3-((2S,4 R)-1-(2-ethoxy-2-oxoethyl)-2- methylpiperidin-4-yl)propoxy)-2-methylphenyl)picolinate (600 mg, crude) as a yellow solid. MS (ESI) m/z: 818.7 [M+H] + . Step H. Procedure for preparation of 2-((2S,4R)-4-(3-(4-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-3- methylphenoxy)propyl)-2-methylpiperidin-1-yl)acetic acid A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4-(3-((2S,4R)-1-(2-ethoxy-2-oxoethyl)-2-methylpiperid in-4-yl)propoxy)-2- methylphenyl)picolinate (600.00 mg, 733.47 μmol, 1.0 equiv.), LiOH•H2O (92.34 mg, 2.20 mmol, 3.0 equiv.) in H 2 O (2 mL) and THF (6 mL) was stirred at 25 °C for 2 hours. The reaction mixture was diluted with H2O (5 mL) and concentrated to a turbid liquid. Then the pH of the turbid liquid was adjusted to pH = 3 with 1 N HCl (5 mL). The solution was extracted with DCM/MeOH (20:1) and the combined organic layers were filtered and concentrated to give 2- ((2S,4R)-4-(3-(4-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-d ihydroisoquinolin-2(1H)-yl)-2- (tert-butoxycarbonyl)pyridin-3-yl)-3-methylphenoxy)propyl)-2 -methylpiperidin-1-yl)acetic acid (550 mg, 696.22 μmol, 94.92% yield) as a yellow solid, which was used in the next step without further purification. MS (ESI) m/z: 790.7 [M+H] + . Step I. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(3-((2S,4R)-1-(2-((3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)-2-methylpiperidin- 4-yl)propoxy)-2- methylphenyl)picolinate A mixture of 2-((2S,4R)-4-(3-(4-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4 - dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin- 3-yl)-3- methylphenoxy)propyl)-2-methylpiperidin-1-yl)acetic acid (200.00 mg, 253.17 μmol, 1.0 equiv.), 3-(6-amino-1-methyl-indazol-3-yl)piperidine-2,6-dione (78.47 mg, 303.81 μmol, 1.2 equiv.) and EDCI (72.80 mg, 379.76 μmol, 1.5 equiv.) in pyridine (2 mL) was stirred at 25 °C for 2 hours. The mixture was diluted with H2O (5 mL × 3) and extracted with ethyl acetate (10 mL × 3). The combined organic layers were filtered and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3- (4-(3-((2S,4R)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1 H-indazol-6-yl)amino)-2- oxoethyl)-2-methylpiperidin-4-yl)propoxy)-2-methylphenyl)pic olinate (200 mg, crude) as a yellow solid, which was used in the next step without further purification. MS (ESI) m/z: 1030.5 [M+H] + . Step J. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(3-((2S,4R)-1-(2-((3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)-2-methylpiperidin- 4-yl)propoxy)-2- methylphenyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4-(3-((2S,4R)-1-(2-((3-(2,6-dioxopiperidin-3-yl)-1-me thyl-1H-indazol-6-yl)amino)-2- oxoethyl)-2-methylpiperidin-4-yl)propoxy)-2-methylphenyl)pic olinate (200.00 mg, 194.13 μmol, 1.0 equiv.) in TFA (1 mL) and DCM (1 mL) was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(3-((2S,4R)-1-(2-((3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-6- yl)amino)-2-oxoethyl)-2-methylpiperidin-4-yl)propoxy)-2-meth ylphenyl)picolinic acid (98.90 mg, 98.35 μmol, 50.66% yield) as a yellow solid. MS (ESI) m/z: 974.5[M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.02 - 12.76 (m, 1H), 12.73 - 12.39 (m, 1H), 10.89 (s, 1H), 9.99 - 9.24 (m, 1H), 8.07 - 7.99 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.72 - 7.60 (m, 2H), 7.51 - 7.42 (m, 3H), 7.41 - 7.31 (m, 2H), 7.21 - 7.10 (m, 1H), 6.94 (dd, J = 8.8, 13.6 Hz, 2H), 6.81 - 6.77 (m, 1H), 6.74 - 6.69 (m, 1H), 4.97 (s, 2H), 4.34 (dd, J = 5.2, 9.6 Hz, 1H), 4.22 - 4.04 (m, 1H), 3.98 - 3.89 (m, 7H), 3.03 (t, J = 5.6 Hz, 2H), 2.68 - 2.61 (m, 2H), 2.56 - 2.51 (m, 4H), 2.37 - 2.31 (m, 1H), 2.24 - 2.14 (m, 1H), 2.03 (s, 3H), 1.90 - 1.60 (m, 6H), 1.53 - 1.20 (m, 6H). Example 294. Preparation of Compound 346a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- ((2R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)-2- (trifluoromethyl)piperazin-1-yl)butyl)cyclohexyl)oxy)-2-meth ylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl (2R)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1- methyl-indazol-6-yl]-2-(trifluoromethyl)piperazine-1-carboxy late A mixture of 6-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (650 mg, 1.30 mmol, 1.0 equiv.), tert-butyl (2R)-2-(trifluoromethyl)piperazine-1-carboxylate (495.40 mg, 1.95 mmol, 1.5 equiv.), Pd2(dba)3 (118.95 mg, 129.90 μmol, 0.1 equiv.), RuPhos (121.23 mg, 259.80 μmol, 0.2 equiv.), and Cs2CO3 (1.27 g, 3.90 mmol, 3.0 equiv.) in toluene (10 mL) was degassed and purged with N 2 three times. The mixture was stirred at 100 °C for 16 hours under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~23% ethyl acetate/petroleum ether) to afford tert-butyl (2R)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl- indazol-6-yl]-2-(trifluoromethyl)piperazine-1-carboxylate (700 mg, 997.44 μmol, 76.79% yield) as a yellow oil. MS (ESI) m/z: 674.3 [M+H] + . Step B. Procedure for preparation of tert-butyl (2R)-4-[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]-2-(trifluoromethyl)piperazine-1-carboxy late To a solution of tert-butyl (2R)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-yl] -2- (trifluoromethyl)piperazine-1-carboxylate (700 mg, 1.04 mmol, 1.0 equiv.) in THF (10 mL) and EtOH (10 mL) was added Pd/C (350 mg, 328.89 μmol, 10% purity, 0.317 equiv.) and Pd(OH)2 (350 mg, 2.49 mmol, 2.40 equiv.) under N2 atmosphere. The suspension was degassed and purged with H 2 three times. The mixture was stirred under H 2 (15 Psi) at 40 °C for 16 hours. The reaction mixture was filtered and washed with THF (100 mL). The filtrate was concentrated under reduced pressure to afford tert-butyl (2R)-4-[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]-2-(trifluoromethyl)piperazine-1-carboxy late (600 mg, crude) as a yellow oil, which was used in the next step without further purification. MS (ESI) m/z: 496.4 [M+H] + . Step C. Procedure for preparation of 3-[1-methyl-6-[(3R)-3-(trifluoromethyl)piperazin-1- yl]indazol-3-yl]piperidine-2,6-dione A solution of tert-butyl (2R)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]-2- (trifluoromethyl)piperazine-1-carboxylate (600 mg, 1.21 mmol, 1.0 equiv.) in HCl/dioxane (10 mL) was stirred at 25 °C for 16 hours. The reaction mixture was filtered to afford 3-[1-methyl- 6-[(3R)-3-(trifluoromethyl)piperazin-1-yl]indazol-3-yl]piper idine-2,6-dione (400 mg, crude) as a white solid, which was used in the next step without further purification. MS (ESI) m/z: 396.1 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.86 (s, 1H), 7.59 (d, J = 8.8 Hz, 1H), 7.10 (s, 1H), 7.00 (dd, J = 1.2, 9.2 Hz, 1H), 4.70 - 4.61 (m, 1H), 4.07 - 3.99 (m, 2H), 3.94 (s, 3H), 3.37 - 3.14 (m, 5H), 2.65 - 2.58 (m, 2H), 2.32 (J = 4.8, 8.4 Hz, 1H), 2.15 (dd, J = 5.6, 13.6 Hz, 1H) Step D. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-((2R)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-2-(trifluoromethyl)piperazin-1-yl)bu tyl)cyclohexyl)oxy)-2- methylphenyl)picolinate To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2-methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)o xy)phenyl)picolinate (100 mg, 134.24 μmol, 1.0 equiv.) and 3-[1-methyl-6-[(3R)-3-(trifluoromethyl)piperazin-1-yl]indazo l-3- yl]piperidine-2,6-dione (63.69 mg, 161.09 μmol, 1.2 equiv.) in DCM (5 mL) was added NaBH(OAc)3 (85.35 mg, 402.72 μmol, 3.0 equiv.). The mixture was stirred at 25 °C for 5 hours. The reaction mixture was diluted with H 2 O (30 mL) and extracted with ethyl acetate (20 mL × 3). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (dichloromethane : methanol = 15 : 1) to afford tert-butyl 6-(8-(benzo[d]thiazol- 2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4 r)-4-(4-((2R)-4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)-2-(trifluorom ethyl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (70 mg, 44.20 μmol, 32.93% yield) as a light yellow solid. MS (ESI) m/z: 1124.5 [M+H] + . Step E. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-((2R)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-2-(trifluoromethyl)piperazin-1-yl)bu tyl)cyclohexyl)oxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4-((2R)-4-(3-(2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-6- yl)-2-(trifluoromethyl)piperazin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinate (70 mg, 62.26 μmol, 1.0 equiv.) in DCM (1 mL) was added TFA (3.07 g, 26.92 mmol, 2 mL, 432.45 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was then concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(4-((2R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl -1H-indazol-6-yl)-2- (trifluoromethyl)piperazin-1-yl)butyl)cyclohexyl)oxy)-2-meth ylphenyl)picolinic acid (24.17 mg, 22.36 μmol, 35.91% yield) as a white solid. MS (ESI) m/z: 1068.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.91 - 12.83 (m, 1H), 10.86 (s, 1H), 8.04 (d, J = 8.0 Hz, 1H), 7.80 (d, J = 7.6 Hz, 1H), 7.63 (d, J = 6.4 Hz, 1H), 7.54 - 7.49 (m, 1H), 7.49 - 7.43 (m, 3H), 7.40 - 7.33 (m, 2H), 7.12 - 7.03 (m, 1H), 6.95 (dd, J = 8.4, 18.4 Hz, 2H), 6.87 - 6.82 (m, 2H), 6.61 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.27 (dd, J = 5.2, 9.2 Hz, 1H), 4.23 - 4.15 (m, 1H), 3.90 (s, 4H), 3.64 (d, J = 9.6 Hz, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.88 - 2.82 (m, 1H), 2.75 (d, J = 6.4 Hz, 2H), 2.67 (d, J = 1.6 Hz, 1H), 2.61 (t, J = 6.0 Hz, 2H), 2.35 - 2.32 (m, 2H), 2.19 - 2.15 (m, 1H), 2.11 - 2.05 (m, 2H), 1.87 (s, 3H), 1.79 (d, J = 12.8 Hz, 2H), 1.49 (dd, J = 4.0, 6.0 Hz, 2H), 1.40 - 1.20 (m, 9H), 1.11 - 1.02 (m, 2H) Example 295. Preparation of Compound 346b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(4- ((2S)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)-2-(trifluoromethyl)piperazin- 1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 4-[3-(2,6-dibenzyloxy-3-pyridyl)-1- methyl-indazol-6-yl]-2-(trifluoromethyl)piperazine-1-carboxy late A mixture of 6-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (1.3 g, 2.60 mmol, 1.0 equiv.), tert-butyl 2-(trifluoromethyl)piperazine-1-carboxylate (990.81 mg, 3.90 mmol, 1.5 equiv.), Pd2(dba)3 (237.90 mg, 259.80 μmol, 0.1 equiv.), RuPhos (242.47 mg, 519.60 μmol, 0.2 equiv.) and Cs2CO3 (2.54 g, 7.79 mmol, 3 equiv.) in toluene (20 mL) was degassed and purged with N 2 three times. The mixture was stirred at 100 °C for 16 hours under a N 2 atmosphere. The reaction mixture was filtered and concentrated under pressure to give a residue. The residue was purified by flash silica gel chromatography (0~24% ethyl acetate/petroleum ether) to give tert-butyl 4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-yl]-2- (trifluoromethyl)piperazine-1-carboxylate (1.1 g, 1.55 mmol, 59.70% yield) as a yellow oil. MS (ESI) m/z: 674.3 [M+H] + . Step B. Procedure for preparation of tert-butyl (2R)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1- methyl-indazol-6-yl]-2-(trifluoromethyl)piperazine-1-carboxy late and tert-butyl (2S)-4- [3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-yl]-2-(tri fluoromethyl)piperazine-1- carboxylate The stereoisomers of tert-butyl 4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-yl]-2- (trifluoromethyl)piperazine-1-carboxylate mixture were separated by SFC to afford tert-butyl (2R)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-yl] -2-(trifluoromethyl)piperazine- 1-carboxylate (390 mg, 578.87 μmol, 35.45% yield) as a white solid and tert-butyl (2S)-4-[3- (2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-yl]-2-(triflu oromethyl)piperazine-1- carboxylate (290 mg, 430.44 μmol, 26.36% yield) as a light yellow solid. Step C. Procedure for preparation of tert-butyl (2S)-4-[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]-2-(trifluoromethyl)piperazine-1-carboxy late To a solution of tert-butyl (2S)-4-[3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazol-6-yl] -2- (trifluoromethyl)piperazine-1-carboxylate (290 mg, 430.44 μmol, 1 equiv.) in THF (5 mL) and EtOH (5 mL) was added Pd/C (150 mg, 140.95 μmol, 10% purity, 0.327 equiv.) and Pd(OH) 2 (150 mg, 1.07 mmol, 2.48 equiv.) under N 2 atmosphere. The suspension was degassed and purged with H2 three times. The mixture was stirred under H2 (15 Psi) at 40 °C for 16 hours. The reaction mixture was filtered and washed with THF (50 mL). The filtrate was concentrated to give tert-butyl (2S)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]-2- (trifluoromethyl)piperazine-1-carboxylate (260 mg, crude) as a yellow oil, which was used in the next step without further purification. MS (ESI) m/z: 496.3 [M+H] + . Step D. Procedure for preparation of 3-[1-methyl-6-[(3S)-3-(trifluoromethyl)piperazin-1- yl]indazol-3-yl]piperidine-2,6-dione A solution of tert-butyl (2S)-4-[3-(2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]-2- (trifluoromethyl)piperazine-1-carboxylate (260.00 mg, 524.73 μmol, 1.0 equiv.) in HCl/dioxane (10 mL) was stirred at 25 °C for 16 hours. The reaction mixture was filtered to give 3-[1-methyl-6-[(3S)-3-(trifluoromethyl)piperazin-1-yl]indazo l-3-yl]piperidine-2,6-dione (170 mg, crude) as a light-yellow solid, which was used in the next step without further purification. MS (ESI) m/z: 396.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.87 (s, 1H), 7.59 (d, J = 8.8 Hz, 1H), 7.11 (s, 1H), 7.01 (dd, J = 1.6, 8.8 Hz, 1H), 4.72 - 4.60 (m, 1H), 4.30 (dd, J = 5.2, 9.6 Hz, 1H), 4.03 (d, J = 11.2 Hz, 1H), 3.94 (s, 3H), 3.90 (d, J = 12.4 Hz, 1H), 3.48 - 3.42 (m, 1H), 3.36 - 3.18 (m, 3H), 2.72 - 2.55 (m, 2H), 2.39 - 2.27 (m, 1H), 2.22 - 2.10 (m, 1H) Step E. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(4-((2S)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-2-(trifluoromethyl)piperazin-1-yl)bu tyl)cyclohexyl)oxy)-2- methylphenyl)picolinate To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2-methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)o xy)phenyl)picolinate (100 mg, 134.24 μmol, 1.0 equiv.) and 3-[1-methyl-6-[(3S)-3-(trifluoromethyl)piperazin-1-yl]indazo l-3- yl]piperidine-2,6-dione (58.38 mg, 147.66 μmol, 1.1 equiv.) in DCM (5 mL) was added NaBH(OAc)3 (85.35 mg, 402.72 μmol, 3.0 equiv.). The mixture was stirred at 25 °C for 16 hours. The reaction mixture was then diluted with H 2 O (30 mL) and extracted with ethyl acetate (20 mL × 3). The combined organic layers were washed with brine (50 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r) -4-(4-((2S)-4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)-2-(trifluorom ethyl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (150 mg, crude) as a light yellow solid, which was used in the next step without further purification. MS (ESI) m/z: 1125.8 [M+H] + . Step F. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(4-((2S)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-2-(trifluoromethyl)piperazin-1-yl)bu tyl)cyclohexyl)oxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(4-((2S)-4-(3-(2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-6-yl)- 2-(trifluoromethyl)piperazin-1-yl)butyl)cyclohexyl)oxy)-2-me thylphenyl)picolinate (150 mg, 133.41 μmol, 1.0 equiv.) in DCM (3 mL) was added TFA (9.21 g, 80.77 mmol, 6 mL, 605.43 equiv.). The mixture was stirred at 30 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1r,4r)-4-(4-((2S)- 4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)-2-( trifluoromethyl)piperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (56.36 mg, 51.33 μmol, 38.47% yield) as a white solid. MS (ESI) m/z: 1068.3 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.91 - 12.81 (m, 1H), 10.85 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.53 - 7.48 (m, 1H), 7.48 - 7.42 (m, 3H), 7.39 - 7.32 (m, 2H), 7.10 - 7.03 (m, 1H), 6.94 (dd, J = 8.4, 18.4 Hz, 2H), 6.87 - 6.81 (m, 2H), 6.61 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.26 (dd, J = 5.2, 9.2 Hz, 1H), 4.23 - 4.14 (m, 1H), 3.93 - 3.88 (m, 5H), 3.63 (d, J = 10.0 Hz, 2H), 3.16 - 3.06 (m, 2H), 3.02 (t, J = 5.6 Hz, 2H), 2.90 - 2.81 (m, 1H), 2.74 (s, 2H), 2.65 - 2.57 (m, 2H), 2.33 - 2.27 (m, 1H), 2.15 (dd, J = 5.2, 13.2 Hz, 1H), 2.07 (d, J = 9.6 Hz, 2H), 1.92 - 1.85 (m, 3H), 1.78 (d, J = 11.6 Hz, 2H), 1.52 - 1.45 (m, 2H), 1.43 - 1.13 (m, 9H), 1.09 – 1.01 (m, 2H) Example 296. Preparation of Compound 347a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3- ((2R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)-2-(trifluoromethyl)piperazin- 1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-((2R)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-2-(trifluoromethyl)piperazin-1-yl)pr opyl)cyclohexyl)oxy)-2- methylphenyl)picolinate To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2-methyl-3-(((1r,4r)-4-(3-oxopropyl)cyclohexyl) oxy)phenyl)picolinate (40 mg, 54.73 μmol, 1 equiv.) in DCM (2 mL) was added 3-[1-methyl-6-[(3R)-3- (trifluoromethyl)piperazin-1-yl]indazol-3-yl]piperidine-2,6- dione (25.97 mg, 65.67 μmol, 1.2 equiv.), and NaBH(OAc) 3 (57.99 mg, 273.63 μmol, 5 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH = 15:1) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3- ((2R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)-2-(trifluoromethyl)piperazin- 1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (40 mg, crude) as a yellow oil. MS (ESI) m/z: 1110.6[M+H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-((2R)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-2-(trifluoromethyl)piperazin-1-yl)pr opyl)cyclohexyl)oxy)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3-((2R)-4-(3-(2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-6- yl)-2-(trifluoromethyl)piperazin-1-yl)propyl)cyclohexyl)oxy) -2-methylphenyl)picolinate (40 mg, 36.03 μmol, 1 equiv.) in DCM (1 mL) was added TFA (1 mL). The mixture was stirred at 25 °C for 16 hours. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-((2R)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-2-(trifluoromethyl)piperazin-1-yl)pr opyl)cyclohexyl)oxy)-2- methylphenyl)picolinic acid (16.26 mg, 15.13 μmol, 41.99 % yield) as an off-white solid. MS (ESI) m/z: 1054.5[M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 10.85 (s, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.84 - 7.74 (m, 1H), 7.62 ( d, J = 7.6 Hz, 1H), 7.54 - 7.41 (m, 4H), 7.40 - 7.31 (m, 2H), 7.10 - 7.03 (m, 1H), 6.97 - 6.88 (m, 2H), 6.88 - 6.76 (m, 2H), 6.62 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.30 - 4.14 (m, 2H), 3.94 - 3.88 (m, 4H), 3.63 (d, J = 12.0 Hz, 2H), 3.14 - 2.99 (m, 5H), 2.85 (d, J = 7.6 Hz, 1H), 2.74 - 2.69 (m, 2H), 2.63 - 2.58 (m, 2H), 2.29 (dd, J = 4.4, 9.6 Hz, 1H), 2.20 - 2.00 (m, 4H), 1.87 (s, 3H), 1.80 (d, J = 12.0 Hz, 2H), 1.50 (d, J = 6.0 Hz, 2H), 1.43 - 1.29 (m, 3H), 1.26 - 1.19 (m, 3H), 1.12 - 1.01 (m, 2H). Example 297. Preparation of Compound 347b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(3- ((2S)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)-2-(trifluoromethyl)piperazin- 1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-((2S)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-2-(trifluoromethyl)piperazin-1-yl)pr opyl)cyclohexyl)oxy)-2- methylphenyl)picolinate To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2-methyl-3-(((1r,4r)-4-(3-oxopropyl)cyclohexyl) oxy)phenyl)picolinate (100 mg, 136.82 μmol, 1.0 equiv.) and 3-(1-methyl-6-((S)-3-(trifluoromethyl)piperazin-1-yl)-1H- indazol-3-yl)piperidine-2,6-dione (64.99 mg, 150.50 μmol, 1.1 equiv., HCl) in DCM (1 mL) was added NaBH(OAc) 3 (86.99 mg, 410.45 μmol, 3.0 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , DCM: MeOH= 15:1) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1r,4r)-4-(3-((2S)- 4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)-2-( trifluoromethyl)piperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (74 mg, 66.65 μmol, 48.71% yield) as a yellow solid. MS (ESI) m/z: 555.9 [M/2+H] + . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-((2S)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-2-(trifluoromethyl)piperazin-1-yl)pr opyl)cyclohexyl)oxy)-2- methylphenyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1r,4r)-4-(3-((2S)-4-(3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-6-yl)-2- (trifluoromethyl)piperazin-1-yl)propyl)cyclohexyl)oxy)-2-met hylphenyl)picolinate (74 mg, 66.65 μmol, 1.0 equiv.) in TFA (1 mL) and DCM (1 mL) was stirred at 25 °C for 16 hours. The reaction mixture was concentrated under reduced pressure to remove solvent to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(3-((2S)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-2-(trifluoromethyl)piperazin-1-yl)pr opyl)cyclohexyl)oxy)-2- methylphenyl)picolinic acid (27.59 mg, 26.17 μmol, 39.27% yield) as a white solid. MS (ESI) m/z: 1054.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 13.05 - 12.69 (m, 1H), 10.90 - 10.83 (m, 1H), 8.04 (d, J = 7.6 Hz, 1H), 7.79 (d, J = 8.4 Hz, 1H), 7.63 (d, J = 7.6 Hz, 1H), 7.54 - 7.43 (m, 4H), 7.41 - 7.33 (m, 2H), 7.11 - 7.05 (m, 1H), 6.99 - 6.91 (m, 2H), 6.88 - 6.82 (m, 2H), 6.62 (d, J = 7.2 Hz, 1H), 4.98 (s, 2H), 4.29 - 4.17 (m, 2H), 3.96 - 3.88 (m, 6H), 3.43 (m, 1H), 3.37 - 3.31 (m, 1H), 3.15 - 3.01 (m, 4H), 2.94 - 2.83 (m, 1H), 2.79 - 2.71 (m, 2H), 2.63 - 2.59 (m, 2H), 2.35 - 2.29 (m, 1H), 2.20 - 2.05 (m, 3H), 1.87 (s, 3H), 1.85 - 1.73 (m, 2H), 1.58 - 1.47 (m, 2H), 1.45 - 1.19 (m, 6H), 1.15 - 1.00 (m, 2H) Example 298. Preparation of Compound 349a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4- ((4S)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 7-yl)piperazin-1- yl)pentyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1R,4r)-4- ((4S*)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -7-yl)piperazin-1- yl)pentyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-7-(4- ((S)-5-((1r,4r)-4-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2- yl)phenoxy)cyclohexyl)pentan-2-yl)piperazin-1-yl)-1H-indazol e To a solution of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-7-(4-((S)-5-((1r,4R)-4-(3 -bromo-2- methylphenoxy)cyclohexyl)pentan-2-yl)piperazin-1-yl)-1-methy l-1H-indazole (200 mg, 237.28 μmol, 1.0 equiv.) in dioxane (2 mL) was added Pd(dppf)Cl2 (17.36 mg, 23.73 μmol, 0.1 equiv.), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (180.76 mg, 711.83 μmol, 3.0 equiv.), and KOAc (69.86 mg, 711.83 μmol, 3.0 equiv.). The mixture was stirred at 90 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was diluted with H2O (50 mL) and extracted with ethyl acetate 100 mL (50 mL × 2). The combined organic layers were washed with ethyl acetate 30 mL, dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, petroleum ether / ethyl acetate = 3 / 1) to give 3-(2,6- bis(benzyloxy)pyridin-3-yl)-1-methyl-7-(4-((S)-5-((1r,4r)-4- (2-methyl-3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)cyclohexyl)penta n-2-yl)piperazin-1-yl)-1H- indazole (170 mg, 181.47 μmol, 76.48 % yield) as a yellow oil. MS (ESI) m/z: 890.5 [M+H] + . Step B. Procedure for preparation of 3-(1-methyl-7-(4-((S)-5-((1r,4r)-4-(2-methyl-3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)cyclohe xyl)pentan-2-yl)piperazin- 1-yl)-1H-indazol-3-yl)piperidine-2,6-dione A mixture of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-7-(4-((S)-5-((1r ,4R)-4-(2-methyl-3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)cyclohe xyl)pentan-2-yl)piperazin-1- yl)-1H-indazole (170 mg, 191.02 μmol, 1.0 equiv.), Pd/C (40.66 mg, 38.20 μmol, 10 % purity, 0.2 equiv.), EtOH (2 mL), and Pd(OH) 2 (26.83 mg, 38.20 μmol, 20 % purity, 0.2 equiv.) in THF (2 mL) was stirred at 40 °C for 12 hours under H 2 atmosphere. The mixture was then filtered and washed with THF (50 mL). The filtrate was concentrated under reduced pressure to give 3-(1-methyl-7-(4-((S)-5-((1r,4r)-4-(2-methyl-3-(4,4,5,5-tetr amethyl-1,3,2- dioxaborolan-2-yl)phenoxy)cyclohexyl)pentan-2-yl)piperazin-1 -yl)-1H-indazol-3- yl)piperidine-2,6-dione (140 mg, 169.16 μmol, 88.56 % yield) as a yellow solid, which was used in the next step without further purification. MS (ESI) m/z: 712.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.89 (s, 1H), 7.44 - 7.34 (m, 1H), 7.21 - 7.13 (m, 1H), 7.11 - 7.01 (m, 4H), 5.76 (s, 1H), 4.34 (dd, J = 4.8, 9.2 Hz, 1H), 4.28 - 4.11 (m, 5H), 3.93 (s, 1H), 2.90 (s, 2H), 2.74 (s, 2H), 2.69 - 2.60 (m, 4H), 2.38 - 2.31 (m, 2H), 2.30 (s, 3H), 2.19 (s, 3H), 2.08 - 2.01 (m, 2H), 1.82 - 1.75 (m, 2H), 1.56 - 1.44 (m, 2H), 1.26 - 1.19 (m, 5H), 1.17 (s, 1H), 1.09 - 1.01 (m, 12H), 0.99 (d, J = 6.4 Hz, 3H) Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-((4S)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)pentyl)cyclohexyl)oxy) -2- methylphenyl)picolinate A mixture of 3-(1-methyl-7-(4-((S)-5-((1r,4R)-4-(2-methyl-3-(4,4,5,5-tetr amethyl-1,3,2- dioxaborolan-2-yl)phenoxy)cyclohexyl)pentan-2-yl)piperazin-1 -yl)-1H-indazol-3- yl)piperidine-2,6-dione (140 mg, 196.70 μmol, 1.0 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-bromopicolin ate (133.48 mg, 236.04 μmol, 1.2 equiv.), KF (34.29 mg, 590.10 μmol, 13.82 μL, 3.0 equiv.), [2-(2- aminophenyl)phenyl]palladium(1+)bis(1-adamantyl)-butyl-phosp hanemethanesulfonate (28.65 mg, 39.34 μmol, 0.2 equiv.), and H 2 O (0.2 mL) in dioxane (2 mL) was degassed and purged with N2 three times. The mixture was stirred at 100 °C for 1 hour under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was diluted with H 2 O (30 mL) and extracted with ethyl acetate 100 mL (50 mL × 2). The combined organic layers were washed with brine 30 mL, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH = 20 : 1) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-((4S)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)pentyl)cyclohexyl)oxy) -2-methylphenyl)picolinate (80 mg, 66.52 μmol, 33.82 % yield) as a yellow oil. MS (ESI) m/z: 1070.6 [M+H] + . Step D. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-((4S)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)pentyl)cyclohexyl)oxy) -2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3- (((1R,4r)-4-((4S*)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-7-yl)piperazin- 1-yl)pentyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-((4S)-4-(4-(3-(2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-7- yl)piperazin-1-yl)pentyl)cyclohexyl)oxy)-2-methylphenyl)pico linate (80 mg, 74.74 μmol, 1.0 equiv.) in DCM (0.5 mL) was added TFA (1 mL). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-((4S)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)pentyl)cyclohexyl)oxy) -2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1R,4r)- 4-((4S*)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indaz ol-7-yl)piperazin-1- yl)pentyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] (29.37 mg, 28.38 μmol, 37.97 % yield) as a white solid. MS (ESI) m/z: 1014.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 12.96 - 12.77 (m, 1H), 12.73 - 12.41 (m, 1H), 10.89 (s, 1H), 8.04 (d, J = 7.2 Hz, 1H), 7.80 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 7.6 Hz, 1H), 7.50 - 7.34 (m, 6H), 7.10 - 6.91 (m, 5H), 6.62 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.38 - 4.32 (m, 1H), 4.25 (s, 3H), 4.22 - 4.16 (m, 1H), 3.92 (s, 2H), 3.03 (s, 2H), 2.73 - 2.55 (m, 5H), 2.44 - 2.29 (m, 4H), 2.22 - 2.01 (m, 4H), 1.93 (s, 1H), 1.88 (s, 3H), 1.84 - 1.71 (m, 3H), 1.64 - 1.50 (m, 1H), 1.44 - 1.20 (m, 10H), 1.13 - 1.05 (m, 3H). Example 299. Preparation of Compound 349b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1S,4r)-4- ((4R)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 7-yl)piperazin-1- yl)pentyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1S,4r)-4- ((4R*)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -7-yl)piperazin-1- yl)pentyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of 3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-7- piperazin-1-yl-indazole A mixture of 7-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (2.5 g, 5.00 mmol, 1.0 equiv.), piperazine (1.29 g, 14.99 mmol, 3.0 equiv.), Pd 2 (dba) 3 (457.51 mg, 499.61 μmol, 0.1 equiv.), NaOtBu (960.26 mg, 9.99 mmol, 2.0 equiv.), and tritert-butylphosphane (2.02 g, 999.23 μmol, 2.35 mL, 10% purity, 0.2 equiv.) in toluene (100 mL) was degassed and purged with N 2 three times. The mixture was stirred at 100 °C for 4 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (10~30% MeOH /DCM) to give 3-(2,6- dibenzyloxy-3-pyridyl)-1-methyl-7-piperazin-1-yl-indazole (1.5 g, 2.88 mmol, 57.64% yield) as a yellow oil. MS (ESI) m/z: 506.4 [M+H] + . Step B. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-7-(4-(5-((1r,4s)- 4-(3-bromo-2-methylphenoxy) cyclohexyl)pentan-2-yl)piperazin-1-yl)-1-methyl-1H- indazole To a solution of 5-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)pentan-2-on e (1.5 g, 4.25 mmol, 1.0 equiv.) in DCM (30 mL) was added NaBH(OAc)3 (3.60 g, 16.98 mmol, 4.0 equiv.) and 3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-7-piperazin-1-yl-inda zole (2.15 g, 4.25 mmol, 1.0 equiv.). The mixture was stirred at 25 °C for 12 hours. Then the reaction mixture was concentrated under reduced pressure to give a residue. The residue was diluted with H2O (100 mL) and extracted with DCM (50 mL × 3). The combined organic layers were washed with DCM (50 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~28% ethyl acetate/petroleum ether) to give 3-(2,6-bis(benzyloxy)pyridin-3-yl)-7-(4-(5-((1r,4s)-4-(3- bromo-2-methylphenoxy) cyclohexyl)pentan-2-yl)piperazin-1-yl)-1-methyl-1H-indazole (800 mg, 892.16 μmol, 21.01% yield) as a yellow solid. MS (ESI) m/z: 844.3 [M+H] + . Step C. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-7-(4-((S)-5- ((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)pentan-2-yl)p iperazin-1-yl)-1-methyl- 1H-indazole and 3-(2,6-bis(benzyloxy)pyridin-3-yl)-7-(4-((R)-5-((1r,4s)-4-(3 -bromo-2- methylphenoxy)cyclohexyl)pentan-2-yl)piperazin-1-yl)-1-methy l-1H-indazole The stereoisomers of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-7-(4-(5-((1r,4s)-4-(3-bro mo-2- methylphenoxy) cyclohexyl)pentan-2-yl)piperazin-1-yl)-1-methyl-1H-indazole (800 mg, 892.16 μmol, 1.0 equiv.) were separated by SFC to give 3-(2,6-bis(benzyloxy)pyridin-3-yl)- 7-(4-((S)-5-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)p entan-2-yl)piperazin-1-yl)-1- methyl-1H-indazole (450 mg, 533.87 μmol, 56.25% yield) as a yellow solid and 3-(2,6- bis(benzyloxy)pyridin-3-yl)-7-(4-((R)-5-((1r,4s)-4-(3-bromo- 2- methylphenoxy)cyclohexyl)pentan-2-yl)piperazin-1-yl)-1-methy l-1H-indazole (300 mg, 336.34 μmol, 35.44% yield) as a yellow solid. Step D. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-7-(4- ((R)-5-((1r,4s)-4-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2- yl)phenoxy)cyclohexyl)pentan-2-yl)piperazin-1-yl)-1H-indazol e A mixture of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-7-(4-((R)-5-((1r,4S)-4-(3 -bromo-2- methylphenoxy)cyclohexyl)pentan-2-yl)piperazin-1-yl)-1-methy l-1H-indazole (300 mg, 355.91 μmol, 1.0 equiv.), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (271.14 mg, 1.07 mmol, 3.0 equiv.), KOAc (104.79 mg, 1.07 mmol, 3.0 equiv.), and Pd(dppf)Cl 2 (52.08 mg, 71.18 μmol, 0.2 equiv.) in dioxane (3 mL) was degassed and purged with N2 three times. The mixture was stirred at 90 °C for 12 hours under N2 atmosphere. To this solution, ethyl acetate (100 mL) and water (100 mL) were added, and the layers were separated. The aqueous was extracted with ethyl acetate (30 mL × 2), dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum. The residue was purified by prep-TLC (SiO2, petroleum ether : ethyl acetate = 3:1) to give 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-7-(4-((R)-5-((1r ,4s)- 4-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)p henoxy)cyclohexyl)pentan-2- yl)piperazin-1-yl)-1H-indazole (300 mg, 323.17 μmol, 90.80% yield) as a yellow solid. MS (ESI) m/z: 890.8 [M+H] + Step E. Procedure for preparation of 3-(1-methyl-7-(4-((R)-5-((1r,4s)-4-(2-methyl-3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)cyclohe xyl)pentan-2-yl)piperazin- 1-yl)-1H-indazol-3-yl)piperidine-2,6-dione A mixture of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-7-(4-((R)-5-((1r ,4s)-4-(2-methyl-3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)cyclohe xyl)pentan-2-yl)piperazin-1- yl)-1H-indazole (300 mg, 337.09 μmol, 1.0 equiv.) in THF (3 mL) and EtOH (3 mL) was added Pd/C (100 mg, 93.97 μmol, 10% purity) and Pd(OH) 2 (100 mg, 142.41 μmol, 20% purity). The mixture was stirred at 40 °C for 16 hours under H2 atmosphere (15 Psi). The reaction mixture was filtered and concentrated under reduced pressure to give a filter cake with a little EtOH. The collected filtrate was concentrated under reduced pressure to give 3-(1-methyl-7-(4-((R)- 5-((1r,4s)-4-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxabor olan-2- yl)phenoxy)cyclohexyl)pentan-2-yl)piperazin-1-yl)-1H-indazol -3-yl)piperidine-2,6-dione (250 mg, 333.69 μmol, 98.99% yield) as a pink solid. 1 H NMR (400 MHz, DMSO) δ = 10.88 (s, 1H), 7.39 - 7.37 (m, 1H), 7.18 - 7.16 (m, 1H), 7.12 - 7.08 (m, 1H), 7.08 - 7.06 (m, 1H), 7.04 - 6.98 (m, 2H), 4.37 - 4.30 (m, 1H), 4.24 (s, 3H), 4.20 - 4.11 (m, 1H), 3.20 - 2.98 (m, 2H), 2.83 - 2.55 (m, 8H), 2.38 - 2.30 (m, 1H), 2.29 (s, 3H), 2.20 - 2.12 (m, 1H), 2.07 - 1.98 (m, 2H), 1.83 - 1.72 (m, 2H), 1.55 - 1.45 (m, 1H), 1.38 - 1.32 (m, 4H), 1.29 (s, 12H), 1.27 - 1.18 (m, 5H), 1.03 (d, J = 13.2 Hz, 2H), 0.98 (d, J = 6.4 Hz, 3H) Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-((4R)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)pentyl)cyclohexyl)oxy) -2- methylphenyl)picolinate A mixture of 3-(1-methyl-7-(4-((R)-5-((1r,4s)-4-(2-methyl-3-(4,4,5,5-tetr amethyl-1,3,2- dioxaborolan-2-yl)phenoxy)cyclohexyl)pentan-2-yl)piperazin-1 -yl)-1H-indazol-3- yl)piperidine-2,6-dione (143.01 mg, 252.90 μmol, 1.2 equiv.), KF (36.73 mg, 632.25 μmol, 14.81 μL, 3.0 equiv.), and H2O (0.4 mL) in dioxane (4 mL) was degassed and purged with N2 three times. To this mixture was added [2-(2-aminophenyl)phenyl]palladium(1+);bis(1- adamantyl)-butyl-phosphane;methanesulfonate (30.70 mg, 42.15 μmol, 0.2 equiv.). The resulting mixture was stirred at 100 °C for 1 hour under N 2 atmosphere. Ethyl acetate (100 mL) and water (100 mL) were added, and the layers were separated. The organic layers were washed with brine (30 mL × 2), dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum. The residue was purified by prep-TLC (SiO 2 , DCM: MeOH = 20:1) to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4- ((4R)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 7-yl)piperazin-1- yl)pentyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (120 mg, 98.66 μmol, 46.81% yield) as a yellow solid. MS (ESI) m/z: 536.2 [1/2M+H] +. Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-((4R)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperazin-1-yl)pentyl)cyclohexyl)oxy) -2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3- (((1S,4r)-4-((4R*)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-7-yl)piperazin- 1-yl)pentyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1s,4r)-4-((4R)-4-(4-(3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-7- yl)piperazin-1-yl)pentyl)cyclohexyl)oxy)-2-methylphenyl)pico linate (120 mg, 112.11 μmol, 1.0 equiv.) in TFA (2 mL) and DCM (1 mL) was stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1S,4r)-4-((4R)-4-(4-(3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-7- yl)piperazin-1-yl)pentyl)cyclohexyl)oxy)-2-methylphenyl)pico linic acid [6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1S,4r)-4-((4R*)-4- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)pip erazin-1- yl)pentyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] (39.51 mg, 37.88 μmol, 33.78% yield) as a white solid. MS (ESI) m/z: 1014.5 [M+H] + . 1 H NMR (400 MHz, DMSO) δ = 12.90 - 12.81 (m, 1H), 10.88 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.49 - 7.33 (m, 6H), 7.10 - 7.02 (m, 3H), 6.97 - 6.91 (m, 2H), 6.61 (d, J = 7.6 Hz, 1H), 4.98 (s, 2H), 4.38 - 4.31 (m, 1H), 4.24 (s, 3H), 4.22 - 4.14 (m, 1H), 3.91 (t, J = 6.0 Hz, 2H), 3.24 - 3.13 (m, 2H), 3.02 (t, J = 5.6 Hz, 2H), 2.96 - 2.73 (m, 4H), 2.70 - 2.59 (m, 3H), 2.41 - 2.25 (m, 2H), 2.21 - 2.14 (m, 1H), 2.12 - 2.05 (m, 2H), 1.87 (s, 3H), 1.82 - 1.76 (m, 2H), 1.66 - 1.51 (m, 1H), 1.47 - 1.17 (m, 10H), 1.11 - 1.00 (m, 4H) Example 300. Preparation of Compound 350a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pip erazin-1-yl)-4- methylpentyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of 4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butanoic acid To a solution of 4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butanal (670 mg, 1.97 mmol, 1.0 equiv.) in THF (6 mL), was added t-BuOH (10 mL) and H 2 O (3 mL), NaH 2 PO 4 (710.81 mg, 5.92 mmol, 3.0 equiv.), 2-methylbut-2-ene (831.01 mg, 11.85 mmol, 1.26 mL, 6.0 equiv.) and NaClO2 (535.83 mg, 5.92 mmol, 3.0 equiv.). The mixture was stirred at 20 °C for 16 hours under O 2 (15 Psi). The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~6% ethyl acetate/petroleum ether) to give 4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butanoic acid (560 mg, crude) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 11.97 (s, 1H), 7.16 - 7.12 (m, 1H), 7.09 - 7.04 (m, 1H), 7.03 - 6.99 (m, 1H), 4.27 - 4.18 (m, 1H), 2.24 - 2.13 (m, 5H), 2.08 - 2.00 (m, 2H), 1.76 (d, J = 11.2 Hz, 2H), 1.56 - 1.46 (m, 2H), 1.39 - 1.29 (m, 2H), 1.28 - 1.23 (m, 1H), 1.21 - 1.15 (m, 2H), 1.09 - 0.97 (m, 2H) Step B. Procedure for preparation of 1-(4-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1-yl)-4-((1r,4s)-4-(3-bromo-2- methylphenoxy)cyclohexyl)butan-1-one To a solution of 4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butanoic acid (560 mg, 1.58 mmol, 1 equiv.) in DMF (6 mL) was added HATU (719.22 mg, 1.89 mmol, 1.2 equiv.), TEA (478.51 mg, 4.73 mmol, 658.20 μL, 3.0 equiv.), and 3-(2,6-bis(benzyloxy)pyridin-3-yl)- 1-methyl-6-(piperazin-1-yl)-1H-indazole (916.53 mg, 1.81 mmol, 1.15 equiv.). The mixture was stirred at 20 °C for 0.5 hours. The mixture was poured into H2O (20 mL), filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~50% ethyl acetate/petroleum ether) to give 1-(4-(3-(2,6- bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-6-yl)piperaz in-1-yl)-4-((1r,4s)-4-(3- bromo-2-methylphenoxy)cyclohexyl)butan-1-one (1.07 g, 1.19 mmol, 75.54% yield) as a yellow oil. MS (ESI) m/z: 844.0 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.89 (d, J = 8.0 Hz, 1H), 7.53 (d, J = 8.8 Hz, 1H), 7.48 - 7.44 (m, 2H), 7.42 - 7.24 (m, 8H), 7.15 - 7.11 (m, 1H), 7.09 - 7.04 (m, 1H), 7.03 - 6.98 (m, 1H), 6.88 (s, 1H), 6.84 (d, J = 9.2 Hz, 1H), 6.56 (d, J = 8.0 Hz, 1H), 5.43 (d, J = 14.0 Hz, 4H), 4.28 - 4.18 (m, 1H), 3.97 (s, 3H), 3.62 (s, 4H), 3.22 (s, 2H), 3.17 (s, 2H), 2.37 - 2.32 (m, 2H), 2.20 (s, 3H), 2.08 - 2.00 (m, 2H), 1.77 (d, J = 12.0 Hz, 2H), 1.59 - 1.49 (m, 2H), 1.41 - 1.30 (m, 2H), 1.30 - 1.26 (m, 1H), 1.25 - 1.20 (m, 2H), 1.09 - 0.98 (m, 2H) Step C. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-(4-(5-((1r,4s)- 4-(3-bromo-2-methylphenoxy)cyclohexyl)-2-methylpentan-2-yl)p iperazin-1-yl)-1- methyl-1H-indazole A mixture of 1-(4-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol -6-yl)piperazin-1- yl)-4-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)butan-1 -one (500 mg, 593.22 μmol, 1.0 equiv.), and ZrCl 4 (165.89 mg, 711.86 μmol, 59.25 μL, 1.2 equiv.) in THF (10 mL) was degassed and purged with N2 three times. To this solution, MeMgBr (2.5 M, 1.66 mL, 7.0 equiv.) was added dropwise at -10 °C. The resulting mixture was stirred at -10 °C for 15 minutes under N 2 atmosphere. The reaction mixture was poured into saturated NH 4 Cl aq. (40 mL), and then extracted was ethyl acetate 120 mL (40 mL × 3). The combined organic layers were washed with brine 50 mL, dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~50% ethyl acetate/petroleum ether) to give 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-(4-(5-((1r,4s)-4- (3-bromo-2-methylphenoxy)cyclohexyl)-2-methylpentan-2-yl)pip erazin-1-yl)-1-methyl-1H- indazole (60 mg, 65.29 μmol, 11.01% yield) as a yellow oil. MS (ESI) m/z: 858.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 7.88 (d, J = 8.4 Hz, 1H), 7.52 - 7.43 (m, 3H), 7.42 - 7.23 (m, 8H), 7.15 - 7.11 (m, 1H), 7.09 - 7.04 (m, 1H), 7.03 - 6.99 (m, 1H), 6.84 - 6.77 (m, 2H), 6.56 (d, J = 8.0 Hz, 1H), 5.44 (s, 2H), 5.41 (s, 2H), 4.27 - 4.18 (m, 1H), 3.96 (s, 3H), 3.20 - 3.14 (m, 4H), 2.63 (s, 4H), 2.20 (s, 3H), 2.05 (d, J = 12.8 Hz, 2H), 1.78 (d, J = 10.8 Hz, 2H), 1.41 - 1.30 (m, 6H), 1.23 (s, 1H), 1.20 - 1.17 (m, 2H), 1.09 - 1.02 (m, 1H), 1.00 (s, 6H) Step D. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-6-(4-(2- methyl-5-((1r,4s)-4-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-d ioxaborolan-2- yl)phenoxy)cyclohexyl)pentan-2-yl)piperazin-1-yl)-1H-indazol e A mixture of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-(4-(5-((1r,4s)-4-(3-bro mo-2- methylphenoxy)cyclohexyl)-2-methylpentan-2-yl)piperazin-1-yl )-1-methyl-1H-indazole (60 mg, 70.02 μmol, 1.0 equiv.), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (26.67 mg, 105.03 μmol, 1.5 equiv.), KOAc (34.36 mg, 350.09 μmol, 5.0 equiv.), and Pd(dppf)Cl 2 (10.25 mg, 14.00 μmol, 0.2 equiv.) in dioxane (1 mL) was degassed and purged with N 2 three times. The mixture was stirred at 100 °C for 16 hours under N2 atmosphere. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , ethyl acetate: methanol = 20:1) to give 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-6- (4-(2-methyl-5-((1r,4s)-4-(2-methyl-3-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)phenoxy)cyclohexyl)pentan-2-yl)piperazin-1-yl)-1H-indazol e (50 mg, 46.24 μmol, 66.05% yield) as a yellow oil. MS (ESI) m/z: 904.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.88 (d, J = 8.0 Hz, 1H), 7.51 - 7.44 (m, 3H), 7.42 - 7.33 (m, 5H), 7.32 - 7.26 (m, 3H), 7.19 - 7.15 (m, 1H), 7.12 - 7.04 (m, 2H), 6.83 - 6.79 (m, 2H), 6.56 (d, J = 8.0 Hz, 1H), 5.44 (s, 2H), 5.41 (s, 2H), 4.22 - 4.13 (m, 1H), 3.96 (s, 3H), 3.17 (s, 2H), 3.16 (s, 2H), 2.63 (s, 4H), 2.29 (s, 3H), 2.07 - 1.99 (m, 2H), 1.81 - 1.74 (m, 2H), 1.41 - 1.31 (m, 6H), 1.28 (s, 11H), 1.23 (s, 1H), 1.20 - 1.16 (m, 2H), 1.08 - 1.02 (m, 2H), 1.00 (s, 6H) Step E. Procedure for preparation of 3-(1-methyl-6-(4-(2-methyl-5-((1r,4s)-4-(2-methyl- 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)cyclo hexyl)pentan-2- yl)piperazin-1-yl)-1H-indazol-3-yl)piperidine-2,6-dione To a solution of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-6-(4-(2-methyl-5 -((1r,4s)-4-(2- methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenox y)cyclohexyl)pentan-2- yl)piperazin-1-yl)-1H-indazole (50 mg, 55.31 μmol, 1.0 equiv.) in EtOH (2 mL) and THF (2 mL) was added Pd/C (25.31 mg, 23.78 μmol, 10% purity, 0.43 equiv.) and Pd(OH) 2 (25.24 mg, 35.95 μmol, 20% purity, 0.65 equiv.) under N2 atmosphere. The suspension was degassed and purged with H2 three times. The mixture was stirred under H2 (15 psi) at 40 °C for 16 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , ethyl acetate: methanol = 20:1) to give 3-(1- methyl-6-(4-(2-methyl-5-((1r,4s)-4-(2-methyl-3-(4,4,5,5-tetr amethyl-1,3,2-dioxaborolan-2- yl)phenoxy)cyclohexyl)pentan-2-yl)piperazin-1-yl)-1H-indazol -3-yl)piperidine-2,6-dione (35 mg, 41.26 μmol, 74.60% yield) as a yellow oil. MS (ESI) m/z: 726.4 [M+H] + Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1-yl)-4-methylpentyl)cyclohexyl)ox y)-2- methylphenyl)picolinate A mixture of 3-(1-methyl-6-(4-(2-methyl-5-((1r,4s)-4-(2-methyl-3-(4,4,5,5 -tetramethyl-1,3,2- dioxaborolan-2-yl)phenoxy)cyclohexyl)pentan-2-yl)piperazin-1 -yl)-1H-indazol-3- yl)piperidine-2,6-dione (35 mg, 48.22 μmol, 1.0 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-bromopicolin ate (30.00 mg, 53.05 μmol, 1.1 equiv.), and KF (8.41 mg, 144.67 μmol, 3.39 μL, 3.0 equiv.) in dioxane (1 mL) and H2O (0.1 mL) was degassed and purged with N2 three times. To this solution, Ad2nBuP Pd G3 (7.02 mg, 9.64 μmol, 0.2 equiv.) was added, and the mixture was stirred at 100 °C for 2 hours under N 2 atmosphere. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH = 10: 1) to give tert-butyl 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1s,4r)-4-(4-(4-(3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin -1-yl)-4- methylpentyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (20 mg, 15.21 μmol, 31.54% yield) as a yellow solid. MS (ESI) m/z: 1084.8 [M+H] + Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1-yl)-4-methylpentyl)cyclohexyl)ox y)-2- methylphenyl)picolinic acid To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-6- yl)piperazin-1-yl)-4-methylpentyl)cyclohexyl)oxy)-2-methylph enyl)picolinate (20 mg, 18.44 μmol, 1.0 equiv.) in DCM (0.5 mL) was added TFA (767.50 mg, 6.73 mmol, 0.5 mL, 364.95 equiv.). The mixture was stirred at 25 °C for 16 hours. The mixture was concentrated under reduced pressure to give a residue, which was diluted with DCM (2 mL) and adjusted to pH = 7 with triethylamine. This resulting solution was concentrated to give a residue. The residue was purified by prep-HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)piperazin-1-yl)-4-methylpentyl)cyclohexyl)ox y)-2-methylphenyl)picolinic acid (17.26 mg, 16.50 μmol, 89.45% yield) as a yellow solid. MS (ESI) m/z: 1028.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.91 - 12.49 (m, 1H), 10.84 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.4 Hz, 1H), 7.62 (d, J = 7.2 Hz, 1H), 7.51 - 7.40 (m, 4H), 7.40 - 7.31 (m, 2H), 7.09 - 7.03 (m, 1H), 6.97 - 6.87 (m, 3H), 6.82 (s, 1H), 6.61 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.28 - 4.22 (m, 1H), 4.22 - 4.14 (m, 1H), 3.96 - 3.89 (m, 2H), 3.88 (s, 3H), 3.18 (s, 4H), 3.02 (t, J = 5.6 Hz, 2H), 2.70 - 2.60 (m, 6H), 2.31 - 2.24 (m, 1H), 2.18 - 2.12 (m, 1H), 2.11 - 2.04 (m, 2H), 1.86 (s, 3H), 1.81 - 1.74 (m, 2H), 1.42 - 1.29 (m, 7H), 1.10 - 1.03 (m, 2H), 1.09 - 1.03 (m, 2H), 1.00 (s, 6H) Example 301. Preparation of Compound 334a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1,7-dimethyl-1H-indazol-6-yl )piperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1,7- dimethyl-1H-indazol-6-yl)piperazin-1-yl)propyl)cyclohexyl)ox y)-2- methylphenyl)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(((1r,4r)-4-(3-oxopropyl)cyclohexyl)oxy)phenyl)pico linate (60 mg, 1 equiv., 82 μmol) and 3-(1,7-dimethyl-6-(piperazin-1-yl)-1H-indazol-3-yl)piperidin e-2,6-dione, HCl (34 mg, 1.1 equiv.90 μmol) were suspended in DCM (5 mL). To the mixture was added sodium triacetoxyborohydride (52 mg, 3 equiv., 0.25 mmol). The mixture was stirred at room temperature overnight. The mixture was then concentrated and extracted with DCM and water. The organic layer was separated, washed with brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1,7- dimethyl-1H-indazol-6-yl)piperazin-1-yl)propyl)cyclohexyl)ox y)-2-methylphenyl)picolinate, which was carried forward without further purification. MS (ESI) m/z: 528.9 [M+2H] 2+ . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1,7- dimethyl-1H-indazol-6-yl)piperazin-1-yl)propyl)cyclohexyl)ox y)-2- methylphenyl)picolinic acid tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1,7-dimethyl- 1H-indazol-6-yl)piperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (87 mg, 1 equiv., 82 μmol) was dissolved in DCM (4 mL) and TFA (1 mL). The mixture was stirred at room temperature overnight. The reaction mixture was concentrated, and then purified by RP-HPLC to afford 6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2( 1H)-yl)-3-(3-(((1s,4r)-4-(3-(4- (3-(2,6-dioxopiperidin-3-yl)-1,7-dimethyl-1H-indazol-6-yl)pi perazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (47 mg, 47 μmol, 57 %) as a white solid. MS (ESI) m/z: 1000.4 [M+H] + ; 1 H NMR (400 MHz, DMSO-d6) δ 10.79 (s, 1H), 7.96 (dd, J = 7.9, 1.2 Hz, 1H), 7.72 (d, J = 8.0 Hz, 1H), 7.56 (dd, J = 7.7, 1.4 Hz, 1H), 7.44 – 7.35 (m, 4H), 7.29 (ddd, J = 10.4, 8.8, 7.4 Hz, 2H), 7.01 (t, J = 7.9 Hz, 1H), 6.87 (dd, J = 15.4, 8.6 Hz, 3H), 6.55 (d, J = 7.5 Hz, 1H), 4.91 (s, 2H), 4.26 – 4.08 (m, 1H), 4.12 (s, 3H), 3.85 (t, J = 6.0 Hz, 2H), 2.96 (t, J = 6.0 Hz, 2H), 2.82 (s, 4H), 2.57 (s, 3H), 2.66 – 2.45 (m, 2H), 2.22 (td, J = 9.2, 4.6 Hz, 1H), 2.13 – 1.98 (m, 1H), 2.03 (s, 2H), 1.80 (s, 3H), 1.73 (d, J = 12.4 Hz, 2H), 1.30 (d, J = 11.2 Hz, 1H), 1.17 (t, J = 7.5 Hz, 2H), 1.00 (q, J = 12.0 Hz, 2H). Example 302. Preparation of Compound 335a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1,5-dimethyl-1H-indazol-6-yl )piperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1,5- dimethyl-1H-indazol-6-yl)piperazin-1-yl)propyl)cyclohexyl)ox y)-2- methylphenyl)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(((1r,4r)-4-(3-oxopropyl)cyclohexyl)oxy)phenyl)pico linate (60 mg, 1 equiv. 82 μmol) and 3-(1,5-dimethyl-6-(piperazin-1-yl)-1H-indazol-3-yl)piperidin e-2,6-dione, HCl (34 mg, 1.1 equiv.90 μmol) were suspended in DCM (5 mL). To the mixture was added sodium triacetoxyborohydride (52 mg, 3 equiv. 0.25 mmol). The mixture was stirred at room temperature overnight. The mixture was then concentrated and extracted with DCM and water. The organic layer was separated, washed with brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1,5- dimethyl-1H-indazol-6-yl)piperazin-1-yl)propyl)cyclohexyl)ox y)-2-methylphenyl)picolinate, which was carried forward without further purification. MS (ESI) m/z: 528.8 [M+2H] 2+ . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1,5- dimethyl-1H-indazol-6-yl)piperazin-1-yl)propyl)cyclohexyl)ox y)-2- methylphenyl)picolinic acid tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1,5-dimethyl- 1H-indazol-6-yl)piperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (87 mg, 1 equiv.82 μmol) was dissolved in DCM (4 mL). To the mixture was added TFA (1 mL). The mixture was stirred at room temperature overnight. The reaction mixture was concentrated, and then purified by RP-HPLC to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1,5-dimethyl- 1H-indazol-6-yl)piperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (51 mg, 51 μmol, 62 %) as a white solid. MS (ESI) m/z: 1000.6 [M+H] + ; 1 H NMR (400 MHz, DMSO-d 6 ) δ 10.77 (s, 1H), 7.95 (d, J = 1.3 Hz, 0H), 7.72 (d, J = 8.0 Hz, 1H), 7.55 (dd, J = 7.6, 1.4 Hz, 1H), 7.44 – 7.34 (m, 4H), 7.34 – 7.23 (m, 2H), 7.07 (s, 1H), 7.01 (t, J = 7.9 Hz, 1H), 6.87 (dd, J = 15.0, 8.5 Hz, 2H), 6.55 (d, J = 7.5 Hz, 1H), 4.91 (s, 2H), 4.16 (ddd, J = 28.0, 10.0, 5.6 Hz, 2H), 3.85 (d, J = 3.9 Hz, 5H), 2.96 (t, J = 6.0 Hz, 2H), 2.88 (s, 4H), 2.65 – 2.45 (m, 2H), 2.24 (s, 3H), 2.13 – 2.02 (m, 1H), 2.03 (s, 1H), 2.00 (s, 1H), 1.80 (s, 3H), 1.74 (d, J = 12.4 Hz, 2H), 1.46 (s, 2H), 1.30 (d, J = 9.2 Hz, 2H), 1.18 (t, J = 7.3 Hz, 3H), 0.99 (dt, J = 20.5, 10.1 Hz, 2H). Example 303. Preparation of Compound 336a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3- (4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)-2, 2-dimethylpiperazin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-2,2-dimethylpiperazin-1-yl)propyl)cy clohexyl)oxy)-2- methylphenyl)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(((1r,4r)-4-(3-oxopropyl)cyclohexyl)oxy)phenyl)pico linate (60 mg, 1 equiv. 82 μmol) and 3-(6-(3,3-dimethylpiperazin-1-yl)-1-methyl-1H-indazol-3-yl)p iperidine-2,6-dione, HCl (35 mg, 1.1 equiv.90 μmol) were suspended in DCM (5 mL). To the mixture was added sodium triacetoxyborohydride (52 mg, 3 equiv.0.25 mmol). The mixture was stirred at room temperature overnight. The mixture was then concentrated and extracted with DCM and water. The organic layer was separated, washed with brine, dried over Na2SO4, filtered, and evaporated to dryness to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)-2,2-dimethylpiperazin-1-yl)propyl)cyclohexy l)oxy)-2- methylphenyl)picolinate, which was carried forward without further purification. MS (ESI) m/z: 535.9 [M+2H] 2+ . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6- dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)-2,2-dimethylpiperazin-1-yl)propyl)cy clohexyl)oxy)-2- methylphenyl)picolinic acid tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-i ndazol-6-yl)-2,2- dimethylpiperazin-1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl )picolinate (88 mg, 1 equiv. 82 μmol) was dissolved in DCM (4 mL). To the mixture was added TFA (1 mL). The mixture was stirred at room temperature overnight. The reaction mixture was concentrated, and then purified by RP-HPLC to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-(3-(2,6-dioxopiperidin-3-yl )-1-methyl-1H-indazol-6-yl)-2,2- dimethylpiperazin-1-yl)propyl)cyclohexyl)oxy)-2-methylphenyl )picolinic acid (20 mg, 20 μmol, 24 %) as a white solid. MS (ESI) m/z: 1014.6 [M+H] + ; 1 H NMR (400 MHz, DMSO- d6) δ 12.79 (s, 1H), 10.77 (s, 1H), 7.96 (d, J = 7.6 Hz, 1H), 7.72 (d, J = 8.0 Hz, 1H), 7.55 (d, J = 7.6 Hz, 1H), 7.44 – 7.35 (m, 3H), 7.35 – 7.24 (m, 2H), 7.01 (t, J = 7.9 Hz, 1H), 6.87 (dd, J = 15.7, 8.6 Hz, 3H), 6.76 (s, 1H), 6.54 (d, J = 7.4 Hz, 1H), 6.44 (s, 1H), 4.91 (s, 2H), 4.19 (dd, J = 9.2, 5.2 Hz, 1H), 4.13 (s, 1H), 3.85 (d, J = 5.3 Hz, 2H), 3.83 (s, 4H), 2.96 (t, J = 6.0 Hz, 2H), 2.88 (s, 2H), 2.56 (d, J = 8.5 Hz, 1H), 2.56 – 2.43 (m, 1H), 2.47 (s, 2H), 2.23 (d, J = 9.0 Hz, 1H), 2.09 (dd, J = 13.1, 6.0 Hz, 1H), 2.02 (d, J = 9.9 Hz, 3H), 1.80 (s, 3H), 1.73 (d, J = 12.4 Hz, 2H), 1.34 – 1.26 (m, 8H), 1.17 (s, 5H), 1.00 (d, J = 11.4 Hz, 7H). Example 304. Preparation of Compound 337a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3- (4-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)ox y)piperidin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-((3-(2,6 -dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)oxy)piperidin-1-yl)propyl)cyclohexyl) oxy)-2- methylphenyl)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(((1r,4r)-4-(3-oxopropyl)cyclohexyl)oxy)phenyl)pico linate (60 mg, 1 equiv. 82 μmol) and 3-(1-methyl-6-(piperidin-4-yloxy)-1H-indazol-3-yl)piperidine -2,6-dione, HCl (34 mg, 1.1 equiv.90 μmol) were suspended in DCM (5 mL). To the mixture was added sodium triacetoxyborohydride (52 mg, 3 equiv. 0.25 mmol). The mixture was stirred at room temperature overnight. The mixture was then concentrated and extracted with DCM and water. The organic layer was separated, washed with brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-((3-(2,6 -dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)oxy)piperidin-1-yl)propyl)cyclohexyl)oxy)-2- methylphenyl)picolinate, which was carried forward without further purification. MS (ESI) m/z: 529.3 [M+2H] 2+ . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-((3-(2,6 -dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)oxy)piperidin-1-yl)propyl)cyclohexyl) oxy)-2- methylphenyl)picolinic acid tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(3-(4-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)oxy)piperidin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (87 mg, 1 equiv.82 μmol) was dissolved in DCM (4 mL). To the mixture was added TFA (1 mL). The mixture was stirred at room temperature overnight. The reaction mixture was concentrated, and then purified by RP-HPLC to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(3-(4-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)oxy)piperidin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (40 mg, 40 μmol, 49 %) as a white solid. MS (ESI) m/z: 1001.5 [M+H] + ; 1 H NMR (400 MHz, DMSO-d 6 ) δ 10.79 (s, 1H), 7.96 (dd, J = 8.0, 1.2 Hz, 1H), 7.72 (d, J = 8.0 Hz, 1H), 7.55 (dd, J = 7.6, 1.4 Hz, 1H), 7.50 (d, J = 8.8 Hz, 1H), 7.44 – 7.34 (m, 3H), 7.34 – 7.23 (m, 2H), 7.06 – 6.96 (m, 2H), 6.87 (dd, J = 14.4, 8.5 Hz, 2H), 6.69 (dd, J = 8.7, 2.0 Hz, 1H), 6.55 (d, J = 7.5 Hz, 1H), 4.91 (s, 2H), 4.53 (s, 1H), 4.22 (dd, J = 9.5, 5.1 Hz, 1H), 4.12 (s, 1H), 3.85 (s, 5H), 3.84 (d, J = 5.2 Hz, 1H), 2.95 (t, J = 5.9 Hz, 2H), 2.85 (s, 2H), 2.61 – 2.43 (m, 3H), 2.32 – 2.18 (m, 1H), 2.15 – 1.97 (m, 1H), 2.00 (s, 5H), 1.80 (s, 3H), 1.72 (d, J = 12.8 Hz, 5H), 1.45 (d, J = 9.8 Hz, 3H), 1.30 (d, J = 7.7 Hz, 0H), 1.14 (t, J = 7.8 Hz, 2H), 0.99 (q, J = 12.1 Hz, 2H). Example 305. Preparation of Compound 338a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(3- (4-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)ox y)piperidin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-((3-(2,6 -dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)oxy)piperidin-1-yl)propyl)cyclohexyl) oxy)-2- methylphenyl)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(((1r,4r)-4-(3-oxopropyl)cyclohexyl)oxy)phenyl)pico linate (60 mg, 1 equiv. 82 μmol) and 3-(1-methyl-7-(piperidin-4-yloxy)-1H-indazol-3-yl)piperidine -2,6-dione, HCl (34 mg, 1.1 equiv.90 μmol) were suspended in DCM (5 mL). To the mixture was added sodium triacetoxyborohydride (52 mg, 3 equiv. 0.25 mmol). The mixture was stirred at room temperature overnight. The mixture was then concentrated and extracted with DCM and water. The organic layer was separated, washed with brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-((3-(2,6 -dioxopiperidin-3-yl)-1-methyl- 1H-indazol-7-yl)oxy)piperidin-1-yl)propyl)cyclohexyl)oxy)-2- methylphenyl)picolinate, which was carried forward without further purification. MS (ESI) m/z: 529.3 [M+2H] 2+ . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(3-(4-((3-(2,6 -dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)oxy)piperidin-1-yl)propyl)cyclohexyl) oxy)-2- methylphenyl)picolinic acid tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(3-(4-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H- indazol-7-yl)oxy)piperidin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (87 mg, 1 equiv.82 μmol) was dissolved in DCM (4 mL). To the mixture was added TFA (1 mL). The mixture was stirred at room temperature overnight. The reaction mixture was concentrated, and then purified by RP-HPLC to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(3-(4-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H- indazol-7-yl)oxy)piperidin-1- yl)propyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (10 mg, 10 μmol, 12 %) as a white solid. MS (ESI) m/z: 501.4 [M+2H] 2+ ; 1 H NMR (400 MHz, DMSO-d6) δ 10.80 (s, 1H), 7.96 (d, J = 7.5 Hz, 1H), 7.72 (d, J = 8.1 Hz, 1H), 7.55 (d, J = 7.6 Hz, 1H), 7.44 – 7.35 (m, 3H), 7.35 – 7.24 (m, 2H), 7.15 (d, J = 8.0 Hz, 1H), 7.00 (t, J = 7.9 Hz, 1H), 6.95 – 6.79 (m, 4H), 6.54 (d, J = 7.5 Hz, 1H), 6.44 (s, 1H), 4.91 (s, 2H), 4.25 (dd, J = 9.7, 5.1 Hz, 1H), 4.14 (s, 3H), 3.85 (t, J = 5.9 Hz, 2H), 3.10 (d, J = 5.0 Hz, 2H), 2.96 (s, 2H), 2.60 – 2.49 (m, 3H), 2.47 (s, 3H), 2.13 – 2.06 (m, 2H), 2.01 (s, 2H), 1.80 (s, 3H), 1.72 (d, J = 12.8 Hz, 3H), 1.29 (s, 1H), 1.16 (s, 2H), 1.00 (d, J = 12.1 Hz, 2H). Example 306. Preparation of Compound 345a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- ((2R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-y l)-2-methylpiperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-((2R)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)-2-methylpiperazin-1-yl)butyl)cyclohe xyl)oxy)-2- methylphenyl)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)oxy)phenyl)picol inate (60 mg, 1 equiv.81 μmol) and 3-(1-methyl-7-((R)-3-methylpiperazin-1-yl)-1H-indazol-3-yl)p iperidine-2,6-dione, HCl (37 mg, 1.2 equiv.97 μmol) were suspended in DCM (4 mL). To the mixture was added sodium triacetoxyborohydride (51 mg, 36 μL, 3 equiv. 0.24 mmol). The mixture was stirred at room temperature overnight. The mixture was then concentrated and extracted with DCM and water. The organic layer was separated, washed with brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-((2R)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)-2-methylpiperazin-1-yl)butyl)cyclohe xyl)oxy)-2- methylphenyl)picolinate, which was carried forward without further purification. Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-((2R)-4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)-2-methylpiperazin-1-yl)butyl)cyclohe xyl)oxy)-2- methylphenyl)picolinic acid tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(4-((2R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl -1H-indazol-7-yl)-2- methylpiperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)pi colinate (87 mg, 1 equiv. 81 μmol) was dissolved in DCM (4 mL). To the mixture was added TFA (1 mL). The mixture was stirred at room temperature overnight. The reaction mixture was concentrated, and then purified by RP-HPLC to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4-((2R)-4-(3-(2,6-dioxopiperidin -3-yl)-1-methyl-1H-indazol-7- yl)-2-methylpiperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphe nyl)picolinic acid (38 mg, 37 μmol, 46 %). MS (ESI) m/z: 507.9 [M+2H] 2+ ; 1 H NMR (400 MHz, DMSO-d 6 ) δ 10.81 (s, 1H), 7.96 (d, J = 7.8 Hz, 1H), 7.72 (d, J = 8.1 Hz, 1H), 7.56 (d, J = 7.5 Hz, 1H), 7.40 (t, J = 7.6 Hz, 1H), 7.39 (s, 1H), 7.37 (s, 1H), 7.34 (s, 1H), 7.29 (dt, J = 10.0, 7.7 Hz, 2H), 7.00 (t, J = 7.9 Hz, 1H), 6.96 (d, J = 5.1 Hz, 2H), 6.89 (d, J = 8.8 Hz, 1H), 6.85 (d, J = 8.3 Hz, 1H), 6.54 (d, J = 7.5 Hz, 1H), 4.91 (s, 2H), 4.27 (dd, J = 9.5, 5.1 Hz, 1H), 4.18 (s, 2H), 4.19 – 4.10 (m, 1H), 3.85 (t, J = 5.9 Hz, 2H), 3.09 (s, 3H), 2.96 (t, J = 5.9 Hz, 2H), 2.80 (s, 3H), 2.56 (s, 2H), 2.10 (dd, J = 13.4, 5.8 Hz, 1H), 2.01 (t, J = 7.2 Hz, 2H), 1.85 (d, J = 8.1 Hz, 1H), 1.80 (s, 3H), 1.72 (d, J = 12.5 Hz, 2H), 1.43 (s, 3H), 1.28 (s, 4H), 1.19 (s, 4H), 1.06 – 0.95 (m, 5H). Example 307. Preparation of Compound 351a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1S,4r)-4-(4- ((3R)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-y l)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1S,4r)-4-(4- ((3R*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7- yl)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of tert-butyl 3-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1- methyl-1H-indazol-7-yl)-4-oxopiperidine-1-carboxylate A mixture of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-7-bromo-1-methyl-1H-indaz ole (100 g, 199 mmol, 1.00 equiv.), compound tert-butyl 4-oxopiperidine-1-carboxylate (119 g, 599 mmol, 3.00 equiv.), t BuONa (57.6 g, 599 mmol, 3.00 equiv.), RuPhos (27.9 g, 59.9 mmol, 0.30 equiv.) in 1,4-dioxane (750 mL) was degassed and purged with N2 three times. To this solution was added Pd2(dba)3 (27.4 g, 29.9 mmol, 0.15 equiv.), and the mixture was stirred at 65 °C for 2 hours under N 2 atmosphere. The reaction mixture was poured into water (1.00 L) and extracted with ethyl acetate (1.00 L x 3). The combined organic phases were concentrated in vacuo to give a residue. The residue was purified by column chromatography (petroleum ether : ethyl acetate = 8: 1 to 1: 1) to give tert-butyl 3-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H- indazol-7-yl)-4-oxopiperidine-1-carboxylate (200 g, 323 mmol, 80.7% yield) as a yellow solid. MS (ESI) m/z = 619.4 [M+H] + Step B. Procedure for preparation of tert-butyl 3-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1- methyl-1H-indazol-7-yl)piperidine-1-carboxylate To a solution of tert-butyl 3-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-7- yl)-4- oxopiperidine-1-carboxylate (55.0 g, 88.9 mmol, 1.00 equiv.) in diglycol (550 mL) was added N2H4 . H2O (41.8 g, 711 mmol, 40.6 mL, 85.0% purity, 8.00 equiv.) and KOH (29.9 g, 533 mmol, 6.00 equiv.). The mixture was stirred at 120 °C for 6 hrs. The reaction mixture was quenched by addition of 0.50 M HCl (1.20 L) and then extracted with ethyl acetate (1.60 L x 3). The combined organic layers were washed with brine (1.20 L x 2), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (petroleum ether : ethyl acetate = 1 : 0 to 2:1) to give tert-butyl 3-(3-(2,6- bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-7-yl)piperid ine-1-carboxylate (48.0 g, 78.6 mmol, 44.3% yield) as a yellow solid. MS (ESI) m/z = 605.6 (M+H) + Step C. Procedure for preparation of tert-butyl (R)-3-(3-(2,6-bis(benzyloxy)pyridin-3- yl)-1-methyl-1H-indazol-7-yl)piperidine-1-carboxylate and tert-butyl (S)-3-(3-(2,6- bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-7-yl)piperid ine-1-carboxylate Stereoisomers of tert-butyl 3-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-7- yl)piperidine-1-carboxylate (56.5 g, 93.4 mmol) were separated by SFC to give tert-butyl (R)- 3-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-7- yl)piperidine-1-carboxylate (27.4 g, 45.31 mmol, 48.5% yield) and tert-butyl (S)-3-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1- methyl-1H-indazol-7-yl)piperidine-1-carboxylate (26.3 g, 43.4 mmol, 46.5% yield) as yellow solids. MS (ESI) m/z = 605.8 [M+H] + . MS (ESI) m/z = 605.8 [M+H] + Step D. Procedure for preparation of tert-butyl (3R)-3-(3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperidine-1-carboxylate To a solution of tert-butyl (R)-3-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazo l-7- yl)piperidine-1-carboxylate (13.0 g, 21.5 mmol, 1.00 equiv.), and Pd/C (6.50 g, 6.11 mmol, 10.0% purity), Pd(OH)2 (6.50 g, 9.26 mmol, 20.0% purity) in EtOH (130 mL), THF (130 mL) under Ar. The suspension was degassed under vacuum and purged with H 2 three times. The mixture was stirred under H2 (50 psi) at 50 °C for 2 hrs. The reaction mixture was filtered and washed with ethyl acetate (300 mL), and concentrated under reduced pressure to give tert-butyl (3R)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl )piperidine-1-carboxylate (9.00 g, 9.25 mmol, 100% yield) as a light yellow solid. MS (ESI) m/z = 427.2 [M+H] + Step E. Procedure for preparation of 3-(1-methyl-7-((R)-piperidin-3-yl)-1H-indazol-3- yl)piperidine-2,6-dione A mixture of tert-butyl (3R)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7- yl)piperidine-1-carboxylate (8.00 g, 18.7 mmol, 1.00 equiv.) in HCl/dioxane (80.0 mL) was stirred at 25 °C for 1 hr. The reaction mixture was filtered and concentrated under reduced pressure to give 3-(1-methyl-7-((R)-piperidin-3-yl)-1H-indazol-3-yl)piperidin e-2,6-dione (8.49 g, 25.6 mmol, 51.6% yield) as a white solid. MS (ESI) m/z = 327.3 [M+H] + . 1 H NMR: (400 MHz, DMSO-d 6 ) δ 10.89 (s, 1H), 9.78 (br d, J = 10.1 Hz, 1H), 9.43 (br s, 1H), 7.60 (d, J = 8.0 Hz, 1H), 7.33 (d, J = 7.1 Hz, 1H), 7.10 (t, J = 7.6 Hz, 1H), 4.37 (dd, J = 4.9, 9.9 Hz, 1H), 4.27 (s, 3H), 4.05 (br t, J = 11.8 Hz, 1H), 3.41 - 3.28 (m, 2H), 3.16 - 3.06 (m, 1H), 3.02 - 2.87 (m, 1H), 2.74 - 2.55 (m, 2H), 2.41 - 2.27 (m, 1H), 2.15 (br dd, J = 5.5, 13.1 Hz, 1H), 2.06 - 1.96 (m, 2H), 1.88 (br d, J = 14.5 Hz, 2H) Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-(4-((3R)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperidin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)oxy)phenyl)picol inate (60 mg, 1 equiv.81 μmol) and 3-(1-methyl-7-((R)-piperidin-3-yl)-1H-indazol-3-yl)piperidin e-2,6-dione, HCl (35 mg, 1.2 equiv. 97 μmol) were suspended in DCM (5 mL). To the mixture was added sodium triacetoxyborohydride (51 mg, 36 μL, 3 equiv. 0.24 mmol). The mixture was stirred at room temperature overnight. The mixture was then concentrated and extracted with DCM and water. The organic layer was separated, washed with brine, dried over Na2SO4, filtered, and evaporated to dryness to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-(4-((3R)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperidin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinate, which was carried forward without further purification. MS (ESI) m/z: 528.5 [M+2H] 2+ . Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-(4-((3R)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperidin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3- (((1S,4r)-4-(4-((3R*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-7-yl)piperidin- 1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1S,4r)-4-(4-((3R)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl -1H-indazol-7-yl)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinatetert (85 mg, 1 equiv. 81 μmol) was dissolved in DCM (4 mL). To the mixture was added 2 mL TFA. The mixture was stirred at room temperature overnight. The reaction mixture was concentrated, and then purified by RP- HPLC to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1S,4r)-4-(4-((3R)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl -1H-indazol-7-yl)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r) -4-(4-((3R*)-3-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperidin-1-yl )butyl)cyclohexyl)oxy)-2- methylphenyl)picolinic acid] (38 mg, 38 μmol, 47 %) as a white solid. MS (ESI) m/z: 500.4 [M+2H] 2+ ; 1 H NMR (400 MHz, DMSO-d6) δ 10.81 (s, 1H), 7.95 (d, J = 7.9 Hz, 1H), 7.71 (d, J = 8.1 Hz, 1H), 7.53 (dd, J = 18.8, 7.8 Hz, 2H), 7.44 – 7.34 (m, 3H), 7.29 (dt, J = 11.1, 7.5 Hz, 2H), 7.22 (d, J = 7.1 Hz, 1H), 7.00 (q, J = 7.8 Hz, 2H), 6.88 (d, J = 8.8 Hz, 1H), 6.83 (d, J = 8.3 Hz, 1H), 6.55 (d, J = 7.4 Hz, 1H), 4.91 (s, 2H), 4.28 (dd, J = 9.9, 5.1 Hz, 1H), 4.14 (s, 3H), 4.11 (s, 1H), 3.84 (t, J = 6.0 Hz, 2H), 3.59 (s, 1H), 3.04 (s, 1H), 2.95 (t, J = 5.9 Hz, 2H), 2.61 – 2.45 (m, 2H), 2.33 – 2.20 (m, 1H), 2.08 (dd, J = 13.5, 5.5 Hz, 1H), 1.99 (d, J = 10.0 Hz, 2H), 1.92 – 1.77 (m, 4H), 1.77 – 1.65 (m, 4H), 1.60 (s, 1H), 1.43 (s, 2H), 1.22 (d, J = 6.7 Hz, 0H), 1.14 (d, J = 6.4 Hz, 1H), 0.98 (t, J = 12.2 Hz, 2H). Example 308. Preparation of Compound 351b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1R,4r)-4-(4- ((3S)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-y l)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1R,4r)-4-(4- ((3S*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7- yl)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of tert-butyl (3S)-3-(3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperidine-1-carboxylate A solution of tert-butyl (S)-3-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazo l-7- yl)piperidine-1-carboxylate (23.0 g, 38.0 mmol, 1.00 equiv.) and Pd/C (11.5 g, 12.2 mmol, 10.0% purity), Pd(OH) 2 (11.5 g, 18.5 mmol, 20.0% purity) in EtOH (230 mL), THF (230 mL) under Ar. was degassed under vacuum and purged with H2 three times. The mixture was stirred under H2 (50 psi) at 50 °C for 2 hrs. The reaction mixture was filtered and washed with ethyl acetate (300 mL), then concentrated under reduced pressure to give tert-butyl (3S)-3-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperidine-1-c arboxylate (18.0 g, 48.4 mmol, 100% yield) as a light-yellow solid. MS (ESI) m/z = 427.2 [M+H] + Step B. Procedure for preparation of 3-(1-methyl-7-((S)-piperidin-3-yl)-1H-indazol-3- yl)piperidine-2,6-dione A mixture of tert-butyl (3S)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-7- yl)piperidine-1-carboxylate (15.0 g, 35.1 mmol, 1.00 eq)) in HCl/dioxane (150 mL) was stirred at 25 °C for 1 hr. The reaction mixture was filtered and concentrated under reduced pressure to give 3-(1-methyl-7-((S)-piperidin-3-yl)-1H-indazol-3-yl)piperidin e-2,6-dione (8.49g, 25.9 mmol, 61.9% yield, HCl) as light yellow solid. MS (ESI) m/z = 327.3 [M+H] + . 1 H NMR: (400 MHz, DMSO-d 6 ) δ 10.90 (s, 1H), 9.76 (br d, J = 10.4 Hz, 1H), 9.41 (br s, 1H), 7.61 (d, J = 8.0 Hz, 1H), 7.33 (d, J = 7.3 Hz, 1H), 7.10 (t, J = 7.6 Hz, 1H), 4.37 (dd, J = 4.9, 9.9 Hz, 1H), 4.27 (s, 3H), 4.11 - 4.00 (m, 1H), 3.42 - 3.28 (m, 2H), 3.14 (q, J = 11.6 Hz, 1H), 3.01 - 2.88 (m, 1H), 2.74 - 2.55 (m, 2H), 2.34 (ttd, J = 4.7, 9.3, 13.7 Hz, 1H), 2.20 - 2.10 (m, 1H), 2.07 - 1.96 (m, 2H), 1.94 - 1.80 (m, 2H). Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-(4-((3S)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperidin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)oxy)phenyl)picol inate (60 mg, 1 equiv.81 μmol) and rel-3-(1-methyl-7-((R)-piperidin-3-yl)-1H-indazol-3-yl)piper idine-2,6-dione, HCl (35 mg, 1.2 equiv.97 μmol) were suspended in DCM (5 mL). To the mixture was added sodium triacetoxyborohydride (51 mg, 36 μL, 3 equiv. 0.24 mmol). The mixture was stirred at room temperature overnight. The mixture was then concentrated and extracted with DCM and water. The organic layer was separated, washed with brine, dried over Na2SO4, filtered, and evaporated to dryness to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-(4-((3S)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperidin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinate, which was carried forward without further purification. MS (ESI) m/z: 528.5 [M+2H] 2+ . Step D. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-(4-((3S)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-7-yl)piperidin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3- (((1R,4r)-4-(4-((3S*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-7-yl)piperidin- 1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1R,4r)-4-(4-((3S)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl -1H-indazol-7-yl)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (85 mg, 1 equiv.81 μmol) was dissolved in DCM (4 mL). To the mixture was added 2 mL TFA. The mixture was stirred at room temperature overnight. The reaction mixture was concentrated, and then purified by RP-HPLC to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1R,4r)-4-(4-((3S)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl -1H-indazol-7-yl)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r) -4-(4-((3S*)-3-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-7-yl)piperidin-1-yl )butyl)cyclohexyl)oxy)-2- methylphenyl)picolinic acid] (42 mg, 42 μmol, 52 %) as a white solid. MS (ESI) m/z: 500.4 [M+2H] 2+ ; 1 H NMR (400 MHz, DMSO-d 6 ) δ 10.81 (s, 1H), 7.95 (d, J = 7.9 Hz, 1H), 7.71 (d, J = 8.1 Hz, 1H), 7.53 (dd, J = 19.7, 7.8 Hz, 2H), 7.44 – 7.17 (m, 7H), 7.00 (q, J = 7.8 Hz, 2H), 6.87 (d, J = 8.8 Hz, 1H), 6.86 – 6.76 (m, 1H), 6.55 (d, J = 7.5 Hz, 1H), 4.91 (s, 2H), 4.28 (dt, J = 8.4, 4.2 Hz, 1H), 4.13 (d, J = 7.6 Hz, 4H), 4.01 (s, 1H), 3.84 (t, J = 6.0 Hz, 2H), 3.59 (s, 1H), 3.10 (s, 2H), 3.03 (s, 1H), 2.95 (t, J = 6.0 Hz, 2H), 2.61 – 2.45 (m, 2H), 2.34 – 2.20 (m, 1H), 2.13 – 2.04 (m, 1H), 2.04 – 1.96 (m, 2H), 1.82 (d, J = 17.4 Hz, 4H), 1.69 (d, J = 13.3 Hz, 3H), 1.42 (s, 2H), 1.24 (dd, J = 21.7, 9.4 Hz, 1H), 1.17 – 1.09 (m, 1H), 0.98 (t, J = 12.3 Hz, 2H). Example 309. Preparation of Compound 352a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1S,4r)-4-(4- ((3R)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1S,4r)-4-(4- ((3R*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of tert-butyl 3-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1- methyl-1H-indazol-6-yl)-4-oxopiperidine-1-carboxylate A mixture of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-bromo-1-methyl-1H-indaz ole (50.0 g, 99.9 mmol, 1.00 equiv.), compound tert-butyl 4-oxopiperidine-1-carboxylate (59.7 g, 299 mmol, 3.00 equiv.), t BuONa (28.8 g, 299 mmol, 3.00 equiv.), and RuPhos (13.9 g, 29.9 mmol, 0.30 equiv.) in 1,4-dioxane (500 mL) was degassed and purged with N 2 three times. Then, Pd 2 (dba) 3 (13.7 g, 14.9 mmol, 0.15 equiv.) was added, and the mixture was stirred at 65 °C for 2 hours under N2 atmosphere. The reaction mixture was poured into water (1.00 L) and extracted with ethyl acetate (1.00 L x 3). The combined organic phase was concentrated in vacuo to give a residue. The residue was purified by column chromatography (petroleum ether : ethyl acetate = 8 : 1 to 1 : 1) to give tert-butyl 3-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol- 6-yl)-4-oxopiperidine-1-carboxylate (100 g, 146 mmol, 48.9% yield) as a yellow solid. MS (ESI) m/z = 619.6 [M+H] + . MS (ESI) m/z = 619.6 [M+H] + Step B. Procedure for preparation of tert-butyl 3-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1- methyl-1H-indazol-6-yl)piperidine-1-carboxylate To a solution of tert-butyl 3-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-6- yl)-4- oxopiperidine-1-carboxylate (35.0 g, 56.5 mmol, 1.00 equiv.) in diglycol (350 mL) was added N 2 H 4 . H 2 O (26.6 g, 452 mmol, 25.8 mL, 85.0% purity, 8.00 equiv.) and KOH (19.0 g, 339 mmol, 6.00 equiv.). The mixture was stirred at 120 °C for 6 hrs. The reaction mixture was quenched by addition of 0.50 M HCl (500 mL), and then extracted with ethyl acetate (500 mL x 3). The combined organic layers were washed with brine (250 mL x 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (petroleum ether : ethyl acetate = 5 : 1 to 1 :1) to give tert-butyl 3-(3-(2,6- bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-6-yl)piperid ine-1-carboxylate (31.0 g, 51.1 mmol, 37.2% yield) as a yellow solid. MS (ESI) m/z = 605.7 [M+H] + Step C. Procedure for preparation of tert-butyl (R)-3-(3-(2,6-bis(benzyloxy)pyridin-3- yl)-1-methyl-1H-indazol-6-yl)piperidine-1-carboxylate and tert-butyl (S)-3-(3-(2,6- bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-6-yl)piperid ine-1-carboxylate The stereosomers of tert-butyl 3-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-6- yl)piperidine-1-carboxylate (31.0 g, 51.1 mmol) were separated by SFC to give tert-butyl (R)- 3-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazol-6- yl)piperidine-1-carboxylate (13.0 g, 21.50 mmol, 41.9% yield) and tert-butyl (S)-3-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1- methyl-1H-indazol-6-yl)piperidine-1-carboxylate (13.0 g, 21.50 mmol, 41.9% yield) as light- yellow solids. Step D. Procedure for preparation of tert-butyl (3R)-3-(3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperidine-1-carboxylate A solution of tert-butyl (R)-3-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-indazo l-6- yl)piperidine-1-carboxylate (13.0 g, 21.5 mmol, 1.00 equiv.), and Pd/C (6.50 g, 6.11 mmol, 10.0% purity), Pd(OH) 2 (6.50 g, 9.26 mmol, 20.0% purity) in EtOH (130 mL), THF (130 mL) under Ar was degassed under vacuum and purged with H2 three times. The mixture was stirred under H2 (50 psi) at 50 °C for 2 hrs. The reaction mixture was filtered, washed with ethyl acetate (300 mL), and concentrated under reduced pressure to give compound tert-butyl (3R)- 3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)pipe ridine-1-carboxylate (6.20 g, 14.4 mmol, 67.2% yield, 99.4% purity) as a white solid. MS (ESI) m/z = 427.3 [M+H] + Step E. Procedure for preparation of 3-(1-methyl-6-((R)-piperidin-3-yl)-1H-indazol-3- yl)piperidine-2,6-dione A mixture of compound tert-butyl (3R)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 6-yl)piperidine-1-carboxylate (5.00 g, 11.7 mmol, 1.00 eq) in HCl/dioxane (50.0 mL) was stirred at 25 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give 3-(1-methyl-6-((R)-piperidin-3-yl)-1H-indazol-3-yl)piperidin e-2,6-dione (4.62 g, 14.1 mmol, 100% yield, 98.0% purity) as a white solid. MS (ESI) m/z = 327.3 [M+H] + . 1 H NMR: (400 MHz, DMSO-d6) δ 11.02 - 10.77 (m, 1H), 9.68 - 9.32 (m, 2H), 7.67 (d, J = 8.4 Hz, 1H), 7.52 (s, 1H), 7.06 (br d, J = 8.4 Hz, 1H), 4.41 - 4.31 (m, 1H), 3.98 (s, 3H), 3.35 - 3.05 (m, 4H), 2.98 - 2.81 (m, 1H), 2.75 - 2.53 (m, 2H), 2.42 - 2.27 (m, 1H), 2.23 - 2.08 (m, 1H), 1.98 - 1.70 (m, 4H) Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-(4-((3R)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperidin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)oxy)phenyl)picol inate (60 mg, 1 equiv.81 μmol) and 3-(1-methyl-6-((R)-piperidin-3-yl)-1H-indazol-3-yl)piperidin e-2,6-dione, HCl (35 mg, 1.2 equiv. 97 μmol) were suspended in DCM (5 mL). To the mixture was added sodium triacetoxyborohydride (51 mg, 36 μL, 3 equiv. 0.24 mmol). The mixture was stirred at room temperature overnight. The mixture was then concentrated and extracted with DCM and water. The organic layer was separated, washed with brine, dried over Na2SO4, filtered, and evaporated to dryness to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-(4-((3R)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperidin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinate, which was carried forward without further purification. MS (ESI) m/z: 528.4 [M+2H] 2+ . Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r)-4-(4-((3R)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperidin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3- (((1S,4r)-4-(4-((3R*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methy l-1H-indazol-6-yl)piperidin- 1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1S,4r)-4-(4-((3R)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl -1H-indazol-6-yl)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (85 mg, 1 equiv.81 μmol) was dissolved in DCM (4 mL). To the mixture was added 2 mL TFA. The mixture was stirred at room temperature overnight. The reaction mixture was concentrated, and then purified by RP-HPLC to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1S,4r)-4-(4-((3R)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl -1H-indazol-6-yl)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1S,4r) -4-(4-((3R*)-3-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperidin-1-yl )butyl)cyclohexyl)oxy)-2- methylphenyl)picolinic acid] (43 mg, 43 μmol, 53 %) as a white solid. MS (ESI) m/z: 500.4 [M+2H] 2+ ; 1 H NMR (400 MHz, DMSO-d6) δ 10.81 (s, 1H), 7.95 (d, J = 7.9 Hz, 1H), 7.72 (d, J = 8.1 Hz, 1H), 7.56 (dd, J = 8.2, 3.1 Hz, 2H), 7.38 (dt, J = 11.3, 3.8 Hz, 4H), 7.29 (dt, J = 10.7, 7.6 Hz, 2H), 7.03 – 6.93 (m, 2H), 6.87 (d, J = 8.8 Hz, 1H), 6.83 (d, J = 8.3 Hz, 1H), 6.55 (d, J = 7.4 Hz, 1H), 4.91 (s, 2H), 4.27 (dd, J = 9.8, 5.0 Hz, 1H), 4.10 (tt, J = 10.5, 4.4 Hz, 1H), 3.90 (s, 3H), 3.84 (t, J = 6.0 Hz, 2H), 3.05 (s, 2H), 2.95 (t, J = 6.0 Hz, 2H), 2.90 (s, 1H), 2.67 – 2.45 (m, 2H), 2.34 – 2.21 (m, 1H), 2.09 (dq, J = 12.9, 5.2 Hz, 1H), 2.03 – 1.95 (m, 2H), 1.85 (d, J = 5.6 Hz, 1H), 1.82 (s, 1H), 1.80 (s, 3H), 1.71 (s, 3H), 1.64 (d, J = 26.8 Hz, 1H), 1.59 – 1.49 (m, 1H), 1.43 (s, 2H), 1.31 – 1.20 (m, 1H), 1.14 (s, 2H), 0.96 (q, J = 11.9 Hz, 2H). Example 310. Preparation of Compound 352b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1R,4r)-4-(4- ((3S)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-y l)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1R,4r)-4-(4- ((3S*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of tert-butyl (3S)-3-(3-(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperidine-1-carboxylate A solution of compound tert-butyl (S)-3-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H- indazol-6-yl)piperidine-1-carboxylate (13.0 g, 21.5 mmol, 1.00 equiv.), Pd/C (6.50 g, 6.11 mmol, 10.0% purity), and Pd(OH) 2 (6.50 g, 9.26 mmol, 20.0% purity) in EtOH (130 mL) and THF (130 mL) under Ar was degassed under vacuum and purged with H2 three times. The mixture was stirred under H2 (50 psi) at 50 °C for 2 hrs. The reaction mixture was filtered, washed with ethyl acetate (300 mL), and concentrated under reduced pressure to give compound tert-butyl (3S)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)piperidine-1-carboxylate (6.13 g, 14.3 mmol, 66.5% yield) as a white solid. MS (ESI) m/z = 427.2 [M+H] + Step B. Procedure for preparation of 3-(1-methyl-6-((S)-piperidin-3-yl)-1H-indazol-3- yl)piperidine-2,6-dione A mixture of compound tert-butyl (3S)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 6-yl)piperidine-1-carboxylate (5.00 g, 11.7 mmol, 1.00 eq) in HCl/dioxane (50.0 mL) was stirred at 25 °C for 1 hr. The reaction mixture was filtered and concentrated under reduced pressure to give 3-(1-methyl-6-((S)-piperidin-3-yl)-1H-indazol-3-yl)piperidin e-2,6-dione (6.57 g, 20.0 mmol, 100% yield, HCl) as light-yellow solid. 1 H NMR: (400 MHz, DMSO-d6) δ 10.89 (s, 1H), 9.60 - 9.17 (m, 2H), 7.67 (d, J = 8.3 Hz, 1H), 7.52 (s, 1H), 7.06 (br d, J = 8.4 Hz, 1H), 4.35 (dd, J = 5.0, 9.9 Hz, 1H), 3.98 (s, 3H), 3.30 (br d, J = 10.6 Hz, 2H), 2.89 (br d, J = 3.0 Hz, 1H), 2.74 - 2.54 (m, 2H), 2.43 - 2.27 (m, 1H), 2.20 - 2.10 (m, 1H), 1.98 - 1.84 (m, 3H), 1.80 (br dd, J = 9.0, 14.8 Hz, 1H). MS (ESI) m/z = 327.1 [M+H] + . Step C. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-(4-((3S)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperidin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)oxy)phenyl)picol inate (60 mg, 1 equiv.81 μmol) and 3-(1-methyl-6-((R)-piperidin-3-yl)-1H-indazol-3-yl)piperidin e-2,6-dione, HCl (35 mg, 1.2 equiv. 97 μmol) were suspended in DCM (5 mL). To the mixture was added sodium triacetoxyborohydride (51 mg, 36 μL, 3 equiv. 0.24 mmol). The mixture was stirred at room temperature overnight. The mixture was then concentrated and extracted with DCM and water. The organic layer was separated, washed with brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-(4-((3S)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperidin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinate, which was carried forward without further purification. MS (ESI) m/z: 528.5 [M+2H] 2+ . Step D. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-(4-((3S)-3-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperidin [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1R,4r)-4-(4-((3S*)-3-(3 -(2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperidin-1-yl)butyl)cyclohexyl)oxy)- 2-methylphenyl)picolinic acid] tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1R,4r)-4-(4-((3S)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl -1H-indazol-6-yl)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinate (85 mg, 1 equiv.81 μmol) was dissolved in DCM (4 mL). To the mixture was added TFA (2 mL). The mixture was stirred at room temperature overnight. The reaction mixture was concentrated, and then purified by RP-HPLC to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1R,4r)-4-(4-((3S)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl -1H-indazol-6-yl)piperidin [6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(3-(((1R,4r)-4-(4- ((3S*)-3-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6- yl)piperidin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] (35 mg, 35 μmol, 43 %) as a white solid. MS (ESI) m/z: 500.4 [M+2H] 2+ ; 1 H NMR (400 MHz, DMSO-d6) δ 10.80 (s, 1H), 7.95 (d, J = 7.9 Hz, 1H), 7.72 (d, J = 8.0 Hz, 1H), 7.56 (dd, J = 8.3, 2.6 Hz, 2H), 7.39 (ddd, J = 14.6, 8.5, 2.7 Hz, 4H), 7.29 (dt, J = 10.6, 7.6 Hz, 2H), 7.03 – 6.93 (m, 2H), 6.87 (d, J = 8.8 Hz, 1H), 6.83 (d, J = 8.3 Hz, 1H), 6.55 (d, J = 7.5 Hz, 1H), 4.91 (s, 2H), 4.26 (dd, J = 9.8, 5.0 Hz, 1H), 3.90 (d, J = 3.1 Hz, 3H), 3.84 (t, J = 5.9 Hz, 2H), 3.04 (s, 2H), 2.95 (t, J = 6.0 Hz, 2H), 2.89 (s, 1H), 2.67 – 2.45 (m, 2H), 2.34 – 2.21 (m, 1H), 2.15 – 2.04 (m, 1H), 2.03 – 1.96 (m, 2H), 1.85 (d, J = 5.5 Hz, 1H), 1.82 (s, 1H), 1.80 (s, 3H), 1.73 – 1.49 (m, 3H), 1.43 (s, 1H), 1.31 – 1.20 (m, 1H), 1.13 (d, J = 10.0 Hz, 3H), 0.96 (q, J = 12.0 Hz, 2H). Example 311. Preparation of Compound 354a 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- ((2S,6R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 6-yl)-2,6-dimethylpiperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Preparation of tert-butyl (2S,6R)-4-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl- 1H-indazol-6-yl)-2,6-dimethylpiperazine-1-carboxylate 3-(2,6-Bis(benzyloxy)pyridin-3-yl)-6-bromo-1-methyl-1H-indaz ole (1.0 g, 1 equiv.2.0 mmol) was dissolved in 1,4-dioxane (10 mL). To the mixture was added tert-butyl (2S,6R)-2,6- dimethylpiperazine-1-carboxylate (0.51 g, 1.2 equiv. 2.4 mmol), followed by (2- dicyclohexylphosphino-2',6'-diisopropoxy-1,1'-biphenyl)[2-(2 '-amino-1,1'- biphenyl)]palladium(II) methanesulfonate (0.17 g, 0.1 equiv. 0.20 mmol), and sodium tert- butoxide (0.58 g, 3.0 mL, 2 molar, 3 equiv.6.0 mmol). The mixture was then purged with N2, and then heated at 100ºC for ~ 2 hrs. The mixture was cooled down to room temperature, quenched with water, and then extracted with ethyl acetate (3 x). The organic layers were combined, washed with brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness. The residue was then purified by silica gel column chromatography (0-100% ethyl acetate in heptane) to afford tert-butyl (2S,6R)-4-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H- indazol-6-yl)-2,6-dimethylpiperazine-1-carboxylate, which was used in the next step without further purification. Step B. Preparation of tert-butyl (2S,6R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)-2,6-dimethylpiperazine-1-carboxylate tert-butyl (2S,6R)-4-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-in dazol-6-yl)-2,6- dimethylpiperazine-1-carboxylate (1.3 g, 1 equiv.2.0 mmol) was dissolved in ethyl acetate (20 mL). To the mixture was added Pd(OH)2 (0.35 g, 20% Wt, 0.25 equiv.0.50 mmol). The mixture was stirred at 50ºC under H 2 overnight. The mixture was filtered through celite and rinsed with ethyl acetate (3 x). The filtrate was then evaporated to dryness. The residue was then purified by RP-HPLC to afford tert-butyl (2S,6R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)-2,6-dimethylpiperazine-1-carboxylate (0.56 g, 1.2 mmol, 61 %) as a white solid. MS (ESI) m/z: 456.4 [M+H] + . Step C. Preparation of 3-(6-((3S,5R)-3,5-dimethylpiperazin-1-yl)-1-methyl-1H-indazo l- 3-yl)piperidine-2,6-dione, HCl tert-butyl (2S,6R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6 -yl)-2,6- dimethylpiperazine-1-carboxylate (0.56 g, 1 equiv. 1.2 mmol) was suspended in 4M HCl in dioxane (4mL). The mixture was then stirred at room temperature overnight. The mixture was concentrated to afford 3-(6-((3S,5R)-3,5-dimethylpiperazin-1-yl)-1-methyl-1H-indazo l-3- yl)piperidine-2,6-dione, HCl (0.49 g, 1.3 mmol, 100 %) as a white solid. MS (ESI) m/z: 356.2 [M+H] + . Step D. Preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-((2S,6R)-4- (3-(2,6-dioxopiperidin-3-yl)- 1-methyl-1H-indazol-6-yl)-2,6-dimethylpiperazin-1-yl)butyl)c yclohexyl)oxy)-2- methylphenyl)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)oxy)phenyl)picol inate (60 mg, 1 equiv.81 μmol) and 3-(6-((3S,5R)-3,5-dimethylpiperazin-1-yl)-1-methyl-1H-indazo l-3-yl)piperidine-2,6- dione, HCl (35 mg, 1.1 equiv.89 μmol) was suspended in DCM (5 mL). To the mixture was added sodium triacetoxyborohydride (51 mg, 36 μL, 3 equiv. 0.24 mmol). The mixture was stirred at room temperature overnight. The mixture was then concentrated and extracted with DCM and water. The organic layer was separated, washed with brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-(4-((2S,6R )-4-(3-(2,6-dioxopiperidin-3-yl)- 1-methyl-1H-indazol-6-yl)-2,6-dimethylpiperazin-1-yl)butyl)c yclohexyl)oxy)-2- methylphenyl)picolinate, which was carried forward without further purification. MS (ESI) m/z: 543.0 [M+2H] 2+ . Step E. Preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4-((2S,6R)-4-(3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H- indazol-6-yl)-2,6-dimethylpiperazin-1-yl)butyl)cyclohexyl)ox y)-2-methylphenyl)picolinic acid tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-(4-((2S,6R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-met hyl-1H-indazol-6-yl)-2,6- dimethylpiperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl) picolinate (88 mg, 1 equiv.81 μmol) was dissolved in DCM (4 mL). To the mixture was added 1 mL TFA. The mixture was stirred at room temperature. The mixture was concentrated, and purified by RP-HPLC to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4-(4- ((2S,6R)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 6-yl)-2,6-dimethylpiperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (9 mg, 9 μmol, 10 %) as a white solid. MS (ESI) m/z: 514.9 [M+2H] 2+ ; 1 H NMR (400 MHz, DMSO-d6) δ 12.79 (s, 1H), 10.77 (s, 1H), 7.96 (d, J = 7.8 Hz, 1H), 7.72 (d, J = 8.1 Hz, 1H), 7.55 (d, J = 7.4 Hz, 1H), 7.44 – 7.35 (m, 3H), 7.35 – 7.24 (m, 2H), 7.00 (t, J = 7.8 Hz, 1H), 6.87 (dd, J = 17.1, 8.6 Hz, 3H), 6.75 (s, 1H), 6.54 (d, J = 7.4 Hz, 1H), 4.91 (s, 2H), 3.86 (d, J = 5.9 Hz, 1H), 3.83 (s, 4H), 3.57 (s, 2H), 2.96 (t, J = 6.0 Hz, 2H), 2.58 – 2.45 (m, 2H), 2.22 (s, 1H), 2.09 (dd, J = 13.0, 5.9 Hz, 1H), 1.85 (d, J = 6.9 Hz, 1H), 1.80 (s, 3H), 1.71 (d, J = 12.4 Hz, 2H), 1.37 (s, 1H), 1.29 (s, 8H), 1.17 (s, 5H), 1.01 (s, 6H). Example 312. Preparation of Compound 354b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(4- ((2S,6S)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 6-yl)-2,6-dimethylpiperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid Step A. Preparation of tert-butyl (2S,6S)-4-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl- 1H-indazol-6-yl)-2,6-dimethylpiperazine-1-carboxylate 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-bromo-1-methyl-1H-indaz ole (1.0 g, 1 equiv.2.0 mmol) was dissolved in 1,4-dioxane (10 mL). To the mixture was added tert-butyl (2S,6S)-2,6- dimethylpiperazine-1-carboxylate (0.51 g, 1.2 equiv. 2.4 mmol), followed by (2- dicyclohexylphosphino-2',6'-diisopropoxy-1,1'-biphenyl)[2-(2 '-amino-1,1'- biphenyl)]palladium(II) methanesulfonate (0.17 g, 0.1 equiv. 0.20 mmol), and sodium tert- butoxide (0.58 g, 3.0 mL, 2 molar, 3 equiv.6.0 mmol). The mixture was then purged with N 2 , and then heated at 100ºC for ~ 2 hrs. The mixture was cooled down to room temperature, quenched with water, and then extracted with ethyl acetate (3 x). The organic layers were combined, washed with brine, dried over Na2SO4, filtered, and evaporated to dryness. The residue was then purified by silica gel column chromatography (0-100% ethyl acetate in heptane) to afford tert-butyl (2S,6S)-4-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H- indazol-6-yl)-2,6-dimethylpiperazine-1-carboxylate, which was used in the next step without further purification. Step B. Preparation of tert-butyl (2S,6S)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)-2,6-dimethylpiperazine-1-carboxylate tert-Butyl (2S,6S)-4-(3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-1H-in dazol-6-yl)-2,6- dimethylpiperazine-1-carboxylate (1.3 g, 1 equiv.2.0 mmol) was dissolved in ethyl acetate (20 mL). To the mixture was added Pd(OH)2 (0.35 g, 20% Wt, 0.25 equiv.0.50 mmol). The mixture was stirred at 50ºC under H2 overnight. The mixture was filtered through celite and rinsed with ethyl acetate (3x ). The filtrate was then evaporated to dryness. The residue was then purified by RP-HPLC to afford tert-butyl (2S,6S)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 6-yl)-2,6-dimethylpiperazine-1-carboxylate (415 mg, 911 μmol, 46 %) as a white solid. MS (ESI) m/z: 456.3 [M+H] + . Step C. Preparation of 3-(6-((3S,5S)-3,5-dimethylpiperazin-1-yl)-1-methyl-1H-indazo l-3- yl)piperidine-2,6-dione, HCl tert-Butyl (2S,6S)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6 -yl)-2,6- dimethylpiperazine-1-carboxylate (415 mg, 1 equiv. 911 μmol) was suspended in 4M HCl in dioxane (4mL). The mixture was then stirred at room temperature overnight. The mixture was concentrated to afford 3-(6-((3S,5S)-3,5-dimethylpiperazin-1-yl)-1-methyl-1H-indazo l-3- yl)piperidine-2,6-dione, HCl (275 mg, 702 μmol, 77.0 %) as an off-white solid. MS (ESI) m/z: 356.2 [M+H] + . Step D. Preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(4-((2S,6S)-4- (3-(2,6-dioxopiperidin-3-yl)- 1-methyl-1H-indazol-6-yl)-2,6-dimethylpiperazin-1-yl)butyl)c yclohexyl)oxy)-2- methylphenyl)picolinate tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(2- methyl-3-(((1s,4r)-4-(4-oxobutyl)cyclohexyl)oxy)phenyl)picol inate (60 mg, 1 equiv.81 μmol) and 3-(6-((3S,5S)-3,5-dimethylpiperazin-1-yl)-1-methyl-1H-indazo l-3-yl)piperidine-2,6- dione, HCl (35 mg, 1.1 equiv.89 μmol) was suspended in DCM (5 mL). To the mixture was added sodium triacetoxyborohydride (51 mg, 36 μL, 3 equiv. 0.24 mmol). The mixture was stirred at room temperature overnight. The mixture was then concentrated and extracted with DCM and water. The organic layer was separated, washed with brine, dried over Na 2 SO 4 , filtered, and evaporated to dryness to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1r,4r)-4-(4-((2S,6S )-4-(3-(2,6-dioxopiperidin-3-yl)- 1-methyl-1H-indazol-6-yl)-2,6-dimethylpiperazin-1-yl)butyl)c yclohexyl)oxy)-2- methylphenyl)picolinate, which was carried forward without further purification. MS (ESI) m/z: 542.9 [M+2H] 2+ . Step E. Preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(4-((2S,6S)-4-(3-(2,6-dioxopiperi din-3-yl)-1-methyl-1H- indazol-6-yl)-2,6-dimethylpiperazin-1-yl)butyl)cyclohexyl)ox y)-2-methylphenyl)picolinic acid tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1r,4r)-4-(4-((2S,6S)-4-(3-(2,6-dioxopiperidin-3-yl)-1-met hyl-1H-indazol-6-yl)-2,6- dimethylpiperazin-1-yl)butyl)cyclohexyl)oxy)-2-methylphenyl) picolinate (88 mg, 1 equiv.81 μmol) was dissolved in DCM (4 mL). To the mixture was added TFA (1 mL). The mixture was stirred at room temperature. The mixture was concentrated, and purified by RP-HPLC to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1r,4r)-4-(4- ((2S,6S)-4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 6-yl)-2,6-dimethylpiperazin-1- yl)butyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (28 mg, 27 μmol, 34 %) as a white solid. MS (ESI) m/z: 514.9 [M+2H] 2+ ; 1 H NMR (400 MHz, DMSO-d6) δ 12.78 (s, 1H), 10.77 (s, 1H), 7.96 (d, J = 7.9 Hz, 1H), 7.72 (d, J = 8.0 Hz, 1H), 7.55 (d, J = 7.5 Hz, 1H), 7.47 – 7.24 (m, 5H), 7.00 (t, J = 7.9 Hz, 1H), 6.92 – 6.77 (m, 4H), 6.54 (d, J = 7.4 Hz, 1H), 4.91 (s, 2H), 4.19 (dd, J = 9.2, 5.1 Hz, 1H), 4.13 (s, 1H), 3.85 (d, J = 5.9 Hz, 1H), 3.83 (s, 3H), 2.96 (t, J = 6.0 Hz, 2H), 2.85 (s, 9H), 2.54 (p, J = 7.3 Hz, 2H), 2.27 – 2.19 (m, 1H), 2.15 – 1.98 (m, 3H), 1.80 (s, 2H), 1.72 (d, J = 12.4 Hz, 2H), 1.44 (s, 3H), 1.28 (s, 5H), 1.18 (s, 3H), 1.09 (s, 5H), 0.99 (q, J = 15.0 Hz, 2H). Example 313. Preparation of Compound 333b

6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3-(((1s,4r)-4- ((4R)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol- 6-yl)piperazin-1- yl)pentyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid [6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin -2(1H)-yl)-3-(3-(((1S,4r)-4- ((4R*)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol -6-yl)piperazin-1- yl)pentyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of 3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-6- piperazin-1-yl-indazole A mixture of 6-bromo-3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-indazole (2.5 g, 5.00 mmol, 1.0 equiv.), piperazine (1.29 g, 14.99 mmol, 3.0 equiv.), Pd 2 (dba) 3 (457.51 mg, 499.61 μmol, 0.1 equiv.), NaOtBu (960.26 mg, 9.99 mmol, 2.0 equiv.), and tritertbutylphosphane (2.02 g, 999.23 μmol, 2.35 mL, 10% purity, 0.2 equiv.) in toluene (80 mL) was degassed and purged with N2 three times. The mixture was stirred at 100°C for 16 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (10~20% MeOH /DCM) to give 3-(2,6-dibenzyloxy-3- pyridyl)-1-methyl-6-piperazin-1-yl-indazole (2.4 g, 4.67 mmol, 93.44% yield) as a yellow oil. MS (ESI) m/z: 506.4 [M+H] + . Step B. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-(4-(5-((1r,4s)- 4-(3-bromo-2-methylphenoxy)cyclohexyl)pentan-2-yl)piperazin- 1-yl)-1-methyl-1H- indazole A solution of 5-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)pentan-2-on e (1.5 g, 4.25 mmol, 1.0 equiv.), 3-(2,6-dibenzyloxy-3-pyridyl)-1-methyl-6-piperazin-1-yl-inda zole (2.15 g, 4.25 mmol, 1.0 equiv.) in DCM (15 mL) was stirred at 35 °C for 1 hour. To the mixture was added NaBH(OAc)3 (2.70 g, 12.74 mmol, 3 equiv.). The resulting mixture was stirred at 35 °C for 11 hours. DCM (300 mL) and water (300 mL) were added, and layers were separated. The aqueous layer was extracted with DCM (50 mL × 2), dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum. The residue was purified by flash silica gel chromatography (0~50% ethyl acetate/petroleum ether) to give 3-(2,6-bis(benzyloxy)pyridin- 3-yl)-6-(4-(5-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl )pentan-2-yl)piperazin-1-yl)- 1-methyl-1H-indazole (1.8 g, 2.06 mmol, 48.60% yield) as a yellow solid. MS (ESI) m/z: 844.6 [M+H] + Step C. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-(4-((S)-5- ((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)pentan-2-yl)p iperazin-1-yl)-1-methyl- 1H-indazole and 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-(4-((R)-5-((1r,4s)-4-(3 -bromo-2- methylphenoxy)cyclohexyl)pentan-2-yl)piperazin-1-yl)-1-methy l-1H-indazole The stereoisomers of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-(4-(5-((1r,4s)-4-(3-bro mo-2- methylphenoxy)cyclohexyl)pentan-2-yl)piperazin-1-yl)-1-methy l-1H-indazole (1.0 g, 949.11 μmol, 1.0 equiv.) were separated by SFC to give 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-(4-((S)- 5-((1r,4r)-4-(3-bromo-2-methylphenoxy)cyclohexyl)pentan-2-yl )piperazin-1-yl)-1-methyl- 1H-indazole (150 mg, 163.01 μmol, 12.90% yield) and 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6- (4-((R)-5-((1r,4s)-4-(3-bromo-2-methylphenoxy)cyclohexyl)pen tan-2-yl)piperazin-1-yl)-1- methyl-1H-indazole (240 mg, 276.17 μmol, 22.85% yield) as yellow solids. MS (ESI) m/z: 844.3 [M+H] + . MS (ESI) m/z: 844.3 [M+H] + Step D. Procedure for preparation of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-6-(4- ((R)-5-((1r,4S)-4-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2- yl)phenoxy)cyclohexyl)pentan-2-yl)piperazin-1-yl)-1H-indazol e A mixture of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-6-(4-((R)-5-((1r,4S)-4-(3 -bromo-2- methylphenoxy)cyclohexyl)pentan-2-yl)piperazin-1-yl)-1-methy l-1H-indazole (240 mg, 284.73 μmol, 1.0 equiv.), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (216.91 mg, 854.19 μmol, 3.0 equiv.) and KOAc (83.83 mg, 854.19 μmol, 3.0 equiv.) in dioxane (4 mL) was degassed and purged with N2 three times. Pd(dppf)Cl2 (41.67 mg, 56.95 μmol, 0.2 equiv.) was then added into the mixture. The reaction mixture was stirred at 90 °C for 12 hours under N 2 atmosphere. Ethyl acetate (100 mL) and water (100 mL) were added, and the layers were separated. The aqueous layer was extracted with ethyl acetate (30 mL × 2), dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum. The residue was purified by prep-TLC (SiO 2 , petroleum ether : ethyl acetate = 1:1) to give 3-(2,6-bis(benzyloxy)pyridin- 3-yl)-1-methyl-6-(4-((R)-5-((1r,4S)-4-(2-methyl-3-(4,4,5,5-t etramethyl-1,3,2-dioxaborolan-2- yl)phenoxy)cyclohexyl)pentan-2-yl)piperazin-1-yl)-1H-indazol e (90 mg, 93.04 μmol, 32.68% yield) as a yellow solid. MS (ESI) m/z: 890.4 [M+H] + Step E. Procedure for preparation of 3-(1-methyl-6-(4-((R)-5-((1r,4s)-4-(2-methyl-3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy)cyclohe xyl)pentan-2-yl)piperazin- 1-yl)-1H-indazol-3-yl)piperidine-2,6-dione To a mixture of 3-(2,6-bis(benzyloxy)pyridin-3-yl)-1-methyl-6-(4-((R)-5-((1r ,4s)-4-(2- methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenox y)cyclohexyl)pentan-2- yl)piperazin-1-yl)-1H-indazole (90 mg, 101.13 μmol, 1.0 equiv.) in THF (2 mL) and EtOH (2 mL) was added Pd/C (30.00 mg, 28.19 μmol, 10% purity) and Pd(OH)2 (30.00 mg, 42.72 μmol, 20% purity) under H2 atmosphere (15 Psi). The mixture was stirred at 40 °C for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a filter cake with a little EtOH. The filtrate was concentrated under reduced pressure to give 3-(1-methyl-6-(4- ((R)-5-((1r,4s)-4-(2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2- yl)phenoxy)cyclohexyl)pentan-2-yl)piperazin-1-yl)-1H-indazol -3-yl)piperidine-2,6-dione (85 mg, 83.60 μmol, 82.67% yield) as a yellow solid. MS (ESI) m/z: 712.4 [M+H] + Step F. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-((4R)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)pentyl)cyclohexyl)oxy) -2- methylphenyl)picolinate A mixture of 3-(1-methyl-6-(4-((R)-5-((1r,4s)-4-(2-methyl-3-(4,4,5,5-tetr amethyl-1,3,2- dioxaborolan-2-yl)phenoxy)cyclohexyl)pentan-2-yl)piperazin-1 -yl)-1H-indazol-3- yl)piperidine-2,6-dione (85 mg, 83.60 μmol, 1.0 equiv.), tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-bromopicolin ate (56.73 mg, 100.32 μmol, 1.2 equiv.), KF (14.57 mg, 250.79 μmol, 5.88 μL, 3.0 equiv.), and H 2 O (0.2 mL) in dioxane (2 mL) was degassed and purged with N2 three times. Then Ad2nBuP Pd G3 (12.18 mg, 16.72 μmol, 0.2 equiv.) was added into the mixture. The resulting mixture was degassed and purged with N 2 three times. The mixture was stirred at 100 °C for 12 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH = 20:1) to give tert-butyl 6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r) -4-((4R)-4-(4-(3-(2,6- dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)piperazin-1-yl )pentyl)cyclohexyl)oxy)-2- methylphenyl)picolinate (60 mg, 52.13 μmol, 62.36% yield) as a yellow solid. MS (ESI) m/z: 1070.5 [M+H] +. Step G. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(3-(((1s,4r)-4-((4R)-4-(4-(3- (2,6-dioxopiperidin-3-yl)-1- methyl-1H-indazol-6-yl)piperazin-1-yl)pentyl)cyclohexyl)oxy) -2-methylphenyl)picolinic acid A solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(3-(((1s,4r)-4-((4R)-4-(4-(3-(2,6-dioxopiperidin-3-yl) -1-methyl-1H-indazol-6- yl)piperazin-1-yl)pentyl)cyclohexyl)oxy)-2-methylphenyl)pico linate (60 mg, 56.06 μmol, 1.0 equiv.) in TFA (2 mL) and DCM (1 mL) was stirred at 40 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep- HPLC to give 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(3- (((1s,4r)-4-((4R)-4-(4-(3-(2,6-dioxopiperidin-3-yl)-1-methyl -1H-indazol-6-yl)piperazin-1- yl)pentyl)cyclohexyl)oxy)-2-methylphenyl)picolinic acid (15.71 mg, 15.06 μmol, 26.87% yield) as a white solid. MS (ESI) m/z: 1014.4 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 12.90 - 12.50 (m, 2H), 10.85 (s, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 7.6 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.51 - 7.48 (m, 1H), 7.47 - 7.43 (m, 3H), 7.40 - 7.33 (m, 2H), 7.10 - 7.03 (m, 1H), 6.96 (d, J = 8.8 Hz, 1H), 6.91 (d, J = 8.4 Hz, 2H), 6.84 (s, 1H), 6.61 (d, J = 7.6 Hz, 1H), 4.97 (s, 2H), 4.29 - 4.23 (m, 1H), 4.22 - 4.13 (m, 1H), 3.94 - 3.86 (m, 5H), 3.27 - 3.12 (m, 4H), 3.02 (t, J = 5.6 Hz, 2H), 2.79 - 2.69 (m, 2H), 2.65 - 2.55 (m, 4H), 2.32 - 2.25 (m, 1H), 2.19 - 2.12 (m, 1H), 2.11 - 2.04 (m, 2H), 1.86 (s, 3H), 1.82 - 1.74 (m, 2H), 1.57 - 1.47 (m, 1H), 1.41 - 1.27 (m, 6H), 1.23 (s, 3H), 1.11 - 0.96 (m, 5H). 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[4-[3-[1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-4-fluoro-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 4-fluoro-4-formyl-piperidine-1- carboxylate To a solution of oxalyl dichloride (2.72 g, 21.43 mmol, 1.88 mL, 2.0 equiv.) in DCM (30 mL) was added dropwise into DMSO (3.35 g, 42.87 mmol, 3.35 mL, 4.0 equiv.) at -78 °C under N 2 atmosphere. The mixture was stirred at -78 °C for 1 hour. After which time tert-butyl 4-fluoro- 4-(hydroxymethyl)piperidine-1-carboxylate (2.5 g, 10.72 mmol, 1.0 equiv.) in DCM (30 mL) was added dropwise at -78 °C. The solution was stirred for 1 hour at -78 °C. Then TEA (6.51 g, 64.30 mmol, 8.95 mL, 6.0 equiv.) was added into the solution. The solution was stirred at - 78 °C for 0.5 hours under N2 atmosphere. The reaction mixture was then partitioned between DCM (100 mL) and water (50 mL). The organic phase was separated, washed with brine (50 mL × 3), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give tert- butyl 4-fluoro-4-formyl-piperidine-1-carboxylate (4.8 g, crude) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 3.86 (s, 2H), 3.16 - 2.76 (m, 2H), 1.86 - 1.47 (m, 4H), 1.47 - 1.33 (m, 9H) Step B. Procedure for preparation of tert-butyl 4-[(E)-3-ethoxy-3-oxo-prop-1-enyl]-4- fluoro-piperidine-1-carboxylate To a solution of NaH (747.13 mg, 18.68 mmol, 60% purity, 1.8 equiv.) in THF (120 mL) was added ethyl 2-diethoxyphosphorylacetate (3.49 g, 15.57 mmol, 3.09 mL, 1.5 equiv.) at 0 °C for 1 hour under N2 atmosphere. To this solution was added tert-butyl 4-fluoro-4-formyl- piperidine-1-carboxylate (2.4 g, 10.38 mmol, 1 equiv.) in THF (10 mL). The mixture was stirred at 0 °C for 11 hours. The reaction mixture was quenched by addition of aq. NH 4 Cl (100 mL) at 0 °C, and then extracted with ethyl acetate (200 mL). The combined organic layers were washed with brine 100 mL, dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~6% ethyl acetate/petroleum ether) to give tert-butyl 4-[(E)-3-ethoxy-3-oxo-prop-1-enyl]-4-fluoro- piperidine-1-carboxylate (1.3 g, 4.31 mmol, 20.78% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 7.06 - 6.89 (m, 1H), 6.02 (d, J = 15.6 Hz, 1H), 4.22 - 4.04 (m, 2H), 3.86 (d, J = 10.4 Hz, 2H), 2.98 (s, 2H), 1.96 - 1.66 (m, 4H), 1.43 - 1.38 (m, 9H), 1.27 - 1.14 (m, 3H) Step C. Procedure for preparation of tert-butyl 4-(3-ethoxy-3-oxo-propyl)-4-fluoro- piperidine-1-carboxylate To a solution of tert-butyl 4-[(E)-3-ethoxy-3-oxo-prop-1-enyl]-4-fluoro-piperidine-1- carboxylate (0.65 g, 2.16 mmol, 1.0 equiv.) in EtOH (20 mL) was added Pd/C (459.08 mg, 431.39 μmol, 10% purity, 0.2 equiv.). The mixture was stirred at 25 °C for 12 hours under a N2 atmosphere. The mixture was filtered by EtOH (100 mL), and the filtrate was concentrated under reduced pressure to give tert-butyl 4-(3-ethoxy-3-oxo-propyl)-4-fluoro-piperidine-1- carboxylate (1.25 g, crude) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 4.10 - 4.01 (m, 2H), 3.90 (d, J = 8.8 Hz, 1H), 3.76 ( d, J = 10.4 Hz, 1H), 3.07 - 2.80 (m, 2H), 2.41 - 2.37 (m, 1H), 2.35 - 2.27 (m, 1H), 2.01 - 1.80 (m, 2H), 1.80 - 1.66 (m, 2H), 1.65 - 1.50 (m, 2H), 1.41 - 1.38 (m, 9H), 1.21 - 1.15 (m, 3H) Step D. Procedure for preparation of tert-butyl 4-fluoro-4-(3-hydroxypropyl)piperidine- 1-carboxylate To a solution of tert-butyl 4-(3-ethoxy-3-oxo-propyl)-4-fluoro-piperidine-1-carboxylate (0.6 g, 1.98 mmol, 1.0 equiv.) in THF (50 mL) was added LiAlH4 (2.5 M, 949.34 μL, 1.2 equiv.) under a N 2 atmosphere. The mixture was stirred at 0 °C for 1 hour. The reaction mixture was quenched by addition of Na 2 SO 4 ·10 H 2 O (1 g) under 0 °C, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~45% ethyl acetate/petroleum ether) to give tert-butyl 4-fluoro-4-(3-hydroxypropyl)piperidine-1- carboxylate (0.92 g, 3.52 mmol, 89.00% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ = 4.44 ( t, J = 4.8 Hz, 1H), 4.00 - 3.66 (m, 2H), 3.42 - 3.34 (m, 2H), 3.09 - 2.78 (m, 2H), 1.72 (t, J = 11.2 Hz, 1H), 1.65 - 1.54 (m, 3H), 1.52 - 1.45 (m, 2H), 1.43 - 1.37 (m, 10H), 1.26 - 1.14 (m, 1H) Step E. Procedure for preparation of tert-butyl 4-[3-(4-bromo-3-methyl-phenoxy)propyl]- 4-fluoro-piperidine-1-carboxylate A mixture of tert-butyl 4-fluoro-4-(3-hydroxypropyl)piperidine-1-carboxylate (460 mg, 1.76 mmol, 1.0 equiv.), 4-bromo-3-methyl-phenol (362.14 mg, 1.94 mmol, 1.1 equiv.), and 2- (tributyl-phosphanylidene)acetonitrile (637.25 mg, 2.64 mmol, 1.5 equiv.) in toluene (20 mL) was degassed and purged with N 2 three times. The mixture was stirred at 125 °C for 12 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~1% ethyl acetate/petroleum ether) to give tert-butyl 4-[3-(4-bromo-3-methyl-phenoxy)propyl]-4-fluoro- piperidine-1-carboxylate (700 mg, 1.63 mmol, 92.41% yield) as a yellow oil. Step F. Procedure for preparation of 4-[3-(4-bromo-3-methyl-phenoxy)propyl]-4-fluoro- piperidine A solution of tert-butyl 4-[3-(4-bromo-3-methyl-phenoxy)propyl]-4-fluoro-piperidine-1 - carboxylate (700 mg, 1.63 mmol, 1.0 equiv.) in HCl/dioxane (5 mL) and dioxane (3 mL) was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give 4-[3-(4-bromo-3-methyl-phenoxy)propyl]-4-fluoro-piperidine (530 mg, crude) as a white solid. MS (ESI) m/z: 332.0 [M+H] + . Step G. Procedure for preparation of ethyl 2-[4-[3-(4-bromo-3-methyl-phenoxy)propyl]- 4-fluoro-1-piperidyl]acetate To a solution of 4-[3-(4-bromo-3-methyl-phenoxy)propyl]-4-fluoro-piperidine (530 mg, 1.60 mmol, 1.0 equiv.) and ethyl 2-bromoacetate (536.05 mg, 3.21 mmol, 355.23 μL, 2.0 equiv.) in DMF (5 mL) was added K2CO3 (665.43 mg, 4.81 mmol, 3.0 equiv.). The mixture was stirred at 40 °C for 5.5 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product was purified by reversed-phase HPLC to give ethyl 2-[4-[3-(4- bromo-3-methyl-phenoxy)propyl]-4-fluoro-1-piperidyl]acetate (160 mg, 372.79 μmol, 23.23% yield) as a yellow oil. MS (ESI) m/z: 416.1 [M+H] + . Step H. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[3-[1-(2-ethoxy-2-oxo- ethyl)-4-fluoro-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate A mixture of ethyl 2-[4-[3-(4-bromo-3-methyl-phenoxy)propyl]-4-fluoro-1-piperid yl]acetate (160 mg, 384.32 μmol, 1.0 equiv.), tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)pyridine-2- carboxylate (258.95 mg, 422.75 μmol, 1.1 equiv.), KF (66.98 mg, 1.15 mmol, 27.01 μL, 3.0 equiv.), and Ad2nBuP Pd G3 (55.98 mg, 76.86 μmol, 0.2 equiv.) in dioxane (5 mL) and H2O (0.5 mL) was degassed and purged with N2 three times. The mixture was stirred at 100 °C for 1 hour under N 2 atmosphere. The reaction mixture was partitioned between ethyl acetate (30 mL) and water (20 mL). The organic phase was separated, washed with brine (20 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , petroleum ether : ethyl acetate = 0:1) to give tert-butyl 6-[8- (1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin- 2-yl]-3-[4-[3-[1-(2-ethoxy-2- oxo-ethyl)-4-fluoro-4-piperidyl]propoxy]-2-methyl-phenyl]pyr idine-2-carboxylate (240 mg, 268.61 μmol, 69.89% yield) as a yellow solid. MS (ESI) m/z: 822.4 [M+H] + . Step I. Procedure for preparation of 2-[4-[3-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyr idyl]-3-methyl- phenoxy]propyl]-4-fluoro-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[4-[3-[1-(2-ethoxy-2-oxo-ethyl)-4-fluoro-4-piperidyl ]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (240 mg, 291.97 μmol, 1.0 equiv.) in THF (2.4 mL) and H 2 O (0.8 mL) was added LiOH•H2O (36.76 mg, 875.92 μmol, 3.0 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was acidified to pH = 3-4 with citric acid and partitioned between DCM (30 mL) and water (15 mL). The organic phase was separated, washed with brine (15 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give 2-[4-[3-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-3-methyl- phenoxy]propyl]-4-fluoro-1- piperidyl]acetic acid (230 mg, crude) as a white solid. MS (ESI) m/z: 794.7 [M+H] + . Step J. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[4-[3-[1-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-4-fluoro-4-piperidyl]propoxy]-2-met hyl-phenyl]pyridine-2- carboxylate To a solution of 2-[4-[3-[4-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-3-methyl- phenoxy]propyl]-4-fluoro-1- piperidyl]acetic acid (230 mg, 289.69 μmol, 1.0 equiv.) and 3-(6-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (149.64 mg, 579.39 μmol, 2.0 equiv.) in pyridine (2 mL) was added EDCI (111.07 mg, 579.39 μmol, 2.0 equiv.). The mixture was stirred at 40 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM : MeOH = 20:1) to give tert-butyl 6-[8-(1,3-benzothiazol- 2-ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[4-[3-[1-[ 2-[[3-(2,6-dioxo-3-piperidyl)- 1-methyl-indazol-6-yl]amino]-2-oxo-ethyl]-4-fluoro-4-piperid yl]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (230 mg, 206.83 μmol, 71.39% yield) as a yellow solid. MS (ESI) m/z: 1034.4 [M+H] + . Step K. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[4-[3-[1-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-fluoro-4-piperidyl]propoxy]-2-methy l-phenyl]pyridine-2- carboxylic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[4-[3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indaz ol-6-yl]amino]-2-oxo-ethyl]-4- fluoro-4-piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carbo xylate (230 mg, 222.39 μmol, 1.0 equiv.) in TFA (2 mL) and DCM (2 mL) was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product was purified by reverse-phase HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[4-[3-[1-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-fluoro-4-piperidyl]propoxy]-2-methy l-phenyl]pyridine-2- carboxylic acid (114.06 mg, 108.34 μmol, 48.72% yield) as a yellow solid. MS (ESI) m/z: 978.6 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.14 - 12.29 (m, 2H), 10.97 - 10.70 (m, 1H), 9.96 - 9.66 (m, 1H), 8.06 - 8.00 (m, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.66 - 7.59 (m, 2H), 7.49 - 7.41 (m, 3H), 7.39 - 7.33 (m, 2H), 7.22 (d, J = 9.2 Hz, 1H), 6.91 (s, 2H), 6.79 (d, J = 2.4 Hz, 1H), 6.71 (dd, J = 2.4, 8.4 Hz, 1H), 4.96 (s, 2H), 4.36 - 4.27 (m, 1H), 4.00 - 3.94 (m, 2H), 3.93 - 3.88 (m, 5H), 3.22 - 3.17 (m, 2H), 3.02 (t, J = 5.6 Hz, 2H), 2.77 - 2.58 (m, 4H), 2.46 - 2.42 (m, 2H), 2.37 - 2.28 (m, 2H), 2.17 ( dd, J = 5.2, 13.2 Hz, 1H), 2.02 (s, 3H), 1.80 ( s, 5H), 1.76 - 1.70 (m, 2H) 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2-yl]-3-[3-[3-[1-[2-[[3- (2,6-dioxo-3-piperidyl)-1-methyl-indazol-6-yl]amino]-2-oxo-e thyl]-4-fluoro-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylic acid Step A. Procedure for preparation of tert-butyl 4-[3-(3-bromo-2-methyl-phenoxy)propyl]- 4-fluoro-piperidine-1-carboxylate A mixture of tert-butyl 4-fluoro-4-(3-hydroxypropyl)piperidine-1-carboxylate (460 mg, 1.76 mmol, 1.0 equiv.), 3-bromo-2-methyl-phenol (362.14 mg, 1.94 mmol, 1.1 equiv.), and 2- (tributyl-phosphanylidene)acetonitrile (637.25 mg, 2.64 mmol, 1.5 equiv.) in toluene (20 mL) was degassed and purged with N2 three times. The mixture was stirred at 125 °C for 12 hours under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~1% ethyl acetate/petroleum ether) to give tert-butyl 4-[3-(3-bromo-2-methyl-phenoxy)propyl]-4-fluoro- piperidine-1-carboxylate (670 mg, 1.56 mmol, 88.45% yield) as a yellow oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ = 7.16 (d, J = 6.8 Hz, 1H), 7.10 (d, J = 7.6 Hz, 1H), 6.96 (d, J = 8.0 Hz, 1H), 4.11 - 3.86 (m, 3H), 3.81 - 3.57 (m, 2H), 2.99 (d, J = 3.2 Hz, 1H), 2.31 - 2.18 (m, 3H), 1.88 - 1.74 (m, 5H), 1.66 - 1.51 (m, 2H), 1.41 (s, 9H), 1.11 - 0.91 (m, 1H) Step B. Procedure for preparation of 4-[3-(3-bromo-2-methyl-phenoxy)propyl]-4-fluoro- piperidine To a solution of tert-butyl 4-[3-(3-bromo-2-methyl-phenoxy)propyl]-4-fluoro-piperidine-1 - carboxylate (670 mg, 1.56 mmol, 1.0 equiv.) in dioxane (3 mL) was added HCl/dioxane (5 mL). The mixture was stirred at 25 °C for 12 hours. The reaction mixture was concentrated under reduced pressure to remove to give 4-[3-(3-bromo-2-methyl-phenoxy)propyl]-4-fluoro- piperidine (530 mg, crude) as a white solid. MS (ESI) m/z: 332.1 [M+H] + . Step C. Procedure for preparation of ethyl 2-[4-[3-(3-bromo-2-methyl-phenoxy)propyl]- 4-fluoro-1-piperidyl]acetate To a solution of 4-[3-(3-bromo-2-methyl-phenoxy)propyl]-4-fluoro-piperidine (530 mg, 1.60 mmol, 1.0 equiv.) and ethyl 2-bromoacetate (268.02 mg, 1.60 mmol, 177.62 μL, 1.0 equiv.) in DMF (5 mL) was added K 2 CO 3 (665.43 mg, 4.81 mmol, 3.0 equiv.). The mixture was stirred at 40 °C for 2.5 hours. The reaction mixture was concentrated under reduced pressure to give ethyl 2-[4-[3-(3-bromo-2-methyl-phenoxy)propyl]-4-fluoro-1-piperid yl]acetate (620 mg, crude) as a yellow oil. MS (ESI) m/z: 416.2 [M+H] + . Step D. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-(2-ethoxy-2-oxo- ethyl)-4-fluoro-4- piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate To a solution of ethyl 2-[4-[3-(3-bromo-2-methyl-phenoxy)propyl]-4-fluoro-1- piperidyl]acetate (570 mg, 1.37 mmol, 1.0 equiv.) and tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-(4,4,5,5-tet ramethyl-1,3,2-dioxaborolan-2- yl)pyridine-2-carboxylate (838.65 mg, 1.37 mmol, 1.0 equiv.) in dioxane (3 mL) and H 2 O (0.3 mL) was added KF (238.63 mg, 4.11 mmol, 96.22 μL, 3.0 equiv.) and Ad2nBuP Pd G3 (199.42 mg, 273.83 μmol, 0.2 equiv.). The mixture was stirred at 100 °C for 1 hour. The reaction mixture was partitioned between ethyl acetate (30 mL) and water (20 mL). The organic phase was separated, washed with brine (20 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~45% ethyl acetate/petroleum ether) to give tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-(2- ethoxy-2-oxo-ethyl)-4-fluoro- 4-piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carboxylate (530 mg, 638.32 μmol, 46.62% yield) as a yellow oil. MS (ESI) m/z: 822.4 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ = 8.02 ( d, J = 7.6 Hz, 1H), 7.78 (d, J = 8.4 Hz, 1H), 7.60 (d, J = 7.2 Hz, 1H), 7.45 (d, J = 8.4 Hz, 5H), 7.14 - 7.04 (m, 1H), 6.93 (dd, J = 8.4, 14.4 Hz, 2H), 6.58 (d, J = 6.8 Hz, 1H), 5.07 - 4.88 (m, 2H), 4.09 - 3.86 (m, 6H), 3.25 - 3.18 (m, 2H), 3.08 - 2.99 (m, 2H), 2.65 - 2.60 (m, 2H), 2.41 - 2.36 (m, 2H), 1.99 (s, 1H), 1.88 (s, 2H), 1.84 - 1.66 (m, 7H), 1.21 - 1.14 (m, 4H), 1.00 (s, 9H) Step E. Procedure for preparation of 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-1H-isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyr idyl]-2-methyl- phenoxy]propyl]-4-fluoro-1-piperidyl]acetic acid To a solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin- 2-yl]-3-[3-[3-[1-(2-ethoxy-2-oxo-ethyl)-4-fluoro-4-piperidyl ]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (200 mg, 243.31 μmol, 1.0 equiv.) in THF (2.1 mL) and H 2 O (0.7 mL) was added LiOH·H2O (30.63 mg, 729.93 μmol, 3.0 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was acidified to pH = 5-6 with citric acid and partitioned between DCM (20 mL) and water (15 mL). The organic phase was separated, washed with brine (15 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]propyl]-4-fluoro-1- piperidyl]acetic acid (120 mg, 151.14 μmol, 62.12% yield) as a white solid. MS (ESI) m/z: 794.4 [M+H] + . Step F. Procedure for preparation of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3- piperidyl)-1-methyl-indazol- 6-yl]amino]-2-oxo-ethyl]-4-fluoro-4-piperidyl]propoxy]-2-met hyl-phenyl]pyridine-2- carboxylate To a solution of 2-[4-[3-[3-[6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydr o-1H- isoquinolin-2-yl]-2-tert-butoxycarbonyl-3-pyridyl]-2-methyl- phenoxy]propyl]-4-fluoro-1- piperidyl]acetic acid (120 mg, 151.14 μmol, 1.0 equiv.) and 3-(6-amino-1-methyl-indazol-3- yl)piperidine-2,6-dione (58.56 mg, 226.72 μmol, 1.5 equiv.) in pyridine (1 mL) was added EDCI (57.95 mg, 302.29 μmol, 2.0 equiv.). The mixture was stirred at 40 °C for 4 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , DCM: MeOH = 20:1) to give tert-butyl 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-1H-isoquinolin-2-yl]-3-[3-[3-[1-[2- [[3-(2,6-dioxo-3-piperidyl)-1- methyl-indazol-6-yl]amino]-2-oxo-ethyl]-4-fluoro-4-piperidyl ]propoxy]-2-methyl- phenyl]pyridine-2-carboxylate (110 mg, 104.23 μmol, 68.96% yield) as a yellow solid. MS (ESI) m/z: 1034.7 [M+H] +. Step G. Procedure for preparation of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro- 1H-isoquinolin-2-yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3-piperidy l)-1-methyl-indazol-6- yl]amino]-2-oxo-ethyl]-4-fluoro-4-piperidyl]propoxy]-2-methy l-phenyl]pyridine-2- carboxylic acid A solution of tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indaz ol-6-yl]amino]-2-oxo-ethyl]-4- fluoro-4-piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carbo xylate (90 mg, 87.02 μmol, 1.0 equiv.) in TFA (1 mL) and DCM (1 mL) was stirred at 40 °C for 1.5 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by reverse- phase HPLC to give 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-1H-isoquin olin-2- yl]-3-[3-[3-[1-[2-[[3-(2,6-dioxo-3-piperidyl)-1-methyl-indaz ol-6-yl]amino]-2-oxo-ethyl]-4- fluoro-4-piperidyl]propoxy]-2-methyl-phenyl]pyridine-2-carbo xylic acid (36.35 mg, 34.56 μmol, 39.71% yield) as a yellow solid. MS (ESI) m/z: 978.3 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 10.87 (s, 1H), 9.86 (s, 1H), 8.06 - 8.00 (m, 2H), 7.78 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 8.8 Hz, 2H), 7.50 - 7.42 (m, 3H), 7.40 - 7.32 (m, 2H), 7.22 (dd, J = 1.2, 8.8 Hz, 1H), 7.10 (s, 1H), 6.96 (d, J = 9.2 Hz, 1H), 6.88 (d, J = 8.0 Hz, 1H), 6.68 - 6.58 (m, 1H), 4.98 (s, 2H), 4.32 (dd, J = 5.2, 9.6 Hz, 1H), 4.03 - 3.95 (m, 2H), 3.94 - 3.88 (m, 5H), 3.18 (s, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.77 - 2.69 (m, 2H), 2.68 - 2.60 (m, 2H), 2.45 (d, J = 3.6 Hz, 2H), 2.36 - 2.31 (m, 1H), 2.21 - 2.13 (m, 1H), 1.91 (s, 3H), 1.88 - 1.72 (m, 8H). Example 316. Preparation of Compound 271b 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-((2S)-3-(1-(2- ((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino )-2-oxoethyl)piperidin-4-yl)-2- methylpropoxy)-2-methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquin olin-2(1H)-yl)-3-(4-((2S)-3-(1- (2-((3-(2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)am ino)-2-oxoethyl)piperidin-4- yl)-2-methylpropoxy)-2-methylphenyl)picolinic acid] Step A. Procedure for preparation of (S)-4-(3-(4-bromo-3-methylphenoxy)-2- methylpropyl)piperidine To a solution of tert-butyl (S)-4-(3-(4-bromo-3-methylphenoxy)-2-methylpropyl)piperidine -1- carboxylate (1 g, 2.35 mmol, 1 equiv.) was added HCl/EtOAc (4 M, 586.32 µL, 1 equiv.). The mixture was stirred at 25 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give (S)-4-(3-(4-bromo-3-methylphenoxy)-2- methylpropyl)piperidine (800 mg, crude) as a white solid. MS (ESI) m/z: 326.0 [M+H] + . Step B. Procedure for preparation of ethyl (S)-2-(4-(3-(4-bromo-3-methylphenoxy)-2- methylpropyl)piperidin-1-yl)acetate To a solution of (S)-4-(3-(4-bromo-3-methylphenoxy)-2-methylpropyl)piperidine (300 mg, 919.48 µmol, 1 equiv.), ethyl 2-bromoacetate (153.55 mg, 919.48 µmol, 101.69 µL, 1 equiv.) in MeCN (4 mL) was added K2CO3 (381.23 mg, 2.76 mmol, 3 equiv.). The mixture was stirred at 40 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give ethyl (S)-2-(4-(3-(4-bromo-3-methylphenoxy)-2-methylpropyl)piperid in-1-yl)acetate (300 mg, crude) as a white solid. MS (ESI) m/z: 412.0 [M+H] + . Step C. Procedure for preparation of tert-butyl (S)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(4-(3-(1-(2-ethoxy-2-oxoe thyl)piperidin-4-yl)-2- methylpropoxy)-2-methylphenyl)picolinate A mixture of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)- yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)picolinat e (222.82 mg, 363.76 µmol, 1 equiv.), ethyl (S)-2-(4-(3-(4-bromo-3-methylphenoxy)-2-methylpropyl)piperid in-1-yl)acetate (150 mg, 363.76 µmol, 1 equiv.) in dioxane (3 mL) was added KF (1.5 M, 727.52 µL, 3 equiv.) and Ad 2 nBuP Pd G 3 (cataCXium® A Pd G 3 ) (26.49 mg, 36.38 µmol, 0.1 equiv.). The reaction mixture was degassed and purged with N 2 three times. The mixture was stirred at 100 °C for 2 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (0~38% ethyl acetate/petroleum ether) to give tert-butyl (S)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(3-(1-(2-ethoxy-2-oxoethyl )piperidin-4-yl)-2- methylpropoxy)-2-methylphenyl)picolinate (180 mg, 184.53 µmol, 50.73% yield) as a yellow solid. MS (ESI) m/z: 818.4 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ = 7.84 (d, J = 8.0 Hz, 1H), 7.65 - 7.48 (m, 2H), 7.42 - 7.27 (m, 5H), 7.02 - 6.83 (m, 2H), 6.77 (d, J = 2.0 Hz, 1H), 6.69 (dd, J = 2.8, 5.6 Hz, 1H), 5.04 (s, 2H), 4.21 (q, J = 7.2 Hz, 2H), 4.09 (t, J = 6.0 Hz, 2H), 3.87 - 3.66 (m, 2H), 3.36 (s, 2H), 3.16 - 2.98 (m, 4H), 2.17 - 1.99 (m, 5H), 1.54 - 1.43 (m, 4H), 1.33 - 1.22 (m, 5H), 1.19 (s, 9H), 1.11 (s, 2H), 1.02 (d, J = 6.4 Hz, 3H) Step D. Procedure for preparation of (S)-2-(4-(3-(4-(6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-2-(tert-butoxy carbonyl)pyridin-3-yl)-3- methylphenoxy)-2-methylpropyl)piperidin-1-yl)acetic acid To a solution of tert-butyl (S)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquino lin- 2(1H)-yl)-3-(4-(3-(1-(2-ethoxy-2-oxoethyl)piperidin-4-yl)-2- methylpropoxy)-2- methylphenyl)picolinate (180 mg, 220.04 µmol, 1 equiv.) in THF (2 mL) was added LiOH•H 2 O (27.70 mg, 660.12 µmol, 3 equiv.) and H 2 O (0.5 mL). The mixture was stirred at 25 °C for 1.5 hours. The reaction mixture was concentrated under reduced pressure to remove THF. The aqueous phase was adjusted to pH = 4~5 with 1M HCl. The reaction mixture was filtered. The filter cake was diluted in ethyl acetate. The combined organic layers were dried over by Na2SO4, filtered and concentrated under reduced pressure to give (S)-2-(4-(3-(4-(6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-3-methylphenoxy)-2-methylpropyl )piperidin-1-yl)acetic acid (170 mg, crude) as a yellow solid. MS (ESI) m/z: 790.7 [M+H] + . Step E. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-((2S)-3-(1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)-2-methylpr opoxy)-2- methylphenyl)picolinate To a solution of (S)-2-(4-(3-(4-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dih ydroisoquinolin- 2(1H)-yl)-2-(tert-butoxycarbonyl)pyridin-3-yl)-3-methylpheno xy)-2-methylpropyl)piperidin- 1-yl)acetic acid (160 mg, 202.54 µmol, 1 equiv.) and 3-(6-amino-1-methyl-1H-indazol-3- yl)piperidine-2,6-dione (52.31 mg, 202.54 µmol, 1 equiv.) in pyridine (2 mL) was added EDCI (58.24 mg, 303.81 µmol, 1.5 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was quenched by the addition of H2O (2 mL). The reaction mixture was filtered, and the filter cake was washed with water (5 mL). The filter cake was then diluted in DCM (10 mL). The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-((2S)-3-(1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl-1H- indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)-2-methylpropo xy)-2-methylphenyl)picolinate (160 mg, crude) as a yellow solid. MS (ESI) m/z: 1030.4 [M+H] + . Step F. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-((2S)-3-(1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)-2-methylpr opoxy)-2- methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-((2S)-3-(1-(2-((3-(2,6-dio xopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)-2-methylpr opoxy)-2- methylphenyl)picolinic acid] To a solution of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-((2S)-3-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-1 -methyl-1H-indazol-6-yl)amino)- 2-oxoethyl)piperidin-4-yl)-2-methylpropoxy)-2-methylphenyl)p icolinate (160 mg, 155.30 µmol, 1 equiv.) in DCM (1 mL) was added TFA (1.54 g, 13.51 mmol, 1 mL, 86.97 equiv.). The mixture was stirred at 25 °C for 1 hour. The reaction mixture was filtered and concentrated under reduced pressure to give a residue which was purified by prep-HPLC to give 6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H) -yl)-3-(4-((2S)-3-(1-(2-((3- (2,6-dioxopiperidin-3-yl)-1-methyl-1H-indazol-6-yl)amino)-2- oxoethyl)piperidin-4-yl)-2- methylpropoxy)-2-methylphenyl)picolinic acid [rel-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)-3-(4-((2S)-3-(1-(2-((3-(2,6 -dioxopiperidin-3-yl)-1-methyl- 1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)-2-methylpr opoxy)-2- methylphenyl)picolinic acid] (61.56 mg, 59.84 µmol, 58.29% yield) as a yellow solid. MS (ESI) m/z: 974.3 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ = 13.28 - 12.39 (m, 1H), 10.89 (s, 1H), 10.66 - 10.20 (m, 1H), 8.14 (s, 1H), 8.07 - 7.98 (m, 2H), 7.79 (d, J = 8.0 Hz, 1H), 7.72 - 7.58 (m, 2H), 7.51 - 7.41 (m, 3H), 7.40 - 7.32 (m, 2H), 7.17 (d, J = 8.8 Hz, 1H), 6.94 (dd, J = 8.8, 4.0 Hz, 2H), 6.80 (s, 1H), 6.75 - 6.67 (m, 1H), 4.97 (s, 2H), 4.34 (dd, J = 4.8, 4.8 Hz, 1H), 3.97 - 3.88 (m, 5H), 3.87 - 3.66 (m, 4H), 3.02 (t, J = 5.2 Hz, 2H), 2.91 - 2.51 (m, 5H), 2.41 - 2.28 (m, 1H), 2.19 - 2.15 (m, 1H), 2.08 - 1.93 (m, 4H), 1.81 (d, J = 11.2 Hz, 2H), 1.66 - 1.52 (m, 1H), 1.51 - 1.08 (m, 5H), 1.05 - 0.93 (m, 3H). Example 317. Preparation of Compound 356 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(3-(1-(2-((3- (2,6-dioxopiperidin-3-yl)-7-fluoro-1-methyl-1H-indazol-6-yl) amino)-2-oxoethyl)piperidin-4- yl)propoxy)-2-methylphenyl)picolinic acid Step A. Procedure for preparation of tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(3-(1-(2-((3-(2,6-dioxopip eridin-3-yl)-7-fluoro-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)prop oxy)-2- methylphenyl)picolinate 2-(4-(3-(4-(6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro isoquinolin-2(1H)-yl)-2-(tert- butoxycarbonyl)pyridin-3-yl)-3-methylphenoxy)propyl)piperidi n-1-yl)acetic acid (94 mg, 1 equiv.0.12 mmol) was dissolved in DCM (5 mL) and DMF (1 mL). To the mixture was added HATU (51 mg, 1.1 equiv.0.13 mmol), followed by 3-(6-amino-7-fluoro-1-methyl-1H-indazol- 3-yl)piperidine-2,6-dione (40 mg, 1.2 equiv.0.15 mmol) and DIEA (39 mg, 53 μL, 2.5 equiv. 0.30 mmol). The mixture was stirred at 40ºC for 48 hrs. The mixture was concentrated to dryness to afford tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(3-(1-(2-((3-(2,6-dioxopiperidin-3-yl)-7-fluo ro-1-methyl-1H-indazol-6- yl)amino)-2-oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl) picolinate, which was carried forward without further purification. MS (ESI) m/z: 517.8 [M+2H] 2+ . Step B. Procedure for preparation of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(4-(3-(1-(2-((3-(2,6-dioxopip eridin-3-yl)-7-fluoro-1- methyl-1H-indazol-6-yl)amino)-2-oxoethyl)piperidin-4-yl)prop oxy)-2- methylphenyl)picolinic acid tert-Butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(3-(1- (2-((3-(2,6-dioxopiperidin-3-yl)-7-fluoro-1-methyl-1H-indazo l-6-yl)amino)-2- oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinate (0.12 g, 1 equiv.0.12 mmol) was dissolved in DCM (4 mL). To the mixture was added TFA (1 mL). The mixture was stirred at room temperature overnight. The mixture was concentrated, and purified by RP-HPLC to afford 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(4-(3-(1-(2- ((3-(2,6-dioxopiperidin-3-yl)-7-fluoro-1-methyl-1H-indazol-6 -yl)amino)-2- oxoethyl)piperidin-4-yl)propoxy)-2-methylphenyl)picolinic acid (24 mg, 25 μmol, 20 %) as a white solid. MS (ESI) m/z: 978.5 [M+H] + . 1 H NMR (400 MHz, DMSO) δ 10.84 (s, 1H), 9.65 (s, 1H), 7.96 (d, J = 7.9 Hz, 1H), 7.72 (d, J = 8.0 Hz, 1H), 7.59 – 7.48 (m, 2H), 7.45 – 7.34 (m, 4H), 7.34 – 7.23 (m, 2H), 6.86 (t, J = 8.8 Hz, 2H), 6.72 (d, J = 2.6 Hz, 1H), 6.64 (dd, J = 8.4, 2.6 Hz, 1H), 4.90 (s, 2H), 4.30 (dd, J = 10.3, 5.0 Hz, 1H), 4.04 (s, 2H), 3.86 (dt, J = 15.3, 6.1 Hz, 4H), 3.09 (s, 2H), 2.95 (t, J = 6.0 Hz, 2H), 2.85 (d, J = 11.0 Hz, 2H), 2.69 – 2.45 (m, 2H), 2.36 – 2.26 (m, 1H), 2.15 – 2.06 (m, 3H), 1.96 (s, 3H), 1.66 (d, J = 8.8 Hz, 4H), 1.32 (s, 2H), 1.22 (s, 3H). The following examples were synthesized using methods similar to those described in Examples 1-317, above.

Example P1 Resolution of stereoisomeric mixtures Mixtures of stereoisomers were resolved using methods described in Table P1: Table P1

Example B1 HiBiT based degradation assay Human BCL-XL protein coding open reading frame fused with N-terminal HiBiT coding sequence was synthesized from Integrated DNA Technologies (IDT). Next, N-HiBiT- BCL-XL sequence was cloned into pLV-UBC-PGK-Puro, a lentivirus plasmid purchased from Vectorbuilder, to generate pLV-UBC-N-HiBiT-BCL-XL-PGK-puro. Lentiviral particles were generated from Lenti-X™ 293T cells (Clontech) by co-transfection of pLV-UBC-N-HiBiT- BCL-XL-PGK-puro plasmids and lentiviral packaging plasmid mix (Cellecta). HT-1080 [HT1080] (ATCC CCL-121) cells were infected with the lentivirus, and HT1080 cells stably integrated with the lentiviral vectors were established by incubation with 1 μg/mL puromycin (Thermofisher). HT1080 cells expressing N-terminal HiBiT tagged BCL-XL were dispensed into a 384- well plate pre-spotted with compounds at varying concentrations. Three thousand cells were seeded into each well in 40 µL of RPMI 1640 media. After 6 hours of incubation at 37°C with 5% CO 2 , 30 µL of the NANO-GLO® HiBiT Lytic Detection System working solution (Promega) was added to each well and incubated at room temperature for 30 min. After incubation, luminescence was read on a PHERAstar FSX Plate Reader (BMG). BCL-X L degradation at each indicated concentration was normalized with the DMSO control. The BCL- XL degradation curves were plotted using a four-parameter logistic model. For compounds displaying hook effects, the curve fitting was performed using the bottom point of the hook curves (i.e., the point of Y min ) as the bottom plateau to calculate EC 50 . Y min and EC 50 values for certain compounds are shown in Table B1. Table B1

Notes: (1) “A”: Y min < 50%; “B”: 50% ≤Y min <70%; “C”: Y min ≥ 70% (2) “+++”: EC50 < 100 nM; : 100 nM ≤ EC50 < 1000 nM; “ : 1000 nM ≤ EC50 < 10,000 nM Example B2 Western Blotting based degradation assay BCL-X L degradation in human cancer cell lines is measured using traditional Western blotting methods as well as with the automated “Jess” instrument from ProteinSimple. Cells are seeded at 0.5-1 million cells per well in up to 2 mL of growth media (RPMI + 10% FBS) in 6-well plates. The plates are treated with compounds at varying concentrations for up to 5 days. Cells are harvested and lysed using RIPA Buffer (#9806S, Cell Signaling Technology) with Halt Protease & Phosphatase Inhibitor Cocktail (#78441, Thermo Fisher). Protein concentrations of each sample are quantified using Pierce™ BCA Protein Assay Kit (#23225, Thermo Fisher). For the Jess instrument the standard protocol provided by the manufacturer using the Jess Separation Module SM-W004 is followed. For traditional Western blotting, samples resolved on a 4-20% Criterion TGX gel (#5671094, BioRad) and transferred to a PVDF membrane using the Trans-Blot Turbo Transfer system (#1704157, BioRad). Chemiluminescent detection of BCL-XL is performed using a BCL-XL antibody (#2764, Cell Signaling Technology) according to the manufacturer’s instructions. Example B3 MOLT-4 proliferation assay MOLT-4 (ATCC CRL-1582) cells are seeded at 4,000 cells per well in 50 μL RPMI 1640 media in black 384-well plates. The plates are pre-spotted with compounds at varying concentrations. After 3 days of incubation at 37 °C with 5% CO2, cells are assessed using CELLTITERGLO® Cell Viability Assay kit according to manufacturer’s instructions (Promega). Relative cell proliferation at each concentration is normalized against the DMSO control. Example B4 Cell line proliferation assay Human cancer cell lines are seeded at 1,000 – 4,000 cells per well in 50 µL growth media (following culture conditions recommended by cell line sources) in 384-well plates. The plates are treated with compounds at varying concentrations using a D300e Digital Dispenser (Tecan) and then incubated at 37 °C with 5% CO 2 for 3-7 days. Relative cell viability is assessed using CELLTITERGLO® Cell Viability Assay (#G9243, Promega) according to the manufacturer’s instructions or Incucyte SX5 Live-Cell Analysis Instrument (Sartorius) with images recorded at 4-12 hour intervals. Example B5 Apoptosis assay Human cancer cell lines are seeded at 1,000 – 4,000 cells per well in 50 µL growth media (following culture conditions recommended by cell line sources) in 384-well plates. The plates are treated with compounds at varying concentrations using a D300e Digital Dispenser (Tecan) and then incubated at 37 °C with 5% CO2 for 3-7 days. Relative caspase-3/7 activity is assessed using Caspase-Glo 3/7 Assay System (#G8093, Promega) according to the manufacturer’s instructions or Incucyte SX5 Live-Cell Analysis Instrument (Sartorius) with Green Incucyte Caspase-3/7 Dye (#4440, Sartorius) added to the media at between 1:500 and 1:1000 dilution at the time of seeding and images recorded at 4-12 hour intervals. Relative annexin V positivity is assessed using Incucyte SX5 Live-Cell Analysis Instrument (Sartorius) with Orange Incucyte Annexin V Dye (#4759, Sartorius) added to the media at a 1:200 dilution at the time of seeding and images recorded at 4-12 hour intervals. Example B6 Platelet viability assay Human platelet-rich plasma (PRP, SER-PRP-SDS) is obtained from Zenbio. Sodium citrate is used as anticoagulant. To remove any contaminating blood cells, the samples are spun at 200 x g at room temperature for 20 minutes in 50 mL falcon tubes, and the supernatant is transferred to a new tube. One hundred microliter PRP are added to each well of a 96-well plate spotted with compounds at varying concentrations. Plates are incubated at 37 °C with 5% CO 2 for 24 hours, and platelet viability are read with CELLTITERGLO® Cell Viability Assay kit according to manufacturer’s instructions (Promega). Relative viability at each concentration is normalized against the DMSO control. EXEMPLARY EMBODIMENTS P01 Embodiments Embodiment 1. A compound of Formula (I) or (II): Formula (II) or a pharmaceutically acceptable salt thereof, wherein: Ring A is selected from the group consisting of: (a) C3-15 cycloalkylene or 3-15 membered heterocyclylene, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; and (b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 substituents independently selected from the group consisting of: R a and L T1 is a bond or C1-3 alkylene optionally substituted with 1-3 substituents independently selected from the group consisting of: oxo and R c , wherein one CH 2 unit of the C 1-3 alkylene is optionally replaced with -O- or -N(R d )-; A* is selected from the group consisting of: (a) C 3-15 cycloalkyl or 3-15 membered heterocyclyl, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; (b) C 6-15 aryl or 5-15 membered heteroaryl, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; and (c) H; R 1 is selected from the group consisting of: (a) C(O)OH, (b) C(O)NR d R e , and (c) C(O)OC 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R c ; each R 2 , R 3 , R 4 , and R 5 is independently selected from the group consisting of: halo, CN, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, OH, and NR d R e ; m2 is 0, 1, or 2; m3 and m4 are independently 0, 1, 2, or 3; m5 is 0, 1, 2, 3, or 4; L is –(L A )n1–, wherein L A and n1 are defined according to (AA) or (BB): (AA) n1 is an integer from 3 to 15; and each L A is independently selected from the group consisting of: L A1 , L A3 , and L A4 , provided that 1-3 occurrences of L A is L A4 ; (BB) n1 is an integer from 0 to 20; and each L A is independently selected from the group consisting of: L A1 and L A3 ; each L A1 is independently selected from the group consisting of: -CH2-, -CHR L -, and - C(R L ) 2 -; each L A3 is independently selected from the group consisting of: -N(R d )-, -N(R b )-, -O- , -S(O)0-2-, and C(=O); each L A4 is independently selected from the group consisting of: (a) C3-15 cycloalkylene or 3-15 membered heterocyclylene, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; and (b) C6-15 arylene or 5-15 membered heteroarylene, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; provided that L does not contain any N-O, N-N, N-S(O) 0 , or O-S(O) 0-2 bonds; wherein each R L is independently selected from the group consisting of: halo; cyano; - OH; -C1-6 alkoxy; -C1-6 haloalkoxy; -NR d R e ; C(=O)N(R f )2; S(O)0-2(C1-6 alkyl); S(O)0-2(C1-6 haloalkyl); S(O) 1-2 N(R f ) 2 ; -R b ; and C 1-6 alkyl optionally substituted with 1-6 R c ; Ring C is selected from the group consisting of: ; c1 is 0, 1, 2, or 3; each R Y is independently selected from the group consisting of: R a and R b ; R aN is H or C1-6 alkyl optionally substituted with 1-3 R c ; Y 1 and Y 2 are independently N, CH, or CR Y ; yy represents the point of attachment to L; X is CH, C, or N; the is a single bond or a double bond; L C is selected from the group consisting of: a bond, -CH 2 -, -CHR a -, -C(R a ) 2 -, -N(R d )-, and O, provided that when X is N, then L C is other than O; each R a is independently selected from the group consisting of: (a) Halo; (b) cyano; (c) -OH; (d) oxo; (e) -C1-6 alkoxy; (f) -C 1-6 haloalkoxy; (g) -NR d R e ; (h) C(=O)C1-6 alkyl; (i) C(=O)C 1-6 haloalkyl; (j) C(=O)OH; (k) C(=O)OC1-6 alkyl; (l) C(=O)OC 1-6 haloalkyl; (m) C(=O)N(R f ) 2 ; (n) S(O)0-2(C1-6 alkyl); (o) S(O)0-2(C1-6 haloalkyl); (p) S(O) 1-2 N(R f ) 2 ; and (q) C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted with 1-6 R c ; each R b is independently selected from the group consisting of: -(L b )b-R b1 and -R b1 , wherein: each b is independently 1, 2, or 3; each -L b is independently selected from the group consisting of: -O-; -N(H)-; -N(C 1-3 alkyl)-; -S(O) 0-2 -; C(=O); and C 1-3 alkylene; and each R b1 is independently selected from the group consisting of: C3-10 cycloalkyl, 4-10 membered heterocyclyl, C6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 R g ; each R c is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C 1-6 haloalkoxy; -NR d R e ; C(=O)C 1-6 alkyl; C(=O)C 1-6 haloalkyl; C(=O)OC 1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)OH; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); and S(O)1-2N(R f )2; each R d and R e is independently selected from the group consisting of: H; C(=O)C1-6 alkyl; C(=O)C 1-6 haloalkyl; C(=O)OC 1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)N(R f ) 2 ; S(O) 1- 2(C1-6 alkyl); S(O)1-2(C1-6 haloalkyl); S(O)1-2N(R f )2; and C1-6 alkyl optionally substituted with 1-3 R h ; each R f is independently selected from the group consisting of: H and C 1-6 alkyl optionally substituted with 1-3 R h ; each R g is independently selected from the group consisting of: R h ; C1-3 alkyl; and C1- 3 haloalkyl; and each R h is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C1-6 haloalkoxy; -NH2, -N(H)(C1-3 alkyl); and -N(C1-3 alkyl)2. Embodiment 2. The compound of Embodiment 1, wherein the compound is a compound of Formula (I) or a pharmaceutically acceptable salt thereof. Embodiment 3. The compound of Embodiment 1, wherein the compound is a compound of Formula (II) or a pharmaceutically acceptable salt thereof. Embodiment 4. The compound of any one of Embodiments 1-3, wherein Ring A is a phenylene optionally substituted with 1-3 R a . Embodiment 5. The compound of any one of Embodiments 1-4, wherein Ring A is , wherein aa represents the point of attachment to L or L T1 . Embodiment 6. The compound of any one of Embodiments 1-5, wherein Ring is , wherein aa represents the point of attachment to L or L T1 . Embodiment 7. The compound of any one of Embodiments 1-5, wherein Ring A is , wherein aa represents the point of attachment to L or L T1 . Embodiment 8. The compound of any one of Embodiments 1-3, wherein Ring A is 5-6 membered heteroarylene optionally substituted with 1-3 R a . Embodiment 9. The compound of any one of Embodiments 1-3 or 8, wherein Ring A is 5-6 membered heteroarylene optionally substituted with 1-2 R a . Embodiment 10. The compound of any one of Embodiments 1-3 or 8-9, wherein Ring A is 5-membered heteroarylene optionally substituted with 1-2 R a . Embodiment 11. The compound of any one of Embodiments 1-3 or 8-10, wherein Ring A is pyrazolylene optionally substituted with 1-2 R a . Embodiment 12. The compound of any one of Embodiments 1-3 or 8-11, wherein Ring A is selected from the group consisting of: , wherein aa represents the point of attachment to L or L T1 . Embodiment 13. The compound of any one of Embodiments 1-3, wherein Ring A is C 3-10 cycloalkylene optionally substituted with 1-6 R a . Embodiment 14. The compound of any one of Embodiments 1-13, wherein one R a present on Ring A is C 1-3 alkyl optionally substituted with 1-3 F. Embodiment 15. The compound of any one of Embodiments 1-14, wherein one R a present on Ring A is methyl or CF 3 . Embodiment 16. The compound of any one of Embodiments 1-2 or 4-15, wherein R 1 is C(O)OH. Embodiment 17. The compound of any one of Embodiments 1-2 or 4-15, wherein R 1 is C(O)NHR e . Embodiment 18. The compound of any one of Embodiments 1 or 3-15, wherein L T1 is C 1-3 alkylene. Embodiment 19. The compound of any one of Embodiments 1, 3-15, or 18, wherein L T1 is -CH 2 -. Embodiment 20. The compound of any one of Embodiments 1, 3-15, or 18-19, wherein A* is H. Embodiment 21. The compound of any one of Embodiments 1, 3-15, or 18-20, wherein A* is C3-15 cycloalkyl or 3-15 membered heterocyclyl, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b . Embodiment 22. The compound of any one of Embodiments 1, 3-15, or 18-21, wherein A* is C3-15 cycloalkyl optionally substituted with 1-3 R a . Embodiment 23. The compound of any one of Embodiments 1, 3-15, or 18-22, wherein A* is adamantyl optionally substituted with 1-3 R a . Embodiment 24. The compound of any one of Embodiments 1-23, wherein m2 is 0. Embodiment 25. The compound of any one of Embodiments 1-24, wherein m3 is 0. Embodiment 26. The compound of any one of Embodiments 1-25, wherein m4 is 0. Embodiment 27. The compound of any one of Embodiments 1-26, wherein m5 is 0. Embodiment 28. The compound of any one of Embodiments 1-23, wherein m2 is 0; m3 is 0; m4 is 0; and m5 is 0. Embodiment 29. The compound of any one of Embodiments 1-28, wherein Ring C is . Embodiment 30. The compound of any one of Embodiments 1-29, wherein Ring C is . Embodiment 31. The compound of any one of Embodiments 1-30, wherein Ring C is . Embodiment 32. The compound of any one of Embodiments 1-30, wherein Ring C is Embodiment 34. The compound of any one of Embodiments 1-28, wherein Ring C is . Embodiment 35. The compound of any one of Embodiments 1-28 or 34, wherein Ring C is he compound of any one of Embodiments 1-28 or 34-35, wherein Ring . Embodiment 37. The compound of any one of Embodiments 1-28 or 34-35, wherein Ring . Embodiment 38. The compound of any one of Embodiments 29-37, wherein c1 is 0. Embodiment 39. The compound of any one of Embodiments 29-33, wherein R aN is C 1-3 alkyl. Embodiment 40. The compound of any one of Embodiments 29-33 or 39, wherein R aN is methyl. Embodiment 41. The compound of any one of Embodiments 29-40, wherein L C is a bond or N(R d ). Embodiment 42. The compound of any one of Embodiments 29-41, wherein L C is a bond. Embodiment 43. The compound of any one of Embodiments 29-41, wherein L C is -NH-. Embodiment 44. The compound of any one of Embodiments 29-43, wherein X is CH. Embodiment 45. The compound of any one of Embodiments 1-28, wherein the Embodiment 46. The compound of any one of Embodiments 1-28, wherein the Embodiment 47. The compound of any one of Embodiments 1-28, wherein the Embodiment 49. The compound of any one of Embodiments 1-48, wherein L and L A and n1 are defined according to (AA). Embodiment 50. The compound of any one of Embodiments 1-49, wherein n1 is an integer from 3 to 5. Embodiment 51. The compound of any one of Embodiments 1-49, wherein n1 is an integer from 5 to 9. Embodiment 52. The compound of any one of Embodiments 1-49 or 51, wherein n1 is 6, 7, or 8. Embodiment 53. The compound of any one of Embodiments 1-49, wherein n1 is an integer from 9-12. Embodiment 54. The compound of any one of Embodiments 1-53, wherein 1-2 occurrences of L A is L A4 . Embodiment 55. The compound of any one of Embodiments 1-54, wherein one occurrence of L A is L A4 . Embodiment 56. The compound of any one of Embodiments 1-54, wherein two occurrences of L A are L A4 . Embodiment 57. The compound of any one of Embodiments 1-56, wherein each L A4 is independently selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . Embodiment 58. The compound of any one of Embodiments 1-57, wherein 1-4 occurrences of L A is L A3 . Embodiment 59. The compound of any one of Embodiments 1-58, wherein 1-3 occurrences of L A is L A3 . Embodiment 60. The compound of any one of Embodiments 1-59, wherein 0-1 occurrence of L A3 is C(=O); and each remaining occurrence of L A3 is independently selected from the group consisting of: -O-; -N(H)-; and -N(C1-3 alkyl)-. Embodiment 61. The compound of any one of Embodiments 1-60, wherein 2-7 occurrences of L A is L A1 . Embodiment 62. The compound of any one of Embodiments 1-61, wherein 2-5 occurrences of L A is L A1 . Embodiment 63. The compound of any one of Embodiments 1-62, wherein 0-2 occurrences of L A1 is -CHR L - or -C(R L ) 2 -; and each remaining occurrence of L A1 is -CH 2 -. Embodiment 64. The compound of any one of Embodiments 1-63, wherein each occurrence of L A1 is -CH2-. Embodiment 65. The compound of any one of Embodiments 1-63, wherein one occurrence of L A1 is -CHR L - or -C(R L )2-; and each remaining occurrence of L A1 is -CH2-. Embodiment 66. The compound of any one of Embodiments 1-65, wherein each R L is independently selected from the group consisting of: -F and -C1-3 alkyl optionally substituted with 1-3 F. Embodiment 67. The compound of any one of Embodiments 1-49, wherein L is –(L A )n1-, and L A and n1 are defined according to (AA): n1 is an integer from 5 to 9; 1-2 occurrences of L A is L A4 ; 2-7 occurrences of L A is L A1 ; and 1-3 occurrences of L A is L A3 . Embodiment 68. The compound of any one of Embodiments 1-49, wherein L is selected from the group consisting of: (ii) –(L A3 ) 0-2 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 -L A4 - bb ; (iii) –(L A3 )0-2-(L A1 )0-5-L A4 -(L A1 )0-5-L A3 -L A4 -bb; and (iv) –(L A3 )0-1-(L A1 )2-3-L A3 -L A4 -(L A1 )0-5-(L A3 )1-2-bb; provided that L contains 2-7 L A1 ; and wherein bb represents the point of attachment to Ring C. Embodiment 69. The compound of any one of Embodiments 1-49, wherein L is: –(L A3 ) 0-2 -(L A1 ) 2-9 -(L A3 ) 0-1 -L A4 - bb , wherein bb represents the point of attachment to Ring C. Embodiment 70. The compound of any one of Embodiments 67-69, wherein each L A4 is independently selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . Embodiment 71. The compound of any one of Embodiments 67-70, wherein 0-1 occurrence of L A1 is -CHR L - or -C(R L )2-; and each remaining occurrence of L A1 is -CH2-. Embodiment 72. The compound of any one of Embodiments 67-71, wherein 0-1 occurrence of L A3 is C(=O); and each remaining occurrence of L A3 is independently selected from the group consisting of: -O-; -N(H)-; and -N(C1-3 alkyl)-. Embodiment 73. The compound of any one of Embodiments 1-49 or 68, wherein L is: provided that L contains 2-7 L A1 ; and wherein bb represents the point of attachment to Ring C. Embodiment 74. The compound of any one of Embodiments 1-49, 68, or 73, wherein L is: –(L A3 ) 0-1 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 -(L A3 ) 2 - bb ; provided that L contains 2-5 L A1 ; and wherein bb represents the point of attachment to Ring C. Embodiment 75. The compound of any one of Embodiments 1-49, 68, or 73-74, wherein L is: –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-C(=O)-N(R d )-bb; provided that L contains 2-5 L A1 ; and wherein bb represents the point of attachment to Ring C. Embodiment 76. The compound of any one of Embodiments 73-75, wherein L is a divalent group of Formula (L-1): Formula (L-1) wherein: a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and bb represents the point of attachment to Ring C. Embodiment 77. The compound of Embodiment 76, wherein a3 is 1. Embodiment 78. The compound of Embodiments 76 or 77, wherein L A3 is -O-. Embodiment 79. The compound of Embodiment 76, wherein a3 is 0. Embodiment 80. The compound of any one of Embodiments 76-79, wherein a1a + a1b is 3 or 4. Embodiment 81. The compound of any one of Embodiments 76-79, wherein a1a + a1b is 2 or 5. Embodiment 82. The compound of any one of Embodiments 1-49, 68, or 73, wherein L is: –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-L A3 -bb; provided that L contains 2-7 L A1 ; and wherein bb represents the point of attachment to Ring C. Embodiment 83. The compound of Embodiments 73 or 82, wherein L is a divalent group of Formula (L-2): Formula (L-2) wherein: a3a is 0 or 1; L A3a and L A3b are independently selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 7; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L ) 2 -; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and bb represents the point of attachment to Ring C. Embodiment 84. The compound of Embodiment 83, wherein a3a is 1. Embodiment 85. The compound of Embodiments 83 or 84, wherein L A3a is -O-. Embodiment 86. The compound of any one of Embodiments 83-85, wherein L A3b is -N(H)- or –N(C1-3 alkyl)-. Embodiment 87. The compound of any one of Embodiments 83-85, wherein L A3b is -O-. Embodiment 88. The compound of any one of Embodiments 83-87, wherein a1a + a1b is 2, 3, or 4. Embodiment 89. The compound of any one of Embodiments 83-87, wherein a1a + a1b is 5 or 6. Embodiment 90. The compound of any one of Embodiments 1-49 or 68, wherein L is: –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-L A4 -bb; or –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-L A3 -L A4 -bb; provided that L contains 2-7 L A1 ; and wherein bb represents the point of attachment to Ring C. Embodiment 91. The compound of Embodiment 90, wherein L is: –(L A3 ) 0-1 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 -(piperazinylene)- bb ; or –(L A3 ) 0-1 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 -C(=O)-(piperazinylene)- bb ; provided that L contains 2-5 L A1 ; and wherein bb represents the point of attachment to Ring C. Embodiment 92. The compound of Embodiments 90 or 91, wherein L is a divalent group of Formula (L-3) or (L-4):

Formula (L-4) wherein: a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and bb represents the point of attachment to Ring C. Embodiment 93. The compound of Embodiment 92, wherein a3 is 1. Embodiment 94. The compound of Embodiments 92 or 93, wherein L A3 is -O-. Embodiment 95. The compound of any one of Embodiments 92-94, wherein a1a + a1b is 3 or 4. Embodiment 96. The compound of any one of Embodiments 92-94, wherein a1a + a1b is 2. Embodiment 97. The compound of any one of Embodiments 76-81, 83-89, or 92-96, wherein each occurrence of L A1a and L A1b is -CH2-. Embodiment 98. The compound of any one of Embodiments 76-81, 83-89, or 92-96, wherein one occurrence of L A1a is -CHR L - or -C(R L )2-; each remaining occurrence of L A1a is -CH2-; and each occurrence of L A1b is -CH 2 -. Embodiment 99. The compound of any one of Embodiments 76-81, 83-89, or 92-96, wherein one occurrence of L A1b is -CHR L - or -C(R L )2-; each remaining occurrence of L A1b is -CH2-; and each occurrence of L A1a is -CH 2 -. Embodiment 100. The compound of any one of Embodiments 1-49 or 68, wherein L is: –(L A3 ) 0-1 -(L A1 ) 2-3 -L A3 -L A4 -(L A1 ) 0-5 -(L A3 ) 1-2 - bb ; provided that L contains 2-7 L A1 ; and wherein bb represents the point of attachment to Ring C. Embodiment 101. The compound of any one of Embodiments 1-49, 68, or 100, wherein L is: –(L A3 )0-1-CH2CH2-O-L A4 -(L A1 )0-5-(L A3 )1-2-bb; wherein bb represents the point of attachment to Ring C. Embodiment 102. The compound of Embodiments 100 or 101, wherein L is a divalent group of Formula (L-5): Formula (L-5) wherein: a1 is an integer from 0 to 3; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and bb represents the point of attachment to Ring C. Embodiment 103. The compound of Embodiment 102, wherein a1 is 1. Embodiment 104. The compound of Embodiments 102 or 103, wherein 0-1 occurrence of L A1 is -CHR L - or -C(R L )2-; and each remaining occurrence of L A1 is -CH2-. Embodiment 105. The compound of any one of Embodiments 73-104, wherein L A4 is 4-10 membered heterocyclylene optionally substituted with 1-3 R a . Embodiment 106. The compound of any one of Embodiments 73-105, wherein L A4 is selected from the group consisting of: , , , , each optionally substituted with 1-3 R a at one or more ring carbon atoms, wherein cc represents the point of attachment to L A1b or L A1 . Embodiment 107. The compound of Embodiments 105 or 106, wherein each R a present on L A4 is independently F or C 1-3 alkyl optionally substituted with 1-3 F. Embodiment 108. The compound of any one of Embodiments 73-104, wherein L A4 is C3-10 cycloalkylene optionally substituted with 1-3 R a . Embodiment 109. The compound of any one of Embodiments 73-104 or 108, wherein L A4 is 1,4-cyclohexylene optionally substituted with 1-3 R a . Embodiment 110. The compound of any one of Embodiments 73-104, wherein L A4 is phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . Embodiment 111. The compound of any one of Embodiments 73-104 or 110, wherein L A4 is: (a) phenylene optionally substituted with 1-3 R a ; (b) 1,2-phenylene optionally substituted with 1-3 R a ; or (c) 1,4-phenylene optionally substituted with 1-3 R a . Embodiment 112. The compound of any one of Embodiments 1-49 or 69, wherein L is: –(L A3 ) 0-1 -(L A1 ) 2-9 -(L A3 ) 0-1 -(piperazinylene)- bb , wherein bb represents the point of attachment to Ring C. Embodiment 113. The compound of Embodiment 112, wherein L is a divalent group of Formula (L-6): Formula (L-6) wherein: L A3a is selected from the group consisting of: -O-, -N(H)-, and -N(C1-3 alkyl)-; a3a is 0 or 1; a1 is an integer from 2 to 11; a3c is 0 or 1; and bb represents the point of attachment to Ring C. Embodiment 114. The compound of Embodiment 113, wherein a3a is 1. Embodiment 115. The compound of Embodiments 113 or 114, wherein L A3a is -O-. Embodiment 116. The compound of any one of Embodiments 112-115, wherein 0-1 occurrence of L A1 is -CHR L - or -C(R L )2-; and each remaining occurrence of L A1 is -CH2-. Embodiment 117. The compound of any one of Embodiments 1-49 or 73-76, wherein L is selected from the group consisting of:

wherein bb represents the point of attachment to Ring C. Embodiment 118. The compound of any one of Embodiments 1-49 or 73-76, wherein L is selected from the group consisting of: wherein bb represents the point of attachment to Ring C. Embodiment 119. The compound of any one of Embodiments 1-49, 73, or 82-83, wherein , wherein bb represents the point of attachment to Ring C. Embodiment 120. The compound of any one of Embodiments 1-49 or 90-92, wherein L is the point of attachment to Ring C. Embodiment 121. The compound of any one of Embodiments 1-49 or 100-102, wherein L , wherein bb represents the point of attachment to Ring C. Embodiment 122. The compound of any one of Embodiments 1-48, wherein L and L A and n1 are defined according to (BB). Embodiment 123. The compound of Embodiment 122, wherein n1 is an integer from 3 to 5. Embodiment 124. The compound of Embodiment 122, wherein n1 is an integer from 5 to 9. Embodiment 125. The compound of Embodiment 122, wherein n1 is an integer from 9 to 15. Embodiment 126. The compound of any one of Embodiments 122-125, wherein 1-4 occurrences of L A is L A3 . Embodiment 127. The compound of any one of Embodiments 122-126, wherein 1-3 occurrences of L A is L A3 . Embodiment 128. The compound of any one of Embodiments 122-127, wherein 2-3 occurrences of L A are L A3 . Embodiment 129. The compound of any one of Embodiments 122-128, wherein 0-1 occurrence of L A3 is C(=O); and each remaining occurrence of L A3 is independently selected from the group consisting of: -O-; -N(H)-; and -N(C 1-3 alkyl)-. Embodiment 130. The compound of any one of Embodiments 122-129, wherein 0-2 occurrences of L A1 is -CHR L - or -C(R L )2-; and each remaining occurrence of L A1 is -CH2-. Embodiment 131. The compound of any one of Embodiments 1, 3-48, or 122, wherein L A and n1 are defined according to (BB); and n1 is an integer from 3 to 5. Embodiment 132. The compound of any one of Embodiments 1-48 or 122, wherein L A and n1 are defined according to (BB); and n1 is an integer from 9 to 15. Embodiment 133. The compound of any one of Embodiments 1-48 or 122, wherein L is: –(L A3 )0-2-(L A1 )1-15-(L A3 )0-2- bb, wherein bb represents the point of attachment to Ring C. Embodiment 134. The compound of any one of Embodiments 131-133, wherein L contains 2 or 3 L A3 . Embodiment 135. The compound of any one of Embodiments 131-134, wherein 0-1 occurrence of L A3 is C(=O); and each remaining occurrence of L A3 is independently selected from the group consisting of: -O-; -N(H)-; and -N(C1-3 alkyl)-. Embodiment 136. The compound of any one of Embodiments 131-135, wherein 0-2 occurrences of L A1 is -CHR L - or -C(R L )2-; and each remaining occurrence of L A1 is -CH2-. Embodiment 137. The compound of any one of Embodiments 131-136, wherein each occurrence of L A1 is -CH 2 -. Embodiment 138. The compound of any one of Embodiments 131-136, wherein one occurrence of L A1 is -CHR L - or -C(R L ) 2 -; and each remaining occurrence of L A1 is -CH 2 -. Embodiment 139. The compound of any one of Embodiments 122-138, wherein each R L is independently selected from the group consisting of: -F and -C1-3 alkyl optionally substituted with 1-3 F. Embodiment 140. The compound of any one of Embodiments 1-48, 122, or 133, wherein L –(L A3 ) 0-1 -(L A1 ) 1-15 -(L A3 ) 1-2 - bb , wherein bb represents the point of attachment to Ring C. Embodiment 141. The compound of Embodiment 140, wherein L is a divalent group of Formula (L-7): Formula (L-7) wherein: L A3a is selected from the group consisting of: -O-, -N(H)-, and -N(C1-3 alkyl)-; a3a is 0 or 1; a1 is an integer from 1 to 15; each L A3b is independently selected from the group consisting of: C(=O), -O-, N(H)-, and -N(C 1-3 alkyl)-; a3b is 1 or 2; and bb represents the point of attachment to Ring C. Embodiment 142. The compound of Embodiment 141 wherein –(L A3b )a3b- is . Embodiment 143. The compound of Embodiment 141, wherein –(L A3b )a3b- is -N(H)- or - N(C 1-3 alkyl)-. Embodiment 144. The compound of any one of Embodiments 141-143, wherein 0-2 occurrences of L A1 is -CHR L - or -C(R L ) 2 -; and each remaining occurrence of L A1 is -CH 2 -. Embodiment 145. The compound of any one of Embodiments 141-144, wherein each occurrence of L A1 is -CH2-. Embodiment 146. The compound of Embodiment 76, wherein the compound is a compound of Formula (I-A): Formula (I-A) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: c) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and d) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . Embodiment 147. The compound of Embodiment 76, wherein the compound is a compound of Formula (I-B):

Formula (I-B) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . Embodiment 148. The compound of Embodiments 146 or 147, wherein a3 is 1. Embodiment 149. The compound of any one of Embodiments 146-148, wherein L A3 is -O-. Embodiment 150. The compound of Embodiments 146 or 147, wherein a3 is 0. Embodiment 151. The compound of Embodiment 83, wherein the compound is a compound of Formula (I-C):

Formula (I-C) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3a is 0 or 1; L A3a and L A3b are independently selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 7; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L ) 2 -; and L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . Embodiment 152. The compound of Embodiment 83, wherein the compound is a compound of Formula (I-D): Formula (I-D) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3a is 0 or 1; L A3a and L A3b are independently selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 7; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . Embodiment 153. The compound Embodiments 151 or 152, wherein L A3b is -N(H)- or - N(C1-3 alkyl)-. Embodiment 154. The compound of any one of Embodiments 151-153, wherein a3a is 1. Embodiment 155. The compound of any one of Embodiments 151-154, wherein L A3a is -O- . Embodiment 156. The compound of any one of Embodiments 151-153, wherein a3a is 0. Embodiment 157. The compound of any one of Embodiments 146-156, wherein a1a + a1b is 3. Embodiment 158. The compound of any one of Embodiments 146-156, wherein a1a + a1b is 4. Embodiment 159. The compound of any one of Embodiments 146-156, wherein a1a + a1b is 2 or 5. Embodiment 160. The compound of any one of Embodiments 146-159, wherein each occurrence of L A1a and L A1b is -CH 2 -. Embodiment 161. The compound of any one of Embodiments 146-159, wherein one occurrence of L A1a is -CHR L - or -C(R L )2-; each remaining occurrence of L A1a is -CH2-; and each occurrence of L A1b is -CH 2 -. Embodiment 162. The compound of any one of Embodiments 146-159, wherein one occurrence of L A1b is -CHR L - or -C(R L )2-; each remaining occurrence of L A1b is -CH2-; and each occurrence of L A1a is -CH 2 -. Embodiment 163. The compound of any one of Embodiments 146-162, wherein L A4 is 4-10 membered heterocyclylene optionally substituted with 1-3 R a . Embodiment 164. The compound of any one of Embodiments 146-163, wherein L A4 is selected from the group consisting of: , , , , each optionally substituted with 1-3 R a at one or more ring carbon atoms, wherein cc represents the point of attachment to L A1b . Embodiment 165. The compound of Embodiments 163 or 164, wherein each R a present on L A4 is independently a C1-3 alkyl optionally substituted with 1-3 F. Embodiment 166. The compound of any one of Embodiments 146-162, wherein L A4 is C 3- 10 cycloalkylene optionally substituted with 1-3 R a . Embodiment 167. The compound of any one of Embodiments 146-162 or 166, wherein L A4 is 1,4-cyclohexylene optionally substituted with 1-3 R a . Embodiment 168. The compound of any one of Embodiments 146-162, wherein L A4 is phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . Embodiment 169. The compound of any one of Embodiments 146-162 or 168, wherein L A4 is 1,4-phenylene optionally substituted with 1-3 R a . Embodiment 170. The compound of any one of Embodiments 146-162 or 168, wherein L A4 is 1,2-phenylene or 1,3-phenylene, each of which is optionally substituted with 1-3 R a . Embodiment 171. The compound of any one of Embodiments 146-170, wherein m6 is 0. Embodiment 172. The compound of any one of Embodiments 146-171, wherein the R a present on Ring A is C1-3 alkyl optionally substituted with 1-3 -F. Embodiment 173. The compound of any one of Embodiments 146-172, wherein R 1 is C(O)OH. Embodiment 174. The compound of Embodiment 141, wherein a3a is 0; and the compound is a compound of Formula (II-A): Formula (II-A) or a pharmaceutically acceptable salt thereof, wherein: Ring A is phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; L T1 is a bond or CH 2 ; A * is H or C 3-15 cycloalkyl optionally substituted with 1-3 R a ; L A3b is selected from the group consisting of: C(=O), -O-, N(H)-, and -N(C1-3 alkyl)-; a3b is 1 or 2; and a1 is an integer from 1 to 15. Embodiment 175. The compound of Embodiment 174, wherein Ring A is 5-membered heteroarylene optionally substituted with 1-2 R a . Embodiment 176. The compound of Embodiments 174 or 175, wherein Ring A is selected from the group consisting of: , wherein aa represents the point of attachment to L T1 . Embodiment 177. The compound of any one of Embodiments 174-176, wherein L T1 is CH2. Embodiment 178. The compound of any one of Embodiments 174-177, wherein A* is H. Embodiment 179. The compound of any one of Embodiments 174-177, wherein A* is C3-15 cycloalkyl optionally substituted with 1-3 R a . Embodiment 180. The compound of any one of Embodiments 174-177 or 179, wherein A* is adamantyl optionally substituted with 1-3 R a . Embodiment 181. The compound of any one of Embodiments 174-180, wherein a1 is an integer from 1 to 3. Embodiment 182. The compound of any one of Embodiments 174-180, wherein a1 is an integer from 4 to 6. Embodiment 183. The compound of any one of Embodiments 174-180, wherein a1 is an integer from 7 to 9. Embodiment 184. The compound of any one of Embodiments 174-180, wherein a1 is an integer from 10 to 15. Embodiment 185. The compound of any one of Embodiments 174-184, wherein –(L A3b )a3b- is , wherein bb represents the point of attachment to Ring C. Embodiment 186. The compound of any one of Embodiments 146-185, wherein m2 is 0. Embodiment 187. The compound of any one of Embodiments 146-186, wherein m4 is 0. Embodiment 188. The compound of any one of Embodiments 146-187, wherein Ring C is . Embodiment 189. The compound of any one of Embodiments 146-188, wherein Ring C is . Embodiment 190. The compound of any one of Embodiments 146-188, wherein Ring C is . Embodiment 191. The compound of any one of Embodiments 146-190, wherein R aN is C1-3 alkyl. Embodiment 192. The compound of any one of Embodiments 146-191, wherein R aN is methyl. Embodiment 193. The compound of any one of Embodiments 146-187, wherein Ring C is . Embodiment 194. The compound of any one of Embodiments 146-187, wherein Ring C is . Embodiment 195. The compound of any one of Embodiments 146-194, wherein c1 is 0. Embodiment 196. The compound of any one of Embodiments 146-195, wherein L C is a bond. Embodiment 197. The compound of any one of Embodiments 146-195, wherein L C is -NH- . Embodiment 198. The compound of any one of Embodiments 146-197, wherein X is CH. Embodiment 199. The compound of any one of Embodiments 146-187, wherein the Embodiment 200. The compound of any one of Embodiments 146-187, wherein the Embodiment 201. The compound of any one of Embodiments 146-187, wherein the Embodiment 202. The compound of any one of Embodiments 146-187, wherein the Embodiment 203. The compound of any one of Embodiments 1-202, wherein the compound is selected from the group consisting of Compound No.101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 116a, 117, 117a, 118, 119, 120, 121, 122, 123, 124, 124a, 125, 125a, 126, 127, 128, 129, 130, 131, 131a, 132, 133, 134, 135, 136, 137, 138, 138a, 139, 140, 141, 142, 143, 144, 145, 146, 146a, 147, 147a, 148, 148a, 149, 150, 151, 152, 153, 154, 154a, 155, and 156 as depicted in Table C1, or a pharmaceutically acceptable salt thereof. Embodiment 204. A pharmaceutical composition comprising a compound of any one of Embodiments 1-203, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. Embodiment 205. A Bcl-xL protein non-covalently bound with a compound of any one of Embodiments 1-203, or a pharmaceutically acceptable salt thereof. Embodiment 206. A ternary complex comprising a Bcl-xL protein, a compound of in any one of Embodiments 1-203, or a pharmaceutically acceptable salt thereof, and a CRBN protein. Embodiment 207. A method for treating a cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of any one of Embodiments 1-203, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to Embodiment 204. Embodiment 208. A method for inducing degradation of a Bcl-xL protein in a mammalian cell, the method comprising contacting the mammalian cell with an effective amount of a compound of any one of Embodiments 1-203, or a pharmaceutically acceptable salt thereof. Embodiment 209. The method of Embodiment 208, wherein the contacting occurs in vivo. Embodiment 210. The method of Embodiment 208, wherein the contacting occurs in vitro. Embodiment 211. The method of any one of Embodiments 208-210, wherein the mammalian cell is a mammalian cancer cell. P06 Embodiments Embodiment 1. A compound of Formula (I):

Formula (I) or a pharmaceutically acceptable salt thereof, wherein: Ring A is selected from the group consisting of: (c) C 3-15 cycloalkylene or 3-15 membered heterocyclylene, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; and (d) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 substituents independently selected from the group consisting of: R a and R b ; R 1 is selected from the group consisting of: (d) C(O)OH, (e) C(O)NR d R e , and (f) C(O)OC 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R c ; each R 2 , R 3 , R 4 , and R 5 is independently selected from the group consisting of: halo, CN, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, OH, and NR d R e ; m2 is 0, 1, or 2; m3 and m4 are independently 0, 1, 2, or 3; m5 is 0, 1, 2, 3, or 4; L is –(L A )n1–, wherein L A and n1 are defined according to (AA) or (BB): n1 is an integer from 3 to 15; and each L A is independently selected from the group consisting of: L A1 , L A3 , and L A4 , provided that 1-3 occurrences of L A is L A4 ; n1 is an integer from 0 to 20; and each L A is independently selected from the group consisting of: L A1 and L A3 ; each L A1 is independently selected from the group consisting of: -CH2-, -CHR L -, and - C(R L )2-; each L A3 is independently selected from the group consisting of: -N(R d )-, -N(R b )-, -O- , -S(O) 0-2 -, and C(=O); each L A4 is independently selected from the group consisting of: (c) C 3-15 cycloalkylene or 3-15 membered heterocyclylene, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; and (d) C 6-15 arylene or 5-15 membered heteroarylene, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; provided that L does not contain any N-O, O-O, N-N, N-S(O) 0 , or O-S(O) 0-2 bonds; wherein each R L is independently selected from the group consisting of: halo; cyano; - OH; -C 1-6 alkoxy; -C 1-6 haloalkoxy; -NR d R e ; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); S(O)1-2N(R f )2; -R b ; and C1-6 alkyl optionally substituted with 1-6 R c ; Ring C is selected from the group consisting of: ; c1 is 0, 1, 2, or 3; each R Y is independently selected from the group consisting of: R a and R b ; R aN is H or C1-6 alkyl optionally substituted with 1-3 R c ; Y 1 and Y 2 are independently N, CH, or CR Y ; yy represents the point of attachment to L; X is CH, C, or N; the is a single bond or a double bond; L C is selected from the group consisting of: a bond, -CH 2 -, -CHR a -, -C(R a ) 2 -, -N(R d )-, and O, provided that when X is N, then L C is other than O; each R a is independently selected from the group consisting of: (a) halo; (b) cyano; (c) -OH; (d) oxo; (e) -C1-6 alkoxy; (f) -C1-6 haloalkoxy; (g) -NR d R e ; (h) C(=O)C1-6 alkyl; (i) C(=O)C1-6 haloalkyl; (j) C(=O)OH; (k) C(=O)OC1-6 alkyl; (l) C(=O)OC1-6 haloalkyl; (m) C(=O)N(R f ) 2 ; (n) S(O) 0-2 (C 1-6 alkyl); (o) S(O)0-2(C1-6 haloalkyl); (p) S(O)1-2N(R f )2; and (q) C 1-6 alkyl, C 2-6 alkenyl, or C 2-6 alkynyl, each optionally substituted with 1-6 R c ; each R b is independently selected from the group consisting of: -(L b )b-R b1 and -R b1 , wherein: each b is independently 1, 2, or 3; each -L b is independently selected from the group consisting of: -O-; -N(H)-; -N(C1-3 alkyl)-; -S(O) 0-2 -; C(=O); and C 1-3 alkylene; and each R b1 is independently selected from the group consisting of: C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 R g ; each R c is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C1-6 haloalkoxy; -NR d R e ; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)OH; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); and S(O)1-2N(R f )2; each R d and R e is independently selected from the group consisting of: H; C(=O)C1-6 alkyl; C(=O)C 1-6 haloalkyl; C(=O)OC 1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)N(R f ) 2 ; S(O) 1- 2 (C 1-6 alkyl); S(O) 1-2 (C 1-6 haloalkyl); S(O) 1-2 N(R f ) 2 ; and C 1-6 alkyl optionally substituted with 1-3 R h ; each R f is independently selected from the group consisting of: H and C1-6 alkyl optionally substituted with 1-3 R h ; each R g is independently selected from the group consisting of: R h ; C1-3 alkyl; and C1- 3 haloalkyl; and each R h is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C 1-6 haloalkoxy; -NH 2 , -N(H)(C 1-3 alkyl); and -N(C 1-3 alkyl) 2 . Embodiment 2. The compound of Embodiment 1, wherein Ring A is a phenylene optionally substituted with 1-3 R a . Embodiment 3. The compound of Embodiment 1 or 2, wherein Ring , wherein aa represents the point of attachment to L. Embodiment 4. The compound of any one of Embodiments 1-3, wherein Ring A is , wherein aa represents the point of attachment to L. Embodiment 5. The compound of any one of Embodiments 1-3, wherein Ring A is , wherein aa represents the point of attachment to L. Embodiment 6. The compound of Embodiment 1, wherein Ring A is 5-6 membered heteroarylene optionally substituted with 1-3 R a . Embodiment 7. The compound of any one of Embodiments 1-6, wherein one R a present on Ring A is C 1-3 alkyl optionally substituted with 1-3 F. Embodiment 8. The compound of any one of Embodiments 1-7, wherein one R a present on Ring A is methyl or CF 3 . Embodiment 9. The compound of any one of Embodiments 1-8, wherein R 1 is C(O)OH. Embodiment 10. The compound of any one of Embodiments 1-9, wherein m2 is 0. Embodiment 11. The compound of any one of Embodiments 1-10, wherein m3 is 0. Embodiment 12. The compound of any one of Embodiments 1-11, wherein m4 is 0. Embodiment 13. The compound of any one of Embodiments 1-12, wherein m5 is 0. Embodiment 14. The compound of any one of Embodiments 1-9, wherein m2 is 0; m3 is 0; m4 is 0; and m5 is 0. Embodiment 15. The compound of any one of Embodiments 1-14, wherein Ring C is . Embodiment 16. The compound of any one of Embodiments 1-15, wherein Ring C is . Embodiment 17. The compound of any one of Embodiments 1-15, wherein Ring C is . Embodiment 18. The compound of any one of Embodiments 15-17, wherein c1 is 0. Embodiment 19. The compound of any one of Embodiments 15-18, wherein R aN is C1-3 alkyl. Embodiment 20. The compound of any one of Embodiments 15-19, wherein R aN is methyl. Embodiment 21. The compound of any one of Embodiments 15-20, wherein L C is a bond. Embodiment 22. The compound of any one of Embodiments 15-21, wherein X is CH. Embodiment 23. The compound of any one of Embodiments 1-15, wherein the Embodiment 24. The compound of any one of Embodiments 1-15, wherein the Embodiment 25. The compound of any one of Embodiments 1-24, wherein L is –(L A )n1-; and L A and n1 are defined according to (AA). Embodiment 26. The compound of any one of Embodiments 1-25, wherein n1 is an integer from 3 to 5. Embodiment 27. The compound of any one of Embodiments 1-25, wherein n1 is an integer from 5 to 9. Embodiment 28. The compound of any one of Embodiments 1-25 or 27, wherein n1 is 6, 7, or 8. Embodiment 29. The compound of any one of Embodiments 1-25, wherein n1 is an integer from 9 to 12. Embodiment 30. The compound of any one of Embodiments 1-29, wherein 1-2 occurrences of L A is L A4 . Embodiment 31. The compound of any one of Embodiments 1-29, wherein one occurrence of L A is L A4 . Embodiment 32. The compound of any one of Embodiments 1-29, wherein two occurrences of L A are L A4 . Embodiment 33. The compound of any one of Embodiments 1-32, wherein each L A4 is independently selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . Embodiment 34. The compound of any one of Embodiments 1-33, wherein 1-4 occurrences of L A is L A3 . Embodiment 35. The compound of any one of Embodiments 1-34, wherein 1-3 occurrences of L A is L A3 . Embodiment 36. The compound of any one of Embodiments 1-35, wherein 0-1 occurrence of L A3 is C(=O); and each remaining occurrence of L A3 is independently selected from the group consisting of: -O-; -N(H)-; and -N(C1-3 alkyl)-. Embodiment 37. The compound of any one of Embodiments 1-36, wherein 2-7 occurrences of L A is L A1 . Embodiment 38. The compound of any one of Embodiments 1-37, wherein 2-5 occurrences of L A is L A1 . Embodiment 39. The compound of any one of Embodiments 1-38, wherein 0-2 occurrences of L A1 is -CHR L - or -C(R L ) 2 -; and each remaining occurrence of L A1 is -CH 2 -. Embodiment 40. The compound of any one of Embodiments 1-39, wherein each occurrence of L A1 is -CH 2 -. Embodiment 41. The compound of any one of Embodiments 1-39, wherein one occurrence of L A1 is -CHR L - or -C(R L ) 2 -; and each remaining occurrence of L A1 is -CH 2 -. Embodiment 42. The compound of any one of Embodiments 1-41, wherein each R L is independently selected from the group consisting of: -F and -C1-3 alkyl optionally substituted with 1-3 F. Embodiment 43. The compound of any one of Embodiments 1-25, wherein L is – , and L A and n1 are defined according to (AA): n1 is an integer from 5 to 9; 1-2 occurrences of L A is L A4 ; 2-7 occurrences of L A are L A1 ; and 1-3 occurrences of L A is L A3 . Embodiment 44. The compound of any one of Embodiments 1-25, wherein L is selected from the group consisting of: (i) –(L A3 ) 0-2 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 -(L A3 ) 1-2 - bb ; (iv) –(L A3 ) 0-1 -(L A1 ) 2-3 -L A3 -L A4 -(L A1 ) 0-5 -(L A3 ) 1-2 - bb ; provided that L contains 1-7 L A1 ; and wherein bb represents the point of attachment to Ring C. Embodiment 45. The compound of any one of Embodiments 1-25, wherein L is: –(L A3 )0-2-(L A1 )2-9-(L A3 )0-1-L A4 -bb, wherein bb represents the point of attachment to Ring C. Embodiment 46. The compound of Embodiments 43-45, wherein each L A4 is independently selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . Embodiment 47. The compound of any one of Embodiments 43-46, wherein 0-1 occurrence of L A1 is -CHR L - or -C(R L )2-; and each remaining occurrence of L A1 is -CH2-. Embodiment 48. The compound of any one of Embodiments 43-47, wherein 0-1 occurrence of L A3 is C(=O); and each remaining occurrence of L A3 is independently selected from the group consisting of: -O-; -N(H)-; and -N(C1-3 alkyl)-. Embodiment 49. The compound of any one of Embodiments 1-25, wherein L is: –(L A3 )0-2-(L A1 )0-5-L A4 -(L A1 )0-5-(L A3 )1-2- bb; provided that L contains 1-7 L A1 ; and wherein bb represents the point of attachment to Ring C. Embodiment 50. The compound of any one of Embodiments 1-25 or 49, wherein L is: –(L A3 ) 0-1 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 -(L A3 ) 2 - bb ; provided that L contains 1-5 L A1 ; and wherein bb represents the point of attachment to Ring C. Embodiment 51. The compound of any one of Embodiments 1-25 or 49-50, wherein L is: –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-C(=O)-N(R d )-bb; provided that L contains 1-5 L A1 ; and wherein bb represents the point of attachment to Ring C. Embodiment 52. The compound of any one of Embodiments 49-51, wherein L is a divalent group of Formula (L-1): Formula (L-1) wherein: a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and bb represents the point of attachment to Ring C. Embodiment 53. The compound of Embodiment 52, wherein a3 is 1. Embodiment 54. The compound of Embodiments 52 or 53, wherein L A3 is -O-. Embodiment 55. The compound of Embodiment 52, wherein a3 is 0. Embodiment 56. The compound of any one of Embodiments 52-55, wherein a1a + a1b is 3 or 4. Embodiment 57. The compound of any one of Embodiments 52-55, wherein a1a + a1b is 2 or 5. Embodiment 58. The compound of any one of Embodiments 1-25 or 49, wherein L is: –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-L A3 -bb; provided that L contains 2-7 L A1 ; and wherein bb represents the point of attachment to Ring C. Embodiment 59. The compound of Embodiments 49 or 58, wherein L is a divalent group of Formula (L-2): Formula (L-2) wherein: a3a is 0 or 1; L A3a and L A3b are independently selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 7; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and bb represents the point of attachment to Ring C. Embodiment 60. The compound of Embodiment 59, wherein a3a is 1. Embodiment 61. The compound of Embodiments 59 or 60, wherein L A3a is -O-. Embodiment 62. The compound of any one of Embodiments 59-61, wherein L A3b is -N(H)- or –N(C1-3 alkyl)-. Embodiment 63. The compound of any one of Embodiments 59-61, wherein L A3b is -O-. Embodiment 64. The compound of any one of Embodiments 59-63, wherein a1a + a1b is 2, 3, or 4. Embodiment 65. The compound of any one of Embodiments 59-63, wherein a1a + a1b is 5 or 6. Embodiment 66. The compound of any one of Embodiments 1-25, wherein L is: –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-L A3 -L A4 -bb; provided that L contains 2-7 L A1 ; and wherein bb represents the point of attachment to Ring C. Embodiment 67. The compound of Embodiment 66, wherein L is: –(L A3 ) 0-1 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 -(piperazinylene)- bb ; –(L A3 ) 0-1 -(L A1 ) 0-5 -L A4 -(L A1 ) 0-5 -(piperidinylene)- bb ; or –(L A3 )0-1-(L A1 )0-5-L A4 -(L A1 )0-5-C(=O)-(piperazinylene)-bb; provided that L contains 2-5 L A1 ; wherein the piperazinylene and piperidinylene are each optionally substituted with 1-3 R a , and wherein bb represents the point of attachment to Ring C. Embodiment 68. The compound of Embodiment 66, wherein L is a divalent group of Formula (L-3), (L-3a), (L-3b),or (L-4): Formula (L-3) Formula (L-3a)

Formula (L-4) wherein: a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; n2 and n3 are independently 0, 1, or 2; m8 is 0, 1, or 2; L A4b is 7-10 membered bicyclic nitrogen-containing heterocyclylene optionally substituted with 1-3 R a ; and bb represents the point of attachment to Ring C. Embodiment 69. The compound of Embodiment 68, wherein a3 is 1. Embodiment 70. The compound of Embodiments 68 or 69, wherein L A3 is -O-. Embodiment 71. The compound of any one of Embodiments 68-70, wherein a1a + a1b is 3 or 4. Embodiment 72. The compound of any one of Embodiments 68-70, wherein a1a + a1b is 2. Embodiment 73. The compound of any one of Embodiments 52-57, 59-65, or 68-72, wherein each occurrence of L A1a and L A1b is -CH 2 -. Embodiment 74. The compound of any one of Embodiments 52-57, 59-65, or 68-72, wherein one occurrence of L A1a is -CHR L - or -C(R L )2-; each remaining occurrence of L A1a is -CH2-; and each occurrence of L A1b is -CH 2 -. Embodiment 75. The compound of any one of Embodiments 52-57, 59-65, or 68-72, wherein one occurrence of L A1b is -CHR L - or -C(R L ) 2 -; each remaining occurrence of L A1b is -CH 2 -; and each occurrence of L A1a is -CH 2 -. Embodiment 76. The compound of any one of Embodiments 49-75, wherein L A4 is 4-10 membered heterocyclylene optionally substituted with 1-3 R a . Embodiment 77. The compound of any one of Embodiments 49-75, wherein L A4 is 4-10 membered nitrogen-containing heterocyclylene optionally substituted with 1-3 R a . Embodiment 78. The compound of any one of Embodiments 49-77, wherein L A4 is selected from the group consisting of: , , , , optionally substituted with 1-3 R a at one or more ring carbon atoms, wherein cc represents the point of attachment to L A1b . Embodiment 79. The compound of Embodiments 77 or 78, wherein each R a present on L A4 is independently F or C1-3 alkyl optionally substituted with 1-3 F. Embodiment 80. The compound of any one of Embodiments 49-76, wherein L A4 is C3-10 cycloalkylene optionally substituted with 1-3 R a . Embodiment 81. The compound of any one of Embodiments 49-76 or 80, wherein L A4 is 1,4-cyclohexylene optionally substituted with 1-3 R a . Embodiment 82. The compound of any one of Embodiments 49-75, wherein L A4 is phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . Embodiment 83. The compound of any one of Embodiments 49-75 or 82, wherein L A4 is: (a) phenylene optionally substituted with 1-3 R a ; (b) 1,2-phenylene optionally substituted with 1-3 R a ; or (c) 1,4-phenylene optionally substituted with 1-3 R a . Embodiment 84. The compound of any one of Embodiments 1-25 or 45, wherein L is: –(L A3 )0-1-(L A1 )2-9-(L A3 )0-1-(piperazinylene)-bb, wherein bb represents the point of attachment to Ring C. Embodiment 85. The compound of Embodiment 84, wherein L is a divalent group of Formula (L-6): Formula (L-6) wherein: L A3a is selected from the group consisting of: -O-, -N(H)-, and -N(C 1-3 alkyl)-; a3a is 0 or 1; a1 is an integer from 2 to 11; a3c is 0 or 1; and bb represents the point of attachment to Ring C. Embodiment 86. The compound of Embodiment 85, wherein a3a is 1. Embodiment 87. The compound of Embodiments 85 or 86, wherein L A3a is -O-. Embodiment 88. The compound of any one of Embodiments 85-87, wherein 0-1 occurrence of L A1 is -CHR L - or -C(R L ) 2 -; and each remaining occurrence of L A1 is -CH 2 -. Embodiment 89. The compound of any one of Embodiments 1-25 or 49-52, wherein L is selected from the group consisting of: wherein bb represents the point of attachment to Ring C. Embodiment 90. The compound of any one of Embodiments 1-25 or 49-52, wherein L is selected from the group consisting of:

wherein bb represents the point of attachment to Ring C. Embodiment 91. The compound of any one of Embodiments 1-25, 49, or 58-59, wherein L , wherein bb represents the point of attachment to Ring C. Embodiment 92. The compound of any one of Embodiments 1-25 or 66-68, wherein L is selected from the group consisting of: wherein bb represents the point of attachment to Ring C. Embodiment 93. The compound of any one of Embodiments 1-25 or 66-68, wherein L is selected from the group consisting of: wherein bb represents the point of attachment to Ring C. Embodiment 94. The compound of Embodiments 1 or 52, wherein the compound of Formula (I) is a compound of Formula (I-A): Formula (I-A) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . Embodiment 95. The compound of Embodiments 1 or 52, wherein the compound of Formula (I) is a compound of Formula (I-B): Formula (I-B) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . Embodiment 96. The compound of Embodiments 94 or 95, wherein a3 is 1. Embodiment 97. The compound of any one of Embodiments 94-96, wherein L A3 is -O-. Embodiment 98. The compound of Embodiments 94 or 95, wherein a3 is 0. Embodiment 99. The compound of Embodiments 1 or 59, wherein the compound of Formula (I) is a compound of Formula (I-C) or (I-D): Formula (I-D) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3a is 0 or 1; L A3a and L A3b are independently selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 7; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; and L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . Embodiment 100. The compound of Embodiment 99, wherein the compound is a compound of Formula (I-C). Embodiment 101. The compound Embodiments 99 or 100, wherein L A3b is -N(H)- or -N(C 1- 3 alkyl)-. Embodiment 102. The compound of any one of Embodiments 99-101, wherein a3a is 1. Embodiment 103. The compound of any one of Embodiments 99-102, wherein L A3a is -O-. Embodiment 104. The compound of any one of Embodiments 99-101, wherein a3a is 0. Embodiment 105 The compound of Embodiments 1 or 68, wherein the compound of Formula (I) is a compound of Formula (I-E): Formula (I-E) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and m8 is 0, 1, or 2. Embodiment 106. The compound of Embodiments 1 or 68, wherein the compound of Formula (I) is a compound of Formula (I-Ea): Formula (I-Ea) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; n2 and n3 are independently 0, 1, or 2; and m8 is 0, 1, or 2. Embodiment 107. The compound of Embodiments 1 or 68, wherein the compound of Formula (I) is a compound of Formula (I-Eb): Formula (I-Eb) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; n2 and n3 are independently 0, 1, or 2; and m8 is 0, 1, or 2. Embodiment 108. The compound of Embodiments 1 or 68, wherein the compound of Formula (I) is a compound of Formula (I-F): Formula (I-F) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L ) 2 -; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and m8 is 0, 1, or 2. Embodiment 109. The compound of Embodiments 1 or 68, wherein the compound of Formula (I) is a compound of Formula (I-G):

Formula (I-G) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and m8 is 0, 1, or 2. Embodiment 110. The compound of Embodiments 1 or 68, wherein the compound of Formula (I) is a compound of Formula (I-H): Formula (I-H) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and m8 is 0, 1, or 2. Embodiment 111. The compound of any one of Embodiments 94-110, wherein a1a + a1b is 3. Embodiment 112. The compound of any one of Embodiments 94-110, wherein a1a + a1b is 4. Embodiment 113. The compound of any one of Embodiments 94-110, wherein a1a + a1b is 2 or 5. Embodiment 114. The compound of any one of Embodiments 94-113, wherein each occurrence of L A1a and L A1b is -CH2-. Embodiment 115. The compound of any one of Embodiments 94-113, wherein one occurrence of L A1a is -CHR L - or -C(R L )2-; each remaining occurrence of L A1a is -CH2-; and each occurrence of L A1b is -CH2-. Embodiment 116. The compound of any one of Embodiments 94-113, wherein one occurrence of L A1b is -CHR L - or -C(R L )2-; each remaining occurrence of L A1b is -CH2-; and each occurrence of L A1a is -CH 2 -. Embodiment 117. The compound of any one of Embodiments 94-116, wherein L A4 is 4-10 membered heterocyclylene optionally substituted with 1-3 R a . Embodiment 118. The compound of any one of Embodiments 94-117, wherein L A4 is selected from the group consisting of: , , , , , , , , , each optionally substituted with 1-3 R a at one or more ring carbon atoms, wherein cc represents the point of attachment to L A1b . Embodiment 119. The compound of any one of Embodiments 94-118, wherein each R a present on L A4 is independently a C1-3 alkyl optionally substituted with 1-3 F. Embodiment 120. The compound of any one of Embodiments 94-116, wherein L A4 is C 3-10 cycloalkylene optionally substituted with 1-3 R a . Embodiment 121. The compound of any one of Embodiments 94-116 or 120, wherein L A4 is 1,4-cyclohexylene optionally substituted with 1-3 R a . Embodiment 122. The compound of any one of Embodiments 94-116, wherein L A4 is phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . Embodiment 123. The compound of any one of Embodiments 94-116 or 122, wherein L A4 is 1,4-phenylene optionally substituted with 1-3 R a . Embodiment 124. The compound of any one of Embodiments 94-116 or 122, wherein L A4 is 1,2-phenylene or 1,3-phenylene, each of which is optionally substituted with 1-3 R a . Embodiment 125. The compound of any one of Embodiments 94-124, wherein m6 is 0. Embodiment 126. The compound of any one of Embodiments 94-125, wherein the R a present on Ring A is C1-3 alkyl optionally substituted with 1-3 -F. Embodiment 127. The compound of any one of Embodiments 94-126, wherein R 1 is C(O)OH. Embodiment 128. The compound of any one of Embodiments 94-127, wherein m2 is 0. Embodiment 129. The compound of any one of Embodiments 94-128, wherein m4 is 0. Embodiment 130. The compound of any one of Embodiments 94-129, wherein Ring C is . Embodiment 131. The compound of any one of Embodiments 94-130, wherein Ring C is . Embodiment 132. The compound of any one of Embodiments 94-130, wherein Ring C is . Embodiment 133. The compound of any one of Embodiments 94-132, wherein R aN is C 1-3 alkyl. Embodiment 134. The compound of any one of Embodiments 94-133, wherein R aN is methyl. Embodiment 135. The compound of any one of Embodiments 94-134, wherein c1 is 0. Embodiment 136. The compound of any one of Embodiments 94-135, wherein L C is a bond. Embodiment 137. The compound of any one of Embodiments 94-136, wherein X is CH. Embodiment 138. The compound of any one of Embodiments 94-129, wherein the Embodiment 140. A compound selected from the group consisting of 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 116a, 117, 117a, 118, 119, 120, 121, 122, 123, 124, 124a, 125, 125a, 126, 127, 128, 129, 130, 131, 131a, 132, 133, 134, 135, 136, 137, 138, 138a, 138b, 139, 140, 141, 142, 143, 144, 145, 146, 146a, 146b, 147, 147a, 147b, 148, 148a, 148b, 149, 150, 151, 152, 153, 154, 154a, 155, 156, 157, 158, 159, 159a, 160, 160a, 160b, 161, 161a, 162, 162a, 162b, 163, 164, 164a, 164b, 165, 166, 167, 167a, 167b, 168, 168a, 169, 169a, 169b, 170, 171, 171a, 172, 173, 173a, 173b, 174, 174a, 174b, 175, 176, 176a, 177, 177a, 178, 179, 179a, 180, 180a, 180b, 181, 182, 182a, 183, 183a, 184, 185, 185a, 186, 186a, 187, 187a, 187b, 188, 189, 190, 191, 192, 193, 193a, 193b, 194, 195, 196, 197, 197a, 197b, 198, 199, 200, 201, 202, 202a, 202b, 203, 204, 204a, 205, 206, 207, 207a, 207b, 208, 209, 209a, 210, 210a, 211, 211a, 212, 212a, 213, 214, 214a, 215, 215a, 216, 216a, 216b, 217, 218, 218a, 219, 219a, 219b, 220, 221, 222, 223, 224, 224a, 224b, 225, 226, 227, 227a, 228, 229, 230, 231, 231a, 231b, 232, 233, 234, 235, 236, 237, 238, 239, 239a, 240, 240a, 240b, 241, 242, 243, 244, 245, 245a, 245b, 246, 246a, 246b, 247, 248, 249, 250, 250a, 251, 251a, 252, 252a, 253, 253a, 254, 254a, 255, 256, 257, 258, 259, 259a, 260, 261, 262, 262a, 262b, 263, 263a, 263b, 264, 265, 266, 266a, 266b, 267, 268, 268a, 268b, 269, 269a, 270, 270a, 271, 271a, 271b, 272, 272a, 272b, 273, 273a, 273b, 274, 274a, 274b, 275, 275a, 276, 276a, 276b, 277, 278, 278a, 278b, 279, 280, 281, 281a, 281b, 282, 282a, 282b, 283, 283a, 283b, 284, 284a, 284b, 285, 286, 287, 288, 289, 289a, 289b, 290, 291, 292, 293, 293a, 294, 295, 296, 296a, 297, 297a, 297b, 298, 298a, 299, 299a, 300, 300a, 300b, 301, 301a, 301b, 302, 303, 304, 305, 306, 307, 307a, 307b, 308, 308a, 308b, 309, 310, 310a, 310b, 311, 311a, 312, 313, 314, 316, 316a, 317, 318, 318a, 319, 319a, 320, 320a, 320b, 321, 321a, 321b, 322, 322a, 323, 323a, 324, 324a, 325, 325a, 326, 326a, 327, 327a, 328, 328a, 329, 329a, 330, 330a, 331, 331a, 331b, 332, 332a, 332b, 333, 333a, 334, 334a, 335, 335a, 336, 336a, 337, 337a, 338, 338a, 339, 339a, 340, 340a, 341, 341a, 342, 342a, 343, 343a, 343b, 343c, 343d, 344, 344a, 344b, 344c, 344d, 345, 345a, 346, 346a, 346b, 347, 347a, 347b, 349, 349a, 349b, 350, 350a, 351, 351a, 351b, 352, 352a, and 352b as depicted in Table C1, or a pharmaceutically acceptable salt thereof, provided that the compound does not exhibit Y min (6 h) ≥ 70% and/or Y min (24 h) ≥ 70% under conditions described in Example B1. Embodiment 141. A pharmaceutical composition comprising a compound of any one of Embodiments 1-140, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. Embodiment 142. A BCL-XL protein non-covalently bound with a compound of any one of Embodiments 1-140, or a pharmaceutically acceptable salt thereof. Embodiment 143. A ternary complex comprising a BCL-XL protein, a compound of any one of Embodiments 1-140, or a pharmaceutically acceptable salt thereof, and a CRBN protein. Embodiment 144. A method for treating a cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of any one of Embodiments 1-140, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to Embodiment 141. Embodiment 145. The method of Embodiment 144, further comprising administering an additional therapy or therapeutic agent. Embodiment 146. The method of Embodiment 145, wherein the additional therapy or therapeutic agent is an ALK inhibitor, a BCL-2 inhibitor, a BCR-Abl inhibitor, a BRaf inhibitor, a CDK2 inhibitor, CDK 4/6 inhibitor, a CDK7 inhibitor, a CDK9 inhibitor, an EGFR inhibitor, an anti-EGFR antibody, an ERK inhibitor, a FGFR1 inhibitor, a FGFR2 inhibitor, a FGFR3 inhibitor, a FGFR4 inhibitor, a HER2 inhibitor, a JAK2 inhibitor, a KRas inhibitor, a MEK inhibitor, a MET inhibitor, a PARP inhibitor, an LSD1 inhibitor, a BET inhibitor, a telomerase inhibitor, a TORC1/2 inhibitor, chemotherapy, radiotherapy, or a combination thereof. Embodiment 147. The method of Embodiment 146, wherein the additional therapy or therapeutic agent is a JAK2 inhibitor. Embodiment 148. The method of Embodiment 147, wherein the JAK2 inhibitor is fedratinib (e.g., fedratinib dihydrochloride monohydrate), momelotinib (e.g., momelotinib dihydrochloride), pacritinib (e.g., pacritinib citrate), ruxolitinib (e.g., ruxolitinib phosphate), or a combination thereof. Embodiment 149. The method of any one of Embodiments 144-148, wherein the cancer is breast cancer, colorectal cancer, bile duct cancer, colorectal cancer, gastrointestinal stromal tumor, pancreatic cancer, bladder cancer, kidney cancer, cervical cancer, ovarian cancer, uterine cancer, head and neck cancer, hematological cancer, lung cancer, skin cancer, or a combination thereof. Embodiment 150. The method of Embodiment 149, wherein the hematological cancer is acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), follicular lymphoma (FL), small lymphocytic lymphoma (SLL), essential thrombocythemia, polycythemia vera, myelofibrosis, or a combination thereof. Embodiment 151. The method of Embodiment 150, wherein the hematological cancer is essential thrombocythemia, polycythemia vera, myelofibrosis, or a combination thereof. Embodiment 152. The method of Embodiment 151, wherein the myelofibrosis is primary myelofibrosis. Embodiment 153. The method of Embodiment 151, wherein the myelofibrosis is post- essential thrombocythemia myelofibrosis. Embodiment 154. The method of Embodiment 151, wherein the myelofibrosis is post- polycythemia vera myelofibrosis. Embodiment 155. The method of any one of Embodiments 151-154, wherein the hematological cancer has a JAK2 mutation (e.g., JAK2 V617F). Embodiment 156. The method of Embodiment 150, wherein the ALL is T-ALL. Embodiment 157. The method of Embodiment 150, wherein the AML is M6-AML. Embodiment 158. The method of Embodiment 150, wherein the AML is M7-AML. Embodiment 159. A method for inducing degradation of a BCL-XL protein in a mammalian cell, the method comprising contacting the mammalian cell with an effective amount of a compound of any one of Embodiments 1-140, or a pharmaceutically acceptable salt thereof. Embodiment 160. The method of Embodiment 159, wherein the contacting occurs in vivo. Embodiment 161. The method of Embodiment 159, wherein the contacting occurs in vitro. Embodiment 162. The method of any one of Embodiments 159-161, wherein the mammalian cell is a mammalian cancer cell. Exemplary Formula (I-A) Embodiments Embodiment 1. A compound of Formula (I-A): Formula (I-A) or a pharmaceutically acceptable salt thereof, wherein: R 1 is selected from the group consisting of: (a) C(O)OH, (b) C(O)NR d R e , and (c) C(O)OC1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with 1-3 R c ; each R 2 and R 4 is independently selected from the group consisting of: halo, CN, C1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, OH, and NR d R e ; m2 is 0, 1, or 2; m4 is 0, 1, 2, or 3; m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; wherein each R L is independently selected from the group consisting of: halo; cyano; - OH; -C 1-6 alkoxy; -C 1-6 haloalkoxy; -NR d R e ; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); S(O)1-2N(R f )2; -R b ; and C1-6 alkyl optionally substituted with 1-6 R c ; Ring C is selected from the group consisting of: ; c1 is 0, 1, 2, or 3; each R Y is independently selected from the group consisting of: R a and R b ; R aN is H or C 1-6 alkyl optionally substituted with 1-3 R c ; Y 1 and Y 2 are independently N, CH, or CR Y ; yy represents the point of attachment to L; X is CH, C, or N; the is a single bond or a double bond; L C is selected from the group consisting of: a bond, -CH2-, -CHR a -, -C(R a )2-, -N(R d )-, and O, provided that when X is N, then L C is other than O; each R a is independently selected from the group consisting of: (a) halo; (b) cyano; (c) -OH; (d) oxo; (e) -C 1-6 alkoxy; (f) -C 1-6 haloalkoxy; (g) -NR d R e ; (h) C(=O)C1-6 alkyl; (i) C(=O)C1-6 haloalkyl; (j) C(=O)OH; (k) C(=O)OC1-6 alkyl; (l) C(=O)OC 1-6 haloalkyl; (m) C(=O)N(R f ) 2 ; (n) S(O) 0-2 (C 1-6 alkyl); (o) S(O) 0-2 (C 1-6 haloalkyl); (p) S(O)1-2N(R f )2; and (q) C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted with 1-6 R c ; each R b is independently selected from the group consisting of: -(L b )b-R b1 and -R b1 , wherein: each b is independently 1, 2, or 3; each -L b is independently selected from the group consisting of: -O-; -N(H)-; -N(C1-3 alkyl)-; -S(O) 0-2 -; C(=O); and C 1-3 alkylene; and each R b1 is independently selected from the group consisting of: C3-10 cycloalkyl, 4-10 membered heterocyclyl, C6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 R g ; each R c is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C 1-6 haloalkoxy; -NR d R e ; C(=O)C 1-6 alkyl; C(=O)C 1-6 haloalkyl; C(=O)OC 1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)OH; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); and S(O)1-2N(R f )2; each R d and R e is independently selected from the group consisting of: H; C(=O)C1-6 alkyl; C(=O)C 1-6 haloalkyl; C(=O)OC 1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)N(R f ) 2 ; S(O) 1- 2(C1-6 alkyl); S(O)1-2(C1-6 haloalkyl); S(O)1-2N(R f )2; and C1-6 alkyl optionally substituted with 1-3 R h ; each R f is independently selected from the group consisting of: H and C 1-6 alkyl optionally substituted with 1-3 R h ; each R g is independently selected from the group consisting of: R h ; C1-3 alkyl; and C1- 3 haloalkyl; and each R h is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C1-6 haloalkoxy; -NH2, -N(H)(C1-3 alkyl); and -N(C1-3 alkyl)2. Embodiment 2. The compound of Embodiment 1, wherein a1a + a1b is 2 to 5. Embodiment 3. The compound of Embodiment 1 or 2, wherein a1a + a1b is 3. Embodiment 4. The compound of Embodiment 1 or 2, wherein a1a + a1b is 4. Embodiment 5. The compound of Embodiment 1 or 2, wherein a1a + a1b is 2 or 5. Embodiment 6. The compound of any one of Embodiments 1-5, wherein a3 is 1. Embodiment 7. The compound of any one of Embodiments 1-6, wherein L A3 is -O-. Embodiment 8. The compound of any one of Embodiments 1-5, wherein a3 is 0. Embodiment 9. The compound of any one of Embodiments 1-8, wherein each occurrence of L A1a and L A1b is -CH 2 -. Embodiment 10. The compound of any one of Embodiments 1-8, wherein one occurrence of L A1a is -CHR L - or -C(R L ) 2 -; each remaining occurrence of L A1a is -CH 2 -; and each occurrence of L A1b is -CH 2 -. Embodiment 11. The compound of any one of Embodiments 1-8, wherein one occurrence of L A1b is -CHR L - or -C(R L )2-; each remaining occurrence of L A1b is -CH2-; and each occurrence of L A1a is -CH2-. Embodiment 12. The compound of any one of Embodiments 1-11, wherein L A4 is 4-10 membered heterocyclylene optionally substituted with 1-3 R a . Embodiment 13. The compound of any one of Embodiments 1-12, wherein L A4 is selected from the group consisting of: , optionally substituted with 1-3 R a at one or more ring carbon atoms, wherein cc represents the point of attachment to L A1b ; or substituted with 1-3 R a at one or more ring carbon atoms, wherein cc represents the point of attachment to L A1b . Embodiment 14. The compound of Embodiments 12 or 13, wherein each R a present on L A4 is independently a C 1-3 alkyl optionally substituted with 1-3 F. Embodiment 15. The compound of any one of Embodiments 1-11, wherein L A4 is C3-10 cycloalkylene optionally substituted with 1-3 R a . Embodiment 16. The compound of any one of Embodiments 1-11 or 15, wherein L A4 is 1,4- cyclohexylene optionally substituted with 1-3 R a . Embodiment 17. The compound of any one of Embodiments 1-11, wherein L A4 is phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . Embodiment 18. The compound of any one of Embodiments 1-11 or 17, wherein L A4 is 1,4- phenylene optionally substituted with 1-3 R a . Embodiment 19. The compound of any one of Embodiments 1-11 or 17, wherein L A4 is 1,2- phenylene or 1,3-phenylene, each of which is optionally substituted with 1-3 R a . Embodiment 20. The compound of any one of Embodiments 1-19, wherein m6 is 0. Embodiment 21. The compound of any one of Embodiments 1-20, wherein the R a present on Ring A is C1-3 alkyl optionally substituted with 1-3 -F. Embodiment 22. The compound of any one of Embodiments 1-21, wherein R 1 is C(O)OH. Embodiment 23. The compound of any one of Embodiments 1-22, wherein m2 is 0. Embodiment 24. The compound of any one of Embodiments 1-23, wherein m4 is 0. Embodiment 25. The compound of any one of Embodiments 1-24, wherein Ring C is . Embodiment 26. The compound of any one of Embodiments 1-25, wherein Ring C is . Embodiment 27. The compound of any one of Embodiments 1-25, wherein Ring C is . Embodiment 28. The compound of any one of Embodiments 1-27, wherein R aN is C1-3 alkyl. Embodiment 29. The compound of any one of Embodiments 1-28, wherein R aN is methyl. Embodiment 30. The compound of any one of Embodiments 1-29, wherein c1 is 0. Embodiment 31. The compound of any one of Embodiments 1-30, wherein L C is a bond. Embodiment 32. The compound of any one of Embodiments 1-31, wherein X is CH. Embodiment 33. The compound of any one of Embodiments 1-24, wherein the Embodiment 34. The compound of any one of Embodiments 1-24, wherein the Embodiment 35. The compound of Embodiment 1, wherein the compound of Formula (I-A) is a compound of Formula (I-A-1): Formula (I-A-1) or a pharmaceutically acceptable salt thereof, wherein: R 1 is C(O)OH or C(O)OC 1-6 alkyl; R a1 is C1-3 alkyl optionally substituted with 1-3 F; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: c) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and d) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . Embodiment 36. The compound of Embodiment 1, wherein the compound of Formula (I-A) is a compound of Formula (I-A-2): Formula (I-A-2) or a pharmaceutically acceptable salt thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and Ring , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 F; and R aN is C1-3 alkyl. Embodiment 37. The compound of any one of Embodiments 1, 35 or 36, wherein the compound of Formula (I-A), Formula (I-A-1), or Formula (I-A-2) is a compound is a compound of Formula (I-A-3): Formula (I-A-3) or a pharmaceutically acceptable salt thereof, wherein: R 1 is C(O)OH or C(O)OC1-6 alkyl; R a1 is C 1-3 alkyl optionally substituted with 1-3 F; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 1 to 5 (e.g., from 2 to 5); L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and Ring C is or , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 F; and R aN is C 1-3 alkyl. Embodiment 38. The compound of any one of Embodiments 36 or 37, wherein R 1 is C(O)OH. Embodiment 39. The compound of any one of Embodiments 35-38, wherein c1 is 0. Embodiment 40. The compound of any one of Embodiments 35-39 , wherein a1a + a1b is from 2 to 5. Embodiment 41. The compound of any one of Embodiments 1 or 35-40, wherein the moiety is selected from the groups depicted in Table (L-I- A): Table (L-I-A)

wherein bb represents the point of attachment to Ring C; or wherein the moiety is selected from the group consisting of:

wherein bb represents the point of attachment to Ring C. Embodiment 42. The compound of any one of Embodiments 1-41, wherein the compound is selected from the group consisting of 101, 102, 103, 104, 105, 106, 107, 116, 116a, 117, 117a, 120, 121, 122, 124, 124a, 125, 125a, 126, 127, 128, 129, 130, 131, 131a, 132, 134, 156, 158, 159, 159a, 160, 160a, 160b, 162, 162a, 162b, 164, 164a, 164b, 166, 167, 167a, 167b, 169, 169a, 169b, 170, 173, 173a, 173b, 177, 177a, 180, 180a, 180b, 181, 186, 186a, 186b, 187, 187a, 187b, 193, 193a, 193b, 196, 197, 197a, 197b, 202, 202a, 202b, 203, 207, 207a, 207b, 209, 209a, 210, 210a, 210b, 213, 216, 216a, 216b, 218, 218a, 218b, 219, 219a, 219b, 226, 229, 231, 231a, 231b, 240, 240a, 240b, 243, 245, 245a, 245b, 246, 246a, 246b, 262, 262a, 262b, 263, 263a, 263b, 267, 268, 268a, 268b, 272, 272a, 272b, 277, 280, 286, 289, 289a, 289b, 292, 295, 302, 309, 313, 317, 343, 343a, 343b, 343c, 343d, and 357, as depicted in Table C1, or pharmaceutically acceptable salts thereof; or the compound is selected from the group consisting of 101, 102, 103, 104, 105, 106, 107, 116, 116a, 117, 117a, 120, 121, 122, 124, 124a, 125, 125a, 126, 127, 128, 129, 130, 131, 131a, 132, 134, 156, 158, 159, 159a, 160, 160a, 160b, 162, 162a, 162b, 164, 164a, 164b, 166, 167, 167a, 167b, 169, 169a, 169b, 170, 173, 173a, 173b, 177, 177a, 180, 180a, 180b, 181, 186, 186a, 187, 187a, 187b, 193, 193a, 193b, 196, 197, 197a, 197b, 202, 202a, 202b, 203, 207, 207a, 207b, 209, 209a, 210, 210a, 213, 216, 216a, 216b, 218, 218a, 219, 219a, 219b, 226, 229, 231, 231a, 231b, 240, 240a, 240b, 243, 245, 245a, 245b, 246, 246a, 246b, 262, 262a, 262b, 263, 263a, 263b, 267, 268, 268a, 268b, 272, 272a, 272b, 277, 280, 286, 289, 289a, 289b, 292, 295, 302, 309, 313, 317, 343, 343a, 343b, 343c, and 343d as depicted in Table C1, or pharmaceutically acceptable salts thereof; or the compound is selected from the group consisting of 101, 102, 103, 104, 105, 106, 107, 116, 116a, 117, 117a, 120, 121, 122, 124, 124a, 125, 125a, 126, 127, 128, 129, 130, 131, 131a, 132, 134, 156, 158, 159, 159a, 160, 160a, 160b, 162, 162a, 162b, 164, 164a, 164b, 166, 167, 167a, 167b, 169, 169a, 169b, 170, 173, 173a, 173b, 177, 177a, 180, 180a, 180b, 181, 186, 186a, 187, 187a, 187b, 193, 193a, 193b, 196, 197, 197a, 197b, 202, 202a, 202b, 203, 207, 207a, 207b, 209, 209a, 210, 210a, 213, 216, 216a, 216b, 218, 218a, 219, 219a, 219b, 226, 229, 231, 231a, 231b, 240, 240a, 240b, 243, 245, 245a, 245b, 246, 246a, 246b, 262, 262a, 262b, 263, 263a, 263b, 267, 268, 268a, 268b, 272, 272a, 272b, 277, 280, 286, 289, 289a, 289b, 292, 295, 302, 309, 313, and 317, as depicted in Table C1, or pharmaceutically acceptable salts thereof; or the compound is selected from the group consisting of 101, 102, 103, 104, 105, 106, 107, 116, 116a, 117, 117a, 120, 121, 122, 124, 124a, 125, 125a, 126, 127, 128, 129, 130, 131, 131a, 132, 134, 156, 158, 159, 159a, 160, 160a, 160b, 162, 162a, 162b, 164, 164a, 164b, 166, 167, 167a, 167b, 169, 169a, 169b, 170, 173, 173a, 173b, 177, 177a, 180, 180a, 180b, 181, 186, 186a, 187, 187a, 187b, 193, 193a, 193b, 196, 197, 197a, 197b, 202, 202a, 202b, 203, 207, 207a, 207b, 209, 209a, 210, 210a, 213, 216, 216a, 216b, 218, 218a, 219, 219a, 219b, 226, 229, 231, 231a, 231b, 240, 240a, 240b, 243, 245, 245a, 245b, 246, 246a, 246b, 262, 262a, 262b, 263, 263a, 263b, 267, 268, 268a, 268b, 272, 272a, 272b, 277, 280, 286, 289, 289a, and 289b as depicted in Table C1, or a pharmaceutically acceptable salt thereof; or the compound is selected from the group consisting of 101, 102, 103, 104, 105, 106, 107, 116, 116a, 117, 117a, 120, 121, 122, 124, 124a, 125, 125a, 126, 127, 128, 129, 130, 131, 131a, 132, 134, 156, 158, 159, 159a, 160, 160a, 160b, 162, 162a, 162b, 164, 164a, 164b, 166, 167, 167a, 167b, 169, 169a, 169b, 170, 173, 173a, 173b, 177, 177a, 180, 180a, 180b, 181, 186, 186a, 187, 187a, 187b, 193, 193a, 193b, 196, 197, 197a, 197b, 202, 202a, 202b, 203, 207, 207a, 207b, 209, 209a, 210, 210a, 213, 216, 216a, 216b, 218, 218a, 219, 219a, 219b, 226, 229, 231, 231a, 231b, 240, 240a, and 240b as depicted in Table C1, or a pharmaceutically acceptable salt thereof; or the compound is selected from the group consisting of 101, 102, 103, 104, 105, 106, 107, 116, 116a, 117, 117a, 120, 121, 122, 124, 124a, 125, 125a, 126, 127, 128, 129, 130, 131, 131a, 132, 134, and 156 as depicted in Table C1, or a pharmaceutically acceptable salt thereof. Embodiment 43. A pharmaceutical composition comprising the compound of any one of Embodiments 1-42, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. Embodiment 44. A compound of any one of Embodiments 1-42, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of Embodiment 43 for use in treatment. Embodiment 45. A compound of any one of Embodiments 1-42 or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of Embodiment 43 for use in the treatment of a cancer selected from the group consisting of essential thrombocythemia, polycythemia vera, myelofibrosis, and a combination thereof. Embodiment 46. The compound or pharmaceutical composition of Embodiment 45 and an additional therapeutic agent or therapy selected from the group consisting of fedratinib (e.g., fedratinib dihydrochloride monohydrate), momelotinib (e.g., momelotinib dihydrochloride), pacritinib (e.g., pacritinib citrate), ruxolitinib (e.g., ruxolitinib phosphate), and a combination thereof for use in treatment. Embodiment 47. The compound or pharmaceutical composition of Embodiment 46 and an additional therapeutic agent or therapy selected from the group consisting of fedratinib (e.g., fedratinib dihydrochloride monohydrate), momelotinib (e.g., momelotinib dihydrochloride), pacritinib (e.g., pacritinib citrate), ruxolitinib (e.g., ruxolitinib phosphate), and a combination thereof for use in treatment of a cancer selected from the group consisting of essential thrombocythemia, polycythemia vera, myelofibrosis, and a combination thereof. Embodiment 48. A compound of any one of Embodiments 1-42, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of Embodiment 43 for use in a method of treating cancer selected from the group consisting of essential thrombocythemia, polycythemia vera, myelofibrosis, and a combination thereof, the method comprising administering to a subject a therapeutically effective amount of a compound of Embodiments 1-42, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of Embodiment 43. Embodiment 49. The compound or pharmaceutical composition of Embodiment 48, wherein the method comprises administering to the subject a therapeutically effective amount of an additional therapeutic agent or therapy selected from the group consisting of fedratinib (e.g., fedratinib dihydrochloride monohydrate), momelotinib (e.g., momelotinib dihydrochloride), pacritinib (e.g., pacritinib citrate), ruxolitinib (e.g., ruxolitinib phosphate), and a combination thereof. Embodiment 50. The compound or pharmaceutical composition of Embodiment 49, wherein the compound of any one of Embodiments 1-42, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of Embodiment 43, and the additional therapeutic agent or therapy are administered simultaneously, separately, or sequentially. Exemplary Formula (I-E) Embodiments Embodiment 1. A compound of Formula (I-E): Formula (I-E) or a pharmaceutically acceptable salt thereof, wherein: R 1 is selected from the group consisting of: (a) C(O)OH, (b) C(O)NR d R e , and (c) C(O)OC 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R c ; each R 2 and R 4 is independently selected from the group consisting of: halo, CN, C 1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, C1-3 haloalkoxy, OH, and NR d R e ; m2 is 0, 1, or 2; m4 is 0, 1, 2, or 3; m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; wherein each R L is independently selected from the group consisting of: halo; cyano; - OH; -C 1-6 alkoxy; -C 1-6 haloalkoxy; -NR d R e ; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); S(O) 1-2 N(R f ) 2 ; -R b ; and C 1-6 alkyl optionally substituted with 1-6 R c ; m8 is 0, 1, or 2; Ring C is selected from the group consisting of: ; c1 is 0, 1, 2, or 3; each R Y is independently selected from the group consisting of: R a and R b ; R aN is H or C1-6 alkyl optionally substituted with 1-3 R c ; Y 1 and Y 2 are independently N, CH, or CR Y ; yy represents the point of attachment to L; X is CH, C, or N; the is a single bond or a double bond; L C is selected from the group consisting of: a bond, -CH2-, -CHR a -, -C(R a )2-, -N(R d )-, and O, provided that when X is N, then L C is other than O; each R a is independently selected from the group consisting of: (a) halo; (b) cyano; (c) -OH; (d) oxo; (e) -C 1-6 alkoxy; (f) -C 1-6 haloalkoxy; (g) -NR d R e ; (h) C(=O)C1-6 alkyl; (i) C(=O)C 1-6 haloalkyl; (j) C(=O)OH; (k) C(=O)OC1-6 alkyl; (l) C(=O)OC 1-6 haloalkyl; (m) C(=O)N(R f )2; (n) S(O)0-2(C1-6 alkyl); (o) S(O) 0-2 (C 1-6 haloalkyl); (p) S(O) 1-2 N(R f ) 2 ; and (q) C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted with 1-6 R c ; each R b is independently selected from the group consisting of: -(L b )b-R b1 and -R b1 , wherein: each b is independently 1, 2, or 3; each -L b is independently selected from the group consisting of: -O-; -N(H)-; -N(C 1-3 alkyl)-; -S(O) 0-2 -; C(=O); and C 1-3 alkylene; and each R b1 is independently selected from the group consisting of: C3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 R g ; each R c is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C1-6 haloalkoxy; -NR d R e ; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)OH; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); and S(O)1-2N(R f )2; each R d and R e is independently selected from the group consisting of: H; C(=O)C1-6 alkyl; C(=O)C 1-6 haloalkyl; C(=O)OC 1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)N(R f ) 2 ; S(O) 1- 2(C1-6 alkyl); S(O)1-2(C1-6 haloalkyl); S(O)1-2N(R f )2; and C1-6 alkyl optionally substituted with 1-3 R h ; each R f is independently selected from the group consisting of: H and C 1-6 alkyl optionally substituted with 1-3 R h ; each R g is independently selected from the group consisting of: R h ; C1-3 alkyl; and C1- 3 haloalkyl; and each R h is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C1-6 haloalkoxy; -NH2, -N(H)(C1-3 alkyl); and -N(C1-3 alkyl)2. Embodiment 2. The compound of Embodiment 1, wherein m8 is 0. Embodiment 3. The compound of Embodiment 1 or 2, wherein a3 is 1. Embodiment 4. The compound of any one of Embodiments 1-3, wherein L A3 is -O-. Embodiment 5. The compound of any one of Embodiments 1-4, wherein a1a + a1b is 3. Embodiment 6. The compound of any one of Embodiments 1-4, wherein a1a + a1b is 4. Embodiment 7. The compound of any one of any one of Embodiments 1-4, wherein a1a + a1b is 2 or 5. Embodiment 8. The compound of any one of Embodiments 1-7, wherein each occurrence of L A1a and L A1b is -CH2-. Embodiment 9. The compound of any one of Embodiments 1-7, wherein one occurrence of L A1a is -CHR L - or -C(R L )2-; each remaining occurrence of L A1a is -CH2-; and each occurrence of L A1b is -CH 2 -. Embodiment 10. The compound of any one of Embodiments 1-7, wherein one occurrence of L A1b is -CHR L - or -C(R L )2-; each remaining occurrence of L A1b is -CH2-; and each occurrence of L A1a is -CH 2 -. Embodiment 11. The compound of any one of Embodiments 1-10, wherein L A4 is 4-10 membered heterocyclylene optionally substituted with 1-3 R a . Embodiment 12. The compound of any one of Embodiments 1-11, wherein L A4 is selected optionally substituted with 1-3 R a at one or more ring carbon atoms, wherein cc represents the point of attachment to L A1b ; or substituted with 1-3 R a at one or more ring carbon atoms, wherein cc represents the point of attachment to L A1b . Embodiment 13. The compound of Embodiments 11 or 12, wherein each R a present on L A4 is independently a C 1-3 alkyl optionally substituted with 1-3 F. Embodiment 14. The compound of any one of Embodiments 1-10, wherein L A4 is C3-10 cycloalkylene optionally substituted with 1-3 R a . Embodiment 15. The compound of any one of Embodiments 1-10 or 14, wherein L A4 is 1,4- cyclohexylene optionally substituted with 1-3 R a . Embodiment 16. The compound of any one of Embodiments 1-10, wherein L A4 is phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . Embodiment 17. The compound of any one of Embodiments 1-10 or 16, wherein L A4 is 1,4- phenylene optionally substituted with 1-3 R a . Embodiment 18. The compound of any one of Embodiments 1-10 or 16, wherein L A4 is 1,2- phenylene or 1,3-phenylene, each of which is optionally substituted with 1-3 R a . Embodiment 19. The compound of any one of Embodiments 1-18, wherein m6 is 0. Embodiment 20. The compound of any one of Embodiments 1-19, wherein the R a present on Ring A is C1-3 alkyl optionally substituted with 1-3 -F. Embodiment 21. The compound of any one of Embodiments 1-20, wherein R 1 is C(O)OH. Embodiment 22. The compound of any one of Embodiments 1-21, wherein m2 is 0. Embodiment 23. The compound of any one of Embodiments 1-22, wherein m4 is 0. Embodiment 24. The compound of any one of Embodiments 1-23, wherein Ring C is . Embodiment 25. The compound of any one of Embodiments 1-24, wherein Ring C is . Embodiment 26. The compound of any one of Embodiments 1-24, wherein Ring C is . Embodiment 27. The compound of any one of Embodiments 1-26, wherein R aN is C 1-3 alkyl. Embodiment 28. The compound of any one of Embodiments 1-27, wherein R aN is methyl. Embodiment 29. The compound of any one of Embodiments 1-27, wherein c1 is 0. Embodiment 30. The compound of any one of Embodiments 1-29, wherein L C is a bond. Embodiment 31. The compound of any one of Embodiments 1-30, wherein X is CH. Embodiment 32. The compound of any one of Embodiments 1-23, wherein the Embodiment 33. The compound of any one of Embodiments 1-23, wherein the Embodiment 34. The compound of Embodiment 1, wherein the compound is a compound of Formula (I-E-1):

Formula (I-E-1) or pharmaceutically acceptable salts thereof, wherein: R 1 is C(O)OH or C(O)OC1-6 alkyl; R a1 is C1-3 alkyl optionally substituted with 1-3 F; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; and m8 is 0, 1, or 2. Embodiment 35. The compound of Embodiment 1, wherein the compound is a compound of Formula (I-E-2):

Formula (I-E-2) or pharmaceutically acceptable salts thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; m8 is 0, 1, or 2; and Ring , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C1-3 alkyl optionally substituted with 1-3 F; and R aN is C 1-3 alkyl. Embodiment 36. The compound of Embodiments 1 or 34-35, wherein the compound is a compound of Formula (I-E-3):

Formula (I-E-3) or pharmaceutically acceptable salts thereof, wherein: R 1 is C(O)OH or C(O)OC 1-6 alkyl; R a1 is C 1-3 alkyl optionally substituted with 1-3 F; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; m8 is 0, 1, or 2; and Ring , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 F; and R aN is C1-3 alkyl. Embodiment 37. The compound of any one of Embodiments 34-36, wherein m8 is 0. Embodiment 38. The compound of any one of Embodiments 1 or 34-37, wherein the moiety is selected from the groups depicted in Table (L-I-E): Table (L-I-E) wherein bb represents the point of attachment to Ring C; or wherein the moiety is selected from the group , , , , and , wherein bb represents the point of attachment to Ring C. Embodiment 39. The compound of any one of Embodiments 1-36, wherein the compound is selected from the group consisting of 147, 147a, 147b, 148, 148a, 148b, 149, 150, 161, 161a, 163, 168, 168a, 175, 188, 195, 200, 201, 204, 204a, 208, 211, 211a, 212, 212a, 215, 215a, 220, 225, 230, 232, 233, 234, 235, 241, 273, 273a, 273b, 274, 274a, 274b, 275, 275a, 275b, 278, 278a, 278b, 281, 281a, 281b, 282, 282a, 282b, 283, 283a, 283b, 284, 284a, 284b, 293, 293a, 296, 296a, 297, 297a, 297b, 300, 300a, 300b, 301, 301a, 301b, 307, 307a, 307b, 308, 308a, 308b, 310, 310a, 316, 316a, 318, 318a, 319, 319a, 322, 322a, 323, 323a, 324, 324a, 325, 325a, 328, 328a, 333, 333a, 334, 334a, 335, 335a, 336, 336a, 339, 339a, 339b, 340, 340a, 341, 341a, 342, 342a, 345, 345a, 346, 346a, 346b, 347, 347a, 347b, 349, 349a, 349b, 350, 350a, 354, 354a, and 354b as depicted in Table C1, or pharmaceutically acceptable salts thereof; or the compound is selected from the group consisting of 147, 147a, 147b, 148, 148a, 148b, 149, 150, 161, 161a, 163, 168, 168a, 175, 188, 195, 200, 201, 204, 204a, 208, 211, 211a, 212, 212a, 215, 215a, 220, 225, 230, 232, 233, 234, 235, 241, 273, 273a, 273b, 274, 274a, 274b, 275, 275a, 278, 278a, 278b, 281, 281a, 281b, 282, 282a, 282b, 283, 283a, 283b, 284, 284a, 284b, 293, 293a, 296, 296a, 297, 297a, 297b, 300, 300a, 300b, 301, 301a, 301b, 307, 307a, 307b, 308, 308a, 308b, 310, 310a, 316, 316a, 318, 318a, 319, 319a, 322, 322a, 323, 323a, 324, 324a, 325, 325a, 328, 328a, 333, 333a, 334, 334a, 335, 335a, 336, 336a, 339, 339a, 340, 340a, 341, 341a, 342, 342a, 345, 345a, 346, 346a, 346b, 347, 347a, 347b, 349, 349a, 349b, 350, and 350a as depicted in Table C1, or pharmaceutically acceptable salts thereof; or the compound is selected from the group consisting of 147, 147a, 147b, 148, 148a, 148b, 149, 150, 161, 161a, 163, 168, 168a, 175, 188, 195, 200, 201, 204, 204a, 208, 211, 211a, 212, 212a, 215, 215a, 220, 225, 230, 232, 233, 234, 235, 241, 273, 273a, 273b, 274, 274a, 274b, 275, 275a, 278, 278a, 278b, 281, 281a, 281b, 282, 282a, 282b, 283, 283a, 283b, 284, 284a, 284b, 293, 293a, 296, 296a, 297, 297a, 297b, 300, 300a, 300b, 301, 301a, 301b, 307, 307a, 307b, 308, 308a, 308b, 310, 310a, 316, 316a, 318, 318a, 319, 319a, 322, 322a, 323, 323a, 324, 324a, 325, and 325a, as depicted in Table C1, or pharmaceutically acceptable salts thereof; or the compound is selected from the group consisting of 147, 147a, 147b, 148, 148a, 148b, 149, 150, 161, 161a, 163, 168, 168a, 175, 188, 195, 200, 201, 204, 204a, 208, 211, 211a, 212, 212a, 215, 215a, 220, 225, 230, 232, 233, 234, 235, 241, 273, 273a, 273b, 274, 274a, 274b, 275, 275a, 278, 278a, 278b, 281, 281a, 281b, 282, 282a, 282b, 283, 283a, 283b, 284, 284a, and 284b as depicted in Table C1, or pharmaceutically acceptable salts thereof; or the compound is selected from the group consisting of 147, 147a, 148, 148a, 149, 150, 161, 161a, 163, 168, 168a, 175, 188, 195, 200, 201, 204, 204a, 208, 211, 211a, 212, 212a, 215, 215a, 220, 225, 230, 232, 233, 234, 235, and 241, as depicted in Table C1, or pharmaceutically acceptable salts thereof; or the compound is selected from the group consisting of 147, 147a, 148, 148a, 149, and 150 as depicted in Table C1, or pharmaceutically acceptable salts thereof; or Embodiment 40. A pharmaceutical composition comprising the compound of any one of Embodiments 1-39, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. Embodiment 41. A compound of any one of Embodiments 1-39, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of Embodiment 40 for use in treatment. Embodiment 42. A compound of any one of Embodiments 1-39 or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of Embodiment 40 for use in the treatment of a cancer selected from the group consisting of essential thrombocythemia, polycythemia vera, myelofibrosis, and a combination thereof. Embodiment 43. The compound or pharmaceutical composition of Embodiment 42 and an additional therapeutic agent or therapy selected from the group consisting of fedratinib (e.g., fedratinib dihydrochloride monohydrate), momelotinib (e.g., momelotinib dihydrochloride), pacritinib (e.g., pacritinib citrate), ruxolitinib (e.g., ruxolitinib phosphate), and a combination thereof for use in treatment. Embodiment 44. The compound or pharmaceutical composition of Embodiment 43 and an additional therapeutic agent or therapy selected from the group consisting of fedratinib (e.g., fedratinib dihydrochloride monohydrate), momelotinib (e.g., momelotinib dihydrochloride), pacritinib (e.g., pacritinib citrate), ruxolitinib (e.g., ruxolitinib phosphate), and a combination thereof for use in treatment of a cancer selected from the group consisting of essential thrombocythemia, polycythemia vera, myelofibrosis, and a combination thereof. Embodiment 45. A compound of any one of Embodiments 1-39, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of Embodiment 40 for use in a method of treating cancer selected from the group consisting of essential thrombocythemia, polycythemia vera, myelofibrosis, and a combination thereof, the method comprising administering to a subject a therapeutically effective amount of a compound of Embodiments 1-39, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of Embodiment 40. Embodiment 46. The compound or pharmaceutical composition of Embodiment 45, wherein the method comprises administering to the subject a therapeutically effective amount of an additional therapeutic agent or therapy selected from the group consisting of fedratinib (e.g., fedratinib dihydrochloride monohydrate), momelotinib (e.g., momelotinib dihydrochloride), pacritinib (e.g., pacritinib citrate), ruxolitinib (e.g., ruxolitinib phosphate), and a combination thereof. Embodiment 47. The compound or pharmaceutical composition of Embodiment 46, wherein the compound of any one of Embodiments 1-39, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of Embodiment 40, and the additional therapeutic agent or therapy are administered simultaneously, separately, or sequentially. Exemplary Formula (I-Eb) Embodiments Embodiment 1. A compound of Formula (I-Eb): Formula (I-Eb) or pharmaceutically acceptable salts thereof, wherein: R 1 is selected from the group consisting of: (a) C(O)OH, (b) C(O)NR d R e , and (c) C(O)OC1-6 alkyl, wherein the C1-6 alkyl is optionally substituted with 1-3 R c ; each R 2 and R 4 is independently selected from the group consisting of: halo, CN, C1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, C1-3 haloalkoxy, OH, and NR d R e ; m2 is 0, 1, or 2; m4 is 0, 1, 2, or 3; m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C 1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH 2 -, -CHR L - , and -C(R L )2-; and L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; wherein each R L is independently selected from the group consisting of: halo; cyano; - OH; -C1-6 alkoxy; -C1-6 haloalkoxy; -NR d R e ; C(=O)N(R f )2; S(O)0-2(C1-6 alkyl); S(O)0-2(C1-6 haloalkyl); S(O) 1-2 N(R f ) 2 ; -R b ; and C 1-6 alkyl optionally substituted with 1-6 R c ; n2 and n3 are independently 0, 1, or 2; and m8 is 0, 1, or 2. Ring C is selected from the group consisting of: ; c1 is 0, 1, 2, or 3; each R Y is independently selected from the group consisting of: R a and R b ; R aN is H or C1-6 alkyl optionally substituted with 1-3 R c ; Y 1 and Y 2 are independently N, CH, or CR Y ; yy represents the point of attachment to L; X is CH, C, or N; the is a single bond or a double bond; L C is selected from the group consisting of: a bond, -CH2-, -CHR a -, -C(R a )2-, -N(R d )-, and O, provided that when X is N, then L C is other than O; each R a is independently selected from the group consisting of: (a) halo; (b) cyano; (c) -OH; (d) oxo; (e) -C 1-6 alkoxy; (f) -C1-6 haloalkoxy; (g) -NR d R e ; (h) C(=O)C 1-6 alkyl; (i) C(=O)C1-6 haloalkyl; (j) C(=O)OH; (k) C(=O)OC 1-6 alkyl; (l) C(=O)OC 1-6 haloalkyl; (m) C(=O)N(R f )2; (n) S(O)0-2(C1-6 alkyl); (o) S(O) 0-2 (C 1-6 haloalkyl); (p) S(O)1-2N(R f )2; and (q) C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted with 1-6 R c ; each R b is independently selected from the group consisting of: -(L b )b-R b1 and -R b1 , wherein: each b is independently 1, 2, or 3; each -L b is independently selected from the group consisting of: -O-; -N(H)-; -N(C 1-3 alkyl)-; -S(O)0-2-; C(=O); and C1-3 alkylene; and each R b1 is independently selected from the group consisting of: C3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 R g ; each R c is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C1-6 haloalkoxy; -NR d R e ; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC1-6 haloalkyl; C(=O)OH; C(=O)N(R f )2; S(O)0-2(C1-6 alkyl); S(O)0-2(C1-6 haloalkyl); and S(O) 1-2 N(R f ) 2 ; each R d and R e is independently selected from the group consisting of: H; C(=O)C 1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC1-6 haloalkyl; C(=O)N(R f )2; S(O)1- 2(C1-6 alkyl); S(O)1-2(C1-6 haloalkyl); S(O)1-2N(R f )2; and C1-6 alkyl optionally substituted with 1-3 R h ; each R f is independently selected from the group consisting of: H and C1-6 alkyl optionally substituted with 1-3 R h ; each R g is independently selected from the group consisting of: R h ; C 1-3 alkyl; and C 1- 3 haloalkyl; and each R h is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C 1-6 haloalkoxy; -NH 2 , -N(H)(C 1-3 alkyl); and -N(C 1-3 alkyl) 2 . Embodiment 2. The compound of Embodiment 1, wherein m8 is 0. Embodiment 3. The compound of Embodiment 1 or 2, wherein a3 is 1. Embodiment 4. The compound of any one of Embodiments 1-3, wherein L A3 is -O-. Embodiment 5. The compound of any one of Embodiments 1-4, wherein a1a + a1b is 3. Embodiment 6. The compound of any one of Embodiments 1-4, wherein a1a + a1b is 4. Embodiment 7. The compound of any one of any one of Embodiments 1-4, wherein a1a + a1b is 2 or 5. Embodiment 8. The compound of any one of Embodiments 1-7, wherein each occurrence of L A1a and L A1b is -CH2-. Embodiment 9. The compound of any one of Embodiments 1-7, wherein one occurrence of L A1a is -CHR L - or -C(R L )2-; each remaining occurrence of L A1a is -CH2-; and each occurrence of L A1b is -CH2-. Embodiment 10. The compound of any one of Embodiments 1-7, wherein one occurrence of L A1b is -CHR L - or -C(R L )2-; each remaining occurrence of L A1b is -CH2-; and each occurrence of L A1a is -CH2-. Embodiment 11. The compound of any one of Embodiments 1-10, wherein L A4 is 4-10 membered heterocyclylene optionally substituted with 1-3 R a . Embodiment 12. The compound of any one of Embodiments 1-11, wherein L A4 is selected optionally substituted with 1-3 R a at one or more ring carbon atoms, wherein cc represents the point of attachment to L A1b ; or substituted with 1-3 R a at one or more ring carbon atoms, wherein cc represents the point of attachment to L A1b . Embodiment 13. The compound of Embodiments 11 or 12, wherein each R a present on L A4 is independently a C 1-3 alkyl optionally substituted with 1-3 F. Embodiment 14. The compound of any one of Embodiments 1-10, wherein L A4 is C3-10 cycloalkylene optionally substituted with 1-3 R a . Embodiment 15. The compound of any one of Embodiments 1-10 or 14, wherein L A4 is 1,4- cyclohexylene optionally substituted with 1-3 R a . Embodiment 16. The compound of any one of Embodiments 1-10, wherein L A4 is phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . Embodiment 17. The compound of any one of Embodiments 1-10 or 16, wherein L A4 is 1,4- phenylene optionally substituted with 1-3 R a . Embodiment 18. The compound of any one of Embodiments 1-10 or 16, wherein L A4 is 1,2- phenylene or 1,3-phenylene, each of which is optionally substituted with 1-3 R a . Embodiment 19. The compound of any one of Embodiments 1-18, wherein m6 is 0. Embodiment 20. The compound of any one of Embodiments 1-19, wherein the R a present on Ring A is C1-3 alkyl optionally substituted with 1-3 -F. Embodiment 21. The compound of any one of Embodiments 1-20, wherein R 1 is C(O)OH. Embodiment 22. The compound of any one of Embodiments 1-21, wherein m2 is 0. Embodiment 23. The compound of any one of Embodiments 1-22, wherein m4 is 0. Embodiment 24. The compound of any one of Embodiments 1-23, wherein Ring C is . Embodiment 25. The compound of any one of Embodiments 1-24, wherein Ring C is . Embodiment 26. The compound of any one of Embodiments 1-24, wherein Ring C is . Embodiment 27. The compound of any one of Embodiments 1-26, wherein R aN is C1-3 alkyl. Embodiment 28. The compound of any one of Embodiments 1-27, wherein R aN is methyl. Embodiment 29. The compound of any one of Embodiments 1-27, wherein c1 is 0. Embodiment 30. The compound of any one of Embodiments 1-29, wherein L C is a bond. Embodiment 31. The compound of any one of Embodiments 1-30, wherein X is CH. Embodiment 32. The compound of any one of Embodiments 1-23, wherein the Embodiment 33. The compound of any one of Embodiments 1-23, wherein the Embodiment 34. The compound of Embodiment 1, wherein the compound is a compound of Formula (I-Eb-1):

Formula (I-Eb-1) or pharmaceutically acceptable salts thereof, wherein: R 1 is C(O)OH or C(O)OC 1-6 alkyl; R a1 is C 1-3 alkyl optionally substituted with 1-3 F; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; and L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; n2 and n3 are independently 0, 1, or 2; and m8 is 0, 1, or 2. Embodiment 35. The compound of Embodiment 1, wherein the compound is a compound of Formula (I-Eb-2):

Formula (I-Eb-2) or pharmaceutically acceptable salts thereof, wherein: m6 is 0 or 1; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L )2-; L A4 is selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; m8 is 0, 1, or 2; n2 and n3 are independently 0, 1, or 2; and Ring , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C 1-3 alkyl optionally substituted with 1-3 F; and R aN is C1-3 alkyl. Embodiment 36. The compound of any one of Embodiments 1, 31, or 32, wherein the compound is a compound of Formula (I-Eb-3):

Formula (I-Eb-3) or pharmaceutically acceptable salts thereof, wherein: R 1 is C(O)OH or C(O)OC 1-6 alkyl; R a1 is C 1-3 alkyl optionally substituted with 1-3 F; a3 is 0 or 1; L A3 is selected from the group consisting of: -O-, -N(H)-, and –N(C1-3 alkyl)-; a1a and a1b are independently integers from 0 to 5, provided that a1a + a1b is from 2 to 5; L A1a and L A1b are independently selected from the group consisting of: -CH2-, -CHR L - , and -C(R L ) 2 -; L A4 is selected from the group consisting of: a) C3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a ; m8 is 0, 1, or 2; n2 and n3 are independently 0, 1, or 2; and Ring , wherein: c1 is 0 or 1; R a2 is selected from the group consisting of: halo (e.g., -F) and C1-3 alkyl optionally substituted with 1-3 F; and R aN is C 1-3 alkyl. In some embodiments of Formula (I-Eb-1), (I-Eb-2), or (I-Eb-3): a3 is 1; L A3 is -O-; a1a is 0; and L A4 is selected from the group consisting of: C3-10 cycloalkylene optionally substituted with 1-3 R a ; and phenylene optionally substituted with 1-3 R a . Embodiment 37. The compound of any one of Embodiments 1-36, wherein the wherein bb represents the point of attachment to Ring C. Embodiment 38. The compound of any one of Embodiments 1-34, wherein the compound is selected from the group consisting of 248, 249, 255, 256, 269, 269a, 270, 270a, 298, 298a, 299, 299a, 320, 320a, 320b, 321, 321a, 321b, 331, 331a, 331b, 332, 332a, 332b, 351, 351a, 351b, 352, 352a, and 352b as depicted in Table C1, or pharmaceutically acceptable salts thereof; or the compound is selected from the group consisting of 248, 249, 255, 256, 269, 269a, 270, 270a, 298, 298a, 299, 299a, 320, 320a, 320b, 321, 321a, and 321b, as depicted in Table C1, or pharmaceutically acceptable salts thereof; or the compound is selected from the group consisting of 248, 249, 255, 256, 269, 269a, 270, and 270a as depicted in Table C1, or pharmaceutically acceptable salts thereof. Embodiment 39. A pharmaceutical composition comprising the compound of any one of Embodiments 1-38, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. Embodiment 40. A compound of any one of Embodiments 1-38, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of Embodiment 39 for use in treatment. Embodiment 41. A compound of any one of Embodiments 1-38 or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of Embodiment 39 for use in the treatment of a cancer selected from the group consisting of essential thrombocythemia, polycythemia vera, myelofibrosis, and a combination thereof. Embodiment 42. The compound or pharmaceutical composition of Embodiment 41 and an additional therapeutic agent or therapy selected from the group consisting of fedratinib (e.g., fedratinib dihydrochloride monohydrate), momelotinib (e.g., momelotinib dihydrochloride), pacritinib (e.g., pacritinib citrate), ruxolitinib (e.g., ruxolitinib phosphate), and a combination thereof for use in treatment. Embodiment 43. The compound or pharmaceutical composition of Embodiment 42 and an additional therapeutic agent or therapy selected from the group consisting of fedratinib (e.g., fedratinib dihydrochloride monohydrate), momelotinib (e.g., momelotinib dihydrochloride), pacritinib (e.g., pacritinib citrate), ruxolitinib (e.g., ruxolitinib phosphate), and a combination thereof for use in treatment of a cancer selected from the group consisting of essential thrombocythemia, polycythemia vera, myelofibrosis, and a combination thereof. Embodiment 44. A compound of any one of Embodiments 1-38, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of Embodiment 39 for use in a method of treating cancer selected from the group consisting of essential thrombocythemia, polycythemia vera, myelofibrosis, and a combination thereof, the method comprising administering to a subject a therapeutically effective amount of a compound of Embodiments 1-38, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of Embodiment 39. Embodiment 45. The compound or pharmaceutical composition of Embodiment 44, wherein the method comprises administering to the subject a therapeutically effective amount of an additional therapeutic agent or therapy selected from the group consisting of fedratinib (e.g., fedratinib dihydrochloride monohydrate), momelotinib (e.g., momelotinib dihydrochloride), pacritinib (e.g., pacritinib citrate), ruxolitinib (e.g., ruxolitinib phosphate), and a combination thereof. Embodiment 46. The compound or pharmaceutical composition of Embodiment 45, wherein the compound of any one of Embodiments 1-38, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of Embodiment 39, and the additional therapeutic agent or therapy are administered simultaneously, separately, or sequentially. Exemplary Formula (I) Embodiments Embodiment 1. A compound of Formula (I):

Formula (I) or a pharmaceutically acceptable salt thereof, wherein: Ring A is selected from the group consisting of: (a) C 3-15 cycloalkylene or 3-15 membered heterocyclylene, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; and (b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 substituents independently selected from the group consisting of: R a and R b ; R 1 is selected from the group consisting of: (a) C(O)OH, (b) C(O)NR d R e , and (c) C(O)OC 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with 1-3 R c ; each R 2 , R 3 , R 4 , and R 5 is independently selected from the group consisting of: halo, CN, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, OH, and NR d R e ; m2 is 0, 1, or 2; m3 and m4 are independently 0, 1, 2, or 3; m5 is 0, 1, 2, 3, or 4; L is –(L A )n1–, wherein L A and n1 are defined according to (AA) or (BB): n1 is an integer from 3 to 15; and each L A is independently selected from the group consisting of: L A1 , L A3 , and L A4 , provided that 1-3 occurrences of L A is L ; n1 is an integer from 0 to 20; and each L A is independently selected from the group consisting of: L A1 and L A3 ; each L A1 is independently selected from the group consisting of: -CH2-, -CHR L -, and - C(R L )2-; each L A3 is independently selected from the group consisting of: -N(R d )-, -N(R b )-, -O- , -S(O) 0-2 -, and C(=O); each L A4 is independently selected from the group consisting of: (a) C 3-15 cycloalkylene or 3-15 membered heterocyclylene, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; and (b) C 6-15 arylene or 5-15 membered heteroarylene, each of which is optionally substituted with 1-6 substituents independently selected from the group consisting of: R a and R b ; provided that L does not contain any N-O, O-O, N-N, N-S(O) 0 , or O-S(O) 0-2 bonds; wherein each R L is independently selected from the group consisting of: halo; cyano; - OH; -C 1-6 alkoxy; -C 1-6 haloalkoxy; -NR d R e ; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); S(O)1-2N(R f )2; -R b ; and C1-6 alkyl optionally substituted with 1-6 R c ; Ring C is selected from the group consisting of: ; c1 is 0, 1, 2, or 3; each R Y is independently selected from the group consisting of: R a and R b ; R aN is H or C1-6 alkyl optionally substituted with 1-3 R c ; Y 1 and Y 2 are independently N, CH, or CR Y ; yy represents the point of attachment to L; X is CH, C, or N; the is a single bond or a double bond; L C is selected from the group consisting of: a bond, -CH 2 -, -CHR a -, -C(R a ) 2 -, -N(R d )-, and O, provided that when X is N, then L C is other than O; each R a is independently selected from the group consisting of: (a) halo; (b) cyano; (c) -OH; (d) oxo; (e) -C1-6 alkoxy; (f) -C1-6 haloalkoxy; (g) -NR d R e ; (h) C(=O)C1-6 alkyl; (i) C(=O)C1-6 haloalkyl; (j) C(=O)OH; (k) C(=O)OC1-6 alkyl; (l) C(=O)OC1-6 haloalkyl; (m) C(=O)N(R f ) 2 ; (n) S(O) 0-2 (C 1-6 alkyl); (o) S(O)0-2(C1-6 haloalkyl); (p) S(O)1-2N(R f )2; and (q) C 1-6 alkyl, C 2-6 alkenyl, or C 2-6 alkynyl, each optionally substituted with 1-6 R c ; each R b is independently selected from the group consisting of: -(L b )b-R b1 and -R b1 , wherein: each b is independently 1, 2, or 3; each -L b is independently selected from the group consisting of: -O-; -N(H)-; -N(C1-3 alkyl)-; -S(O) 0-2 -; C(=O); and C 1-3 alkylene; and each R b1 is independently selected from the group consisting of: C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C6-10 aryl, and 5-10 membered heteroaryl, each of which is optionally substituted with 1-3 R g ; each R c is independently selected from the group consisting of: halo; cyano; -OH; -C1- 6 alkoxy; -C1-6 haloalkoxy; -NR d R e ; C(=O)C1-6 alkyl; C(=O)C1-6 haloalkyl; C(=O)OC1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)OH; C(=O)N(R f ) 2 ; S(O) 0-2 (C 1-6 alkyl); S(O) 0-2 (C 1-6 haloalkyl); and S(O)1-2N(R f )2; each R d and R e is independently selected from the group consisting of: H; C(=O)C1-6 alkyl; C(=O)C 1-6 haloalkyl; C(=O)OC 1-6 alkyl; C(=O)OC 1-6 haloalkyl; C(=O)N(R f ) 2 ; S(O) 1- 2 (C 1-6 alkyl); S(O) 1-2 (C 1-6 haloalkyl); S(O) 1-2 N(R f ) 2 ; and C 1-6 alkyl optionally substituted with 1-3 R h ; each R f is independently selected from the group consisting of: H and C1-6 alkyl optionally substituted with 1-3 R h ; each R g is independently selected from the group consisting of: R h ; C1-3 alkyl; and C1- 3 haloalkyl; and each R h is independently selected from the group consisting of: halo; cyano; -OH; -C 1- 6 alkoxy; -C 1-6 haloalkoxy; -NH 2 , -N(H)(C 1-3 alkyl); and -N(C 1-3 alkyl) 2 . Embodiment 2. The compound of Embodiment 1, wherein Ring A is a phenylene optionally substituted with 1-3 R a . Embodiment 3. The compound of Embodiment 1 or 2, wherein Ring , wherein aa represents the point of attachment to L. Embodiment 4. The compound of any one of Embodiments 1-3, wherein Ring A is , wherein aa represents the point of attachment to L. Embodiment 5. The compound of any one of Embodiments 1-3, wherein Ring A is , wherein aa represents the point of attachment to L. Embodiment 6. The compound of any one of Embodiments 1-5, wherein one R a present on Ring A is C1-3 alkyl optionally substituted with 1-3 F. Embodiment 7. The compound of any one of Embodiments 1-6, wherein one R a present on Ring A is methyl or CF 3 . Embodiment 8. The compound of any one of Embodiments 1-7, wherein R 1 is C(O)OH. Embodiment 9. The compound of any one of Embodiments 1-8, wherein m2 is 0. Embodiment 10. The compound of any one of Embodiments 1-9, wherein m3 is 0. Embodiment 11. The compound of any one of Embodiments 1-10, wherein m4 is 0. Embodiment 12. The compound of any one of Embodiments 1-11, wherein m5 is 0. Embodiment 13. The compound of any one of Embodiments 1-8, wherein m2 is 0; m3 is 0; m4 is 0; and m5 is 0. Embodiment 14. The compound of any one of Embodiments 1-13, wherein Ring C is . Embodiment 15. The compound of any one of Embodiments 1-14, wherein Ring C is . Embodiment 16. The compound of any one of Embodiments 1-14, wherein Ring C is . Embodiment 17. The compound of any one of Embodiments 14-16, wherein c1 is 0. Embodiment 18. The compound of any one of Embodiments 14-17, wherein R aN is C1-3 alkyl. Embodiment 19. The compound of any one of Embodiments 14-18, wherein R aN is methyl. Embodiment 20. The compound of any one of Embodiments 14-19, wherein L C is a bond. Embodiment 21. The compound of any one of Embodiments 14-20, wherein X is CH. Embodiment 22. The compound of any one of Embodiments 1-14, wherein the Embodiment 23. The compound of any one of Embodiments 1-14, wherein the Embodiment 24. The compound of any one of Embodiments 1-23, wherein L is –(L A )n1-; and L A and n1 are defined according to (AA). Embodiment 25. The compound of any one of Embodiments 1-24, wherein n1 is an integer from 3 to 5. Embodiment 26. The compound of any one of Embodiments 1-24, wherein n1 is an integer from 5 to 9. Embodiment 27. The compound of any one of Embodiments 1-24 or 26, wherein n1 is 6, 7, or 8. Embodiment 28. The compound of any one of Embodiments 1-24, wherein n1 is an integer from 9 to 12. Embodiment 29. The compound of any one of Embodiments 1-28, wherein 1-2 occurrences of L A is L A4 . Embodiment 30. The compound of any one of Embodiments 1-28, wherein one occurrence of L A is L A4 . Embodiment 31. The compound of any one of Embodiments 1-28, wherein two occurrences of L A are L A4 . Embodiment 32. The compound of any one of Embodiments 1-31, wherein each L A4 is independently selected from the group consisting of: a) C 3-10 cycloalkylene or 4-10 membered heterocyclylene, each of which is optionally substituted with 1-3 R a ; and b) phenylene or 5-6 membered heteroarylene, each of which is optionally substituted with 1-3 R a . Embodiment 33. The compound of any one of Embodiments 1-32, wherein 1-4 occurrences of L A is L A3 . Embodiment 34. The compound of any one of Embodiments 1-33, wherein 1-3 occurrences of L A is L A3 . Embodiment 35. The compound of any one of Embodiments 1-34, wherein 0-1 occurrence of L A3 is C(=O); and each remaining occurrence of L A3 is independently selected from the group consisting of: -O-; -N(H)-; and -N(C1-3 alkyl)-. Embodiment 36. The compound of any one of Embodiments 1-35, wherein 2-7 occurrences of L A is L A1 . Embodiment 37. The compound of any one of Embodiments 1-36, wherein 2-5 occurrences of L A is L A1 . Embodiment 38. The compound of any one of Embodiments 1-37, wherein 0-2 occurrences of L A1 is -CHR L - or -C(R L )2-; and each remaining occurrence of L A1 is -CH2-. Embodiment 39. The compound of any one of Embodiments 1-38, wherein each occurrence of L A1 is -CH 2 -. Embodiment 40. The compound of any one of Embodiments 1-38, wherein one occurrence of L A1 is -CHR L - or -C(R L ) 2 -; and each remaining occurrence of L A1 is -CH 2 -. Embodiment 41. The compound of any one of Embodiments 1-40, wherein each R L is independently selected from the group consisting of: -F and -C 1-3 alkyl optionally substituted with 1-3 F. Embodiment 42. The compound of any one of Embodiments 1-24, wherein L is –(L A )n1-, and L A and n1 are defined according to (AA): n1 is an integer from 5 to 9; 1-2 occurrences of L A is L A4 ; 2-7 occurrences of L A are L A1 ; and 1-4 occurrences of L A is L A3 . Embodiment 43. A compound selected from the group consisting of 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 116a, 117, 117a, 118, 119, 120, 121, 122, 123, 124, 124a, 125, 125a, 126, 127, 128, 129, 130, 131, 131a, 132, 133, 134, 135, 136, 137, 138, 138a, 138b, 139, 140, 141, 142, 143, 144, 145, 146, 146a, 146b, 147, 147a, 147b, 148, 148a, 148b, 149, 150, 151, 152, 153, 154, 154a, 155, 156, 157, 158, 159, 159a, 160, 160a, 160b, 161, 161a, 162, 162a, 162b, 163, 164, 164a, 164b, 165, 166, 167, 167a, 167b, 168, 168a, 169, 169a, 169b, 170, 171, 171a, 172, 173, 173a, 173b, 174, 174a, 174b, 175, 176, 176a, 177, 177a, 178, 179, 179a, 180, 180a, 180b, 181, 182, 182a, 183, 183a, 184, 185, 185a, 186, 186a, 186b, 187, 187a, 187b, 188, 189, 190, 191, 192, 193, 193a, 193b, 194, 195, 196, 197, 197a, 197b, 198, 199, 200, 201, 202, 202a, 202b, 203, 204, 204a, 205, 206, 207, 207a, 207b, 208, 209, 209a, 210, 210a, 210b, 211, 211a, 212, 212a, 213, 214, 214a, 215, 215a, 216, 216a, 216b, 217, 218, 218a, 218b, 219, 219a, 219b, 220, 221, 222, 223, 224, 224a, 224b, 225, 226, 227, 227a, 228, 229, 230, 231, 231a, 231b, 232, 233, 234, 235, 236, 237, 238, 239, 239a, 240, 240a, 240b, 241, 242, 243, 244, 245, 245a, 245b, 246, 246a, 246b, 247, 248, 249, 250, 250a, 251, 251a, 252, 252a, 253, 253a, 254, 254a, 255, 256, 257, 258, 259, 259a, 260, 261, 262, 262a, 262b, 263, 263a, 263b, 264, 265, 266, 266a, 266b, 267, 268, 268a, 268b, 269, 269a, 270, 270a, 271, 271a, 271b, 272, 272a, 272b, 273, 273a, 273b, 274, 274a, 274b, 275, 275a, 275b, 276, 276a, 276b, 277, 278, 278a, 278b, 279, 280, 281, 281a, 281b, 282, 282a, 282b, 283, 283a, 283b, 284, 284a, 284b, 285, 286, 287, 288, 289, 289a, 289b, 290, 291, 292, 293, 293a, 294, 295, 296, 296a, 297, 297a, 297b, 298, 298a, 299, 299a, 300, 300a, 300b, 301, 301a, 301b, 302, 303, 304, 305, 306, 307, 307a, 307b, 308, 308a, 308b, 309, 310, 310a, 310b, 311, 311a, 311b, 312, 313, 314, 316, 316a, 317, 318, 318a, 319, 319a, 320, 320a, 320b, 321, 321a, 321b, 322, 322a, 323, 323a, 324, 324a, 325, 325a, 326, 326a, 327, 327a, 328, 328a, 329, 329a, 330, 330a, 331, 331a, 331b, 332, 332a, 332b, 333, 333a, 333b, 334, 334a, 335, 335a, 336, 336a, 337, 337a, 338, 338a, 339, 339a, 339b, 340, 340a, 341, 341a, 342, 342a, 343, 343a, 343b, 343c, 343d, 344, 344a, 344b, 344c, 344d, 345, 345a, 346, 346a, 346b, 347, 347a, 347b, 349, 349a, 349b, 350, 350a, 351, 351a, 351b, 352, 352a, 352b, 353, 353a, 353b, 354, 354a, 354b, 355, 356, 357, as depicted in Table C1, or a pharmaceutically acceptable salt thereof; or the compound is selected from the group consisting of 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 116a, 117, 117a, 118, 119, 120, 121, 122, 123, 124, 124a, 125, 125a, 126, 127, 128, 129, 130, 131, 131a, 132, 133, 134, 135, 136, 137, 138, 138a, 138b, 139, 140, 141, 142, 143, 144, 145, 146, 146a, 146b, 147, 147a, 147b, 148, 148a, 148b, 149, 150, 151, 152, 153, 154, 154a, 155, 156, 157, 158, 159, 159a, 160, 160a, 160b, 161, 161a, 162, 162a, 162b, 163, 164, 164a, 164b, 165, 166, 167, 167a, 167b, 168, 168a, 169, 169a, 169b, 170, 171, 171a, 172, 173, 173a, 173b, 174, 174a, 174b, 175, 176, 176a, 177, 177a, 178, 179, 179a, 180, 180a, 180b, 181, 182, 182a, 183, 183a, 184, 185, 185a, 186, 186a, 187, 187a, 187b, 188, 189, 190, 191, 192, 193, 193a, 193b, 194, 195, 196, 197, 197a, 197b, 198, 199, 200, 201, 202, 202a, 202b, 203, 204, 204a, 205, 206, 207, 207a, 207b, 208, 209, 209a, 210, 210a, 211, 211a, 212, 212a, 213, 214, 214a, 215, 215a, 216, 216a, 216b, 217, 218, 218a, 219, 219a, 219b, 220, 221, 222, 223, 224, 224a, 224b, 225, 226, 227, 227a, 228, 229, 230, 231, 231a, 231b, 232, 233, 234, 235, 236, 237, 238, 239, 239a, 240, 240a, 240b, 241, 242, 243, 244, 245, 245a, 245b, 246, 246a, 246b, 247, 248, 249, 250, 250a, 251, 251a, 252, 252a, 253, 253a, 254, 254a, 255, 256, 257, 258, 259, 259a, 260, 261, 262, 262a, 262b, 263, 263a, 263b, 264, 265, 266, 266a, 266b, 267, 268, 268a, 268b, 269, 269a, 270, 270a, 271, 271a, 271b, 272, 272a, 272b, 273, 273a, 273b, 274, 274a, 274b, 275, 275a, 276, 276a, 276b, 277, 278, 278a, 278b, 279, 280, 281, 281a, 281b, 282, 282a, 282b, 283, 283a, 283b, 284, 284a, 284b, 285, 286, 287, 288, 289, 289a, 289b, 290, 291, 292, 293, 293a, 294, 295, 296, 296a, 297, 297a, 297b, 298, 298a, 299, 299a, 300, 300a, 300b, 301, 301a, 301b, 302, 303, 304, 305, 306, 307, 307a, 307b, 308, 308a, 308b, 309, 310, 310a, 310b, 311, 311a, 312, 313, 314, 316, 316a, 317, 318, 318a, 319, 319a, 320, 320a, 320b, 321, 321a, 321b, 322, 322a, 323, 323a, 324, 324a, 325, 325a, 326, 326a, 327, 327a, 328, 328a, 329, 329a, 330, 330a, 331, 331a, 331b, 332, 332a, 332b, 333, 333a, 334, 334a, 335, 335a, 336, 336a, 337, 337a, 338, 338a, 339, 339a, 340, 340a, 341, 341a, 342, 342a, 343, 343a, 343b, 343c, 343d, 344, 344a, 344b, 344c, 344d, 345, 345a, 346, 346a, 346b, 347, 347a, 347b, 349, 349a, 349b, 350, 350a, 351, 351a, 351b, 352, 352a, and 352b as depicted in Table C1, or a pharmaceutically acceptable salt thereof; or the compound is selected from the group consisting of 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 116a, 117, 117a, 118, 119, 120, 121, 122, 123, 124, 124a, 125, 125a, 126, 127, 128, 129, 130, 131, 131a, 132, 133, 134, 135, 136, 137, 138, 138a, 138b, 139, 140, 141, 142, 143, 144, 145, 146, 146a, 146b, 147, 147a, 147b, 148, 148a, 148b, 149, 150, 151, 152, 153, 154, 154a, 155, 156, 157, 158, 159, 159a, 160, 160a, 160b, 161, 161a, 162, 162a, 162b, 163, 164, 164a, 164b, 165, 166, 167, 167a, 167b, 168, 168a, 169, 169a, 169b, 170, 171, 171a, 172, 173, 173a, 173b, 174, 174a, 174b, 175, 176, 176a, 177, 177a, 178, 179, 179a, 180, 180a, 180b, 181, 182, 182a, 183, 183a, 184, 185, 185a, 186, 186a, 187, 187a, 187b, 188, 189, 190, 191, 192, 193, 193a, 193b, 194, 195, 196, 197, 197a, 197b, 198, 199, 200, 201, 202, 202a, 202b, 203, 204, 204a, 205, 206, 207, 207a, 207b, 208, 209, 209a, 210, 210a, 211, 211a, 212, 212a, 213, 214, 214a, 215, 215a, 216, 216a, 216b, 217, 218, 218a, 219, 219a, 219b, 220, 221, 222, 223, 224, 224a, 224b, 225, 226, 227, 227a, 228, 229, 230, 231, 231a, 231b, 232, 233, 234, 235, 236, 237, 238, 239, 239a, 240, 240a, 240b, 241, 242, 243, 244, 245, 245a, 245b, 246, 246a, 246b, 247, 248, 249, 250, 250a, 251, 251a, 252, 252a, 253, 253a, 254, 254a, 255, 256, 257, 258, 259, 259a, 260, 261, 262, 262a, 262b, 263, 263a, 263b, 264, 265, 266, 266a, 266b, 267, 268, 268a, 268b, 269, 269a, 270, 270a, 271, 271a, 272, 272a, 272b, 273, 273a, 273b, 274, 274a, 274b, 275, 275a, 276, 276a, 276b, 277, 278, 278a, 278b, 279, 280, 281, 281a, 281b, 282, 282a, 282b, 283, 283a, 283b, 284, 284a, 284b, 285, 286, 287, 288, 289, 289a, 289b, 290, 291, 292, 293, 293a, 294, 295, 296, 296a, 297, 297a, 297b, 298, 298a, 299, 299a, 300, 300a, 300b, 301, 301a, 301b, 302, 303, 304, 305, 306, 307, 307a, 307b, 308, 308a, 308b, 309, 310, 310a, 310b, 311, 311a, 312, 313, 314, 315, 315a, 316, 316a, 317, 318, 318a, 319, 319a, 320, 320a, 320b, 321, 321a, 321b, 322, 322a, 323, 323a, 324, 324a, 325, 325a, 326, 326a, 327, 327a, 328, 328a, 329, 329a, 330, 330a, 331, 331a, 331b, 332, 332a, 332b, 333, 333a as depicted in Table C1, or a pharmaceutically acceptable salt thereof; or the compound is selected from the group consisting of 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 116a, 117, 117a, 118, 119, 120, 121, 122, 123, 124, 124a, 125, 125a, 126, 127, 128, 129, 130, 131, 131a, 132, 133, 134, 135, 136, 137, 138, 138a, 138b, 139, 140, 141, 142, 143, 144, 145, 146, 146a, 146b, 147, 147a, 147b, 148, 148a, 148b, 149, 150, 151, 152, 153, 154, 154a, 155, 156, 157, 158, 159, 159a, 160, 160a, 160b, 161, 161a, 162, 162a, 162b, 163, 164, 164a, 164b, 165, 166, 167, 167a, 167b, 168, 168a, 169, 169a, 169b, 170, 171, 171a, 172, 173, 173a, 173b, 174, 174a, 174b, 175, 176, 176a, 177, 177a, 178, 179, 179a, 180, 180a, 180b, 181, 182, 182a, 183, 183a, 184, 185, 185a, 186, 186a, 187, 187a, 187b, 188, 189, 190, 191, 192, 193, 193a, 193b, 194, 195, 196, 197, 197a, 197b, 198, 199, 200, 201, 202, 202a, 202b, 203, 204, 204a, 205, 206, 207, 207a, 207b, 208, 209, 209a, 210, 210a, 211, 211a, 212, 212a, 213, 214, 214a, 215, 215a, 216, 216a, 216b, 217, 218, 218a, 219, 219a, 219b, 220, 221, 222, 223, 224, 224a, 224b, 225, 226, 227, 227a, 228, 229, 230, 231, 231a, 231b, 232, 233, 234, 235, 236, 237, 238, 239, 239a, 240, 240a, 240b, 241, 242, 243, 244, 245, 245a, 245b, 246, 246a, 246b, 247, 248, 249, 250, 250a, 251, 251a, 252, 252a, 253, 253a, 254, 254a, 255, 256, 257, 258, 259, 259a, 260, 261, 262, 262a, 262b, 263, 263a, 263b, 264, 265, 266, 266a, 266b, 267, 268, 268a, 268b, 269, 269a, 270, 270a, 271, 271a, 272, 272a, 272b, 273, 273a, 273b, 274, 274a, 274b, 275, 275a, 276, 276a, 276b, 277, 278, 278a, 278b, 279, 280, 281, 281a, 281b, 282, 282a, 282b, 283, 283a, 283b, 284, 284a, 284b, 285, 286, 287, 288, 289, 289a, 289b, 290, 291, 292, 293, 293a, 294, 295, 296, 296a, 297, 297a, 297b, 298, 298a, 299, 299a, 300, 300a, 300b, 301, 301a, 301b, 302, 303, 304, 305, 306, 307, 307a, 307b, 308, 308a, 308b, 309, 310, 310a, 311, 311a, 312, 313, 314, 315, 315a, 316, 316a, 317, 318, 318a, 319, 319a, 320, 320a, 320b, 321, 321a, 321b, 322, 322a, 323, 323a, 324, 324a, 325, 325a, 326, 326a, 327, 327a as depicted in Table C1, or a pharmaceutically acceptable salt thereof; or the compound is selected from the group consisting of 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 116a, 117, 117a, 118, 119, 120, 121, 122, 123, 124, 124a, 125, 125a, 126, 127, 128, 129, 130, 131, 131a, 132, 133, 134, 135, 136, 137, 138, 138a, 138b, 139, 140, 141, 142, 143, 144, 145, 146, 146a, 146b, 147, 147a, 147b, 148, 148a, 148b, 149, 150, 151, 152, 153, 154, 154a, 155, 156, 157, 158, 159, 159a, 160, 160a, 160b, 161, 161a, 162, 162a, 162b, 163, 164, 164a, 164b, 165, 166, 167, 167a, 167b, 168, 168a, 169, 169a, 169b, 170, 171, 171a, 172, 173, 173a, 173b, 174, 174a, 174b, 175, 176, 176a, 177, 177a, 178, 179, 179a, 180, 180a, 180b, 181, 182, 182a, 183, 183a, 184, 185, 185a, 186, 186a, 187, 187a, 187b, 188, 189, 190, 191, 192, 193, 193a, 193b, 194, 195, 196, 197, 197a, 197b, 198, 199, 200, 201, 202, 202a, 202b, 203, 204, 204a, 205, 206, 207, 207a, 207b, 208, 209, 209a, 210, 210a, 211, 211a, 212, 212a, 213, 214, 214a, 215, 215a, 216, 216a, 216b, 217, 218, 218a, 219, 219a, 219b, 220, 221, 222, 223, 224, 224a, 224b, 225, 226, 227, 227a, 228, 229, 230, 231, 231a, 231b, 232, 233, 234, 235, 236, 237, 238, 239, 239a, 240, 240a, 240b, 241, 242, 243, 244, 245, 245a, 245b, 246, 246a, 246b, 247, 248, 249, 250, 250a, 251, 251a, 252, 252a, 253, 253a, 254, 254a, 255, 256, 257, 258, 259, 259a, 260, 261, 262, 262a, 262b, 263, 263a, 263b, 264, 265, 266, 266a, 266b, 267, 268, 268a, 268b, 269, 269a, 270, 270a, 271, 271a, 272, 272a, 272b, 273, 273a, 273b, 274, 274a, 274b, 275, 275a, 276, 276a, 276b, 277, 278, 278a, 278b, 279, 280, 281, 281a, 281b, 282, 282a, 282b, 283, 283a, 283b, 284, 284a, 284b, 285, 286, 287, 288, 289, 289a, 289b, 290, 291 as depicted in Table C1, or a pharmaceutically acceptable salt thereof; or the compound is selected from the group consisting of 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 116a, 117, 117a, 118, 119, 120, 121, 122, 123, 124, 124a, 125, 125a, 126, 127, 128, 129, 130, 131, 131a, 132, 133, 134, 135, 136, 137, 138, 138a, 139, 140, 141, 142, 143, 144, 145, 146, 146a, 147, 147a, 148, 148a, 149, 150, 151, 152, 153, 154, 154a, 155, 156, 157, 158, 159, 159a, 160, 160a, 160b, 161, 161a, 162, 162a, 162b, 163, 164, 164a, 164b, 165, 166, 167, 167a, 167b, 168, 168a, 169, 169a, 169b, 170, 171, 171a, 172, 173, 173a, 173b, 174, 174a, 174b, 175, 176, 176a, 177, 177a, 178, 179, 179a, 180, 180a, 180b, 181, 182, 182a, 183, 183a, 184, 185, 185a, 186, 186a, 187, 187a, 187b, 188, 189, 190, 191, 192, 193, 193a, 193b, 194, 195, 196, 197, 197a, 197b, 198, 199, 200, 201, 202, 202a, 202b, 203, 204, 204a, 205, 206, 207, 207a, 207b, 208, 209, 209a, 210, 210a, 211, 211a, 212, 212a, 213, 214, 214a, 215, 215a, 216, 216a, 216b, 217, 218, 218a, 219, 219a, 219b, 220, 221, 222, 223, 224, 224a, 224b, 225, 226, 227, 227a, 228, 229, 230, 231, 231a, 231b, 232, 233, 234, 235, 236, 237, 238, 239, 239a, 240, 240a, 240b, and 241 as depicted in Table C1, or a pharmaceutically acceptable salt thereof; or the compound is selected from the group consisting of 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 116a, 117, 117a, 118, 119, 120, 121, 122, 123, 124, 124a, 125, 125a, 126, 127, 128, 129, 130, 131, 131a, 132, 133, 134, 135, 136, 137, 138, 138a, 139, 140, 141, 142, 143, 144, 145, 146, 146a, 147, 147a, 148, 148a, 149, 150, 151, 152, 153, 154, 154a, 155, and 156, as depicted in Table C1, or a pharmaceutically acceptable salt thereof. Embodiment 44. The compound of Embodiment 43, wherein the compound does not exhibit Ymin (6 h) ≥ 70% and/or Ymin (24 h) ≥ 70% under conditions described in Example B1. Embodiment 45. A pharmaceutical composition comprising a compound of any one of Embodiments 1-44, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. Embodiment 46. A compound of any one of Embodiments 1-44, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of Embodiment 45 for use in treatment. Embodiment 47. A compound of any one of Embodiments 1-44 or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of Embodiment 45 for use in the treatment of a cancer selected from the group consisting of essential thrombocythemia, polycythemia vera, myelofibrosis, and a combination thereof. Embodiment 48. The compound or pharmaceutical composition of Embodiment 47 and an additional therapeutic agent or therapy selected from the group consisting of fedratinib (e.g., fedratinib dihydrochloride monohydrate), momelotinib (e.g., momelotinib dihydrochloride), pacritinib (e.g., pacritinib citrate), ruxolitinib (e.g., ruxolitinib phosphate), and a combination thereof for use in treatment. Embodiment 49. The compound or pharmaceutical composition of Embodiment 48 and an additional therapeutic agent or therapy selected from the group consisting of fedratinib (e.g., fedratinib dihydrochloride monohydrate), momelotinib (e.g., momelotinib dihydrochloride), pacritinib (e.g., pacritinib citrate), ruxolitinib (e.g., ruxolitinib phosphate), and a combination thereof for use in treatment of a cancer selected from the group consisting of essential thrombocythemia, polycythemia vera, myelofibrosis, and a combination thereof. Embodiment 50. A compound of any one of Embodiments 1-44, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of Embodiment 45 for use in a method of treating cancer selected from the group consisting of essential thrombocythemia, polycythemia vera, myelofibrosis, and a combination thereof, the method comprising administering to a subject a therapeutically effective amount of a compound of Embodiments 1-44, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of Embodiment 45. Embodiment 51. The compound or pharmaceutical composition of Embodiment 50, wherein the method comprises administering to the subject a therapeutically effective amount of an additional therapeutic agent or therapy selected from the group consisting of fedratinib (e.g., fedratinib dihydrochloride monohydrate), momelotinib (e.g., momelotinib dihydrochloride), pacritinib (e.g., pacritinib citrate), ruxolitinib (e.g., ruxolitinib phosphate), and a combination thereof. Embodiment 52. The compound or pharmaceutical composition of Embodiment 54, wherein the compound of any one of Embodiments 1-44, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of Embodiment 45, and the additional therapeutic agent or therapy are administered simultaneously, separately, or sequentially.