Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHODS FOR DELAYING, PREVENTING, AND TREATING ACQUIRED RESISTANCE TO RAS INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2021/257736
Kind Code:
A1
Abstract:
The present disclosure relates to compositions and methods for the treatment of diseases or disorders (e.g., cancer) with bi-steric inhibitors of mTOR in combination with RAS inhibitors. Specifically, in some embodiments this disclosure includes compositions and methods for inducing apoptosis of tumor cells and/or for delaying, preventing, or treating acquired resistance to RAS inhibitors using bi-steric mTOR inhibitors.

Inventors:
SINGH MALLIKA (US)
JIANG JINGJING (US)
YANG YU CHI (US)
EVANS JAMES W (US)
SCHULZE CHRISTOPHER J (US)
Application Number:
PCT/US2021/037679
Publication Date:
December 23, 2021
Filing Date:
June 16, 2021
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
REVOLUTION MEDICINES INC (US)
International Classes:
A61K31/436; A61K31/496; A61K31/506; A61P35/00
Domestic Patent References:
WO2020055761A12020-03-19
WO2019212990A12019-11-07
WO2019212991A12019-11-07
WO2020132597A12020-06-25
WO2021091967A12021-05-14
WO2020132597A12020-06-25
WO2021113595A12021-06-10
WO2021107160A12021-06-03
WO2021106231A12021-06-03
WO2021088458A12021-05-14
WO2021086833A12021-05-06
WO2021085653A12021-05-06
WO2021081212A12021-04-29
WO2021058018A12021-04-01
WO2021057832A12021-04-01
WO2021055728A12021-03-25
WO2021031952A12021-02-25
WO2021027911A12021-02-18
WO2021023247A12021-02-11
WO2020259513A12020-12-30
WO2020259432A12020-12-30
WO2020234103A12020-11-26
WO2020233592A12020-11-26
WO2020216190A12020-10-29
WO2020178282A12020-09-10
WO2020146613A12020-07-16
WO2020118066A12020-06-11
WO2020113071A12020-06-04
WO2020106647A22020-05-28
WO2020102730A12020-05-22
WO2020101736A12020-05-22
WO2020097537A22020-05-14
WO2020086739A12020-04-30
WO2020081282A12020-04-23
WO2020050890A22020-03-12
WO2020047192A12020-03-05
WO2020035031A12020-02-20
WO2020028706A12020-02-06
WO2019241157A12019-12-19
WO2019232419A12019-12-05
WO2019217691A12019-11-14
WO2019217307A12019-11-14
WO2019215203A12019-11-14
WO2019213526A12019-11-07
WO2019213516A12019-11-07
WO2019155399A12019-08-15
WO2019150305A12019-08-08
WO2019110751A12019-06-13
WO2019099524A12019-05-23
WO2019051291A12019-03-14
WO2018218070A22018-11-29
WO2018218071A12018-11-29
WO2018218069A12018-11-29
WO2018217651A12018-11-29
WO2018206539A12018-11-15
WO2018143315A12018-08-09
WO2018140600A12018-08-02
WO2018140599A12018-08-02
WO2018140598A12018-08-02
WO2018140514A12018-08-02
WO2018140513A12018-08-02
WO2018140512A12018-08-02
WO2018119183A22018-06-28
WO2018112420A12018-06-21
WO2018068017A12018-04-12
WO2018064510A12018-04-05
WO2017201161A12017-11-23
WO2017172979A12017-10-05
WO2017100546A12017-06-15
WO2017087528A12017-05-26
WO2017058807A12017-04-06
WO2017058805A12017-04-06
WO2017058728A12017-04-06
WO2017058902A12017-04-06
WO2017058792A12017-04-06
WO2017058768A12017-04-06
WO2017058915A12017-04-06
WO2017015562A12017-01-26
WO2016168540A12016-10-20
WO2016164675A12016-10-13
WO2016049568A12016-03-31
WO2016049524A12016-03-31
WO2015054572A12015-04-16
WO2014152588A12014-09-25
WO2014143659A12014-09-18
WO2013155223A12013-10-17
WO2019212990A12019-11-07
WO2020106640A12020-05-28
WO2020018282A12020-01-23
WO2020033413A22020-02-13
WO2019234405A12019-12-12
WO2019227040A12019-11-28
WO2019217933A12019-11-14
WO2019204442A12019-10-24
WO2019204449A12019-10-24
WO2019204505A22019-10-24
WO2019137985A12019-07-18
WO2019055540A12019-03-21
WO2018237084A12018-12-27
WO2018212774A12018-11-22
WO2018195439A22018-10-25
WO2018011351A22018-01-18
WO2018005678A12018-01-04
WO2017137370A12017-08-17
WO2017112777A12017-06-29
WO2017106520A12017-06-22
WO2017096045A12017-06-08
WO2017079864A12017-05-18
WO2016179558A12016-11-10
WO2016176338A12016-11-03
WO2016100546A12016-06-23
WO2016040806A12016-03-17
WO2018204416A12018-11-08
WO2019212991A12019-11-07
WO2001037820A22001-05-31
WO2001032651A12001-05-10
WO2002068406A22002-09-06
WO2002066470A12002-08-29
WO2002055501A22002-07-18
WO2004005279A22004-01-15
WO2004007481A22004-01-22
WO2004007458A12004-01-22
WO2004009784A22004-01-29
WO2002059110A12002-08-01
WO1999045009A11999-09-10
WO2000059509A12000-10-12
WO1999061422A11999-12-02
WO2000012089A12000-03-09
WO2000002871A12000-01-20
WO2005016894A12005-02-24
WO1996033980A11996-10-31
WO1996030347A11996-10-03
WO1997030034A11997-08-21
WO1997030044A11997-08-21
WO1997038994A11997-10-23
WO1997049688A11997-12-31
WO1998002434A11998-01-22
WO1997038983A11997-10-23
WO1995019774A11995-07-27
WO1995019970A11995-07-27
WO1997013771A11997-04-17
WO1998002437A11998-01-22
WO1998002438A11998-01-22
WO1997032881A11997-09-12
WO1998033798A21998-08-06
WO1997032880A11997-09-12
WO1997002266A11997-01-23
WO1997027199A11997-07-31
WO1998007726A11998-02-26
WO1997034895A11997-09-25
WO1996031510A11996-10-10
WO1998014449A11998-04-09
WO1998014450A11998-04-09
WO1998014451A11998-04-09
WO1995009847A11995-04-13
WO1997019065A11997-05-29
WO1998017662A11998-04-30
WO1999035146A11999-07-15
WO1999035132A11999-07-15
WO1999007701A11999-02-18
WO1992020642A11992-11-26
WO2006044453A12006-04-27
WO2009003608A12009-01-08
WO2009055730A12009-04-30
WO2006122806A22006-11-23
WO2008070740A12008-06-12
WO2005011700A12005-02-10
WO2021110796A12021-06-10
WO2021088945A12021-05-14
WO2021073439A12021-04-22
WO2021061706A12021-04-01
WO2021061515A12021-04-01
WO2021043077A12021-03-11
WO2021033153A12021-02-25
WO2021028362A12021-02-18
WO2021018287A12021-02-04
WO2020259679A12020-12-30
WO2020249079A12020-12-17
WO2020210384A12020-10-15
WO2020201991A12020-10-08
WO2020181283A12020-09-10
WO2020177653A12020-09-10
WO2020165734A12020-08-20
WO2020165733A12020-08-20
WO2020165732A12020-08-20
WO2020156243A12020-08-06
WO2020156242A12020-08-06
WO2020108590A12020-06-04
WO2020104635A12020-05-28
WO2020094104A12020-05-14
WO2020094018A12020-05-14
WO2020081848A12020-04-23
WO2020073949A12020-04-16
WO2020073945A12020-04-16
WO2020072656A12020-04-09
WO2020065453A12020-04-02
WO2020065452A12020-04-02
WO2020063760A12020-04-02
WO2020061103A12020-03-26
WO2020061101A12020-03-26
WO2020033828A12020-02-13
WO2020033286A12020-02-13
WO2020022323A12020-01-30
WO2019233810A12019-12-12
WO2019213318A12019-11-07
WO2019183367A12019-09-26
WO2019183364A12019-09-26
WO2019182960A12019-09-26
WO2019167000A12019-09-06
WO2019165073A12019-08-29
WO2019158019A12019-08-22
WO2019152454A12019-08-08
WO2019051469A12019-03-14
WO2019051084A12019-03-14
WO2018218133A12018-11-29
WO2018172984A12018-09-27
WO2018160731A12018-09-07
WO2018136265A12018-07-26
WO2018136264A12018-07-26
WO2018130928A12018-07-19
WO2018129402A12018-07-12
WO2018081091A12018-05-03
WO2018057884A12018-03-29
WO2018013597A12018-01-18
WO2017216706A12017-12-21
WO2017211303A12017-12-14
WO2017210134A12017-12-07
WO2017156397A12017-09-14
WO2017100279A12017-06-15
WO2017079723A12017-05-11
WO2017078499A22017-05-11
WO2016203406A12016-12-22
WO2016203405A12016-12-22
WO2016203404A12016-12-22
WO2016196591A12016-12-08
WO2016191328A12016-12-01
WO2015107495A12015-07-23
WO2015107494A12015-07-23
WO2015107493A12015-07-23
WO2014176488A12014-10-30
WO2014113584A12014-07-24
WO2006121168A12006-11-16
WO2010003118A12010-01-07
WO2011090754A12011-07-28
WO2011028683A12011-03-10
WO2013039954A12013-03-21
WO2005007190A12005-01-27
WO2007133822A12007-11-22
WO2005055808A22005-06-23
WO1999040196A11999-08-12
WO2001003720A22001-01-18
WO1999020758A11999-04-29
WO2006083289A22006-08-10
WO2005115451A22005-12-08
WO2011051726A22011-05-05
WO1996033172A11996-10-24
WO1996027583A11996-09-12
WO1998007697A11998-02-26
WO1998003516A11998-01-29
WO1998034918A11998-08-13
WO1998034915A11998-08-13
WO1998033768A11998-08-06
WO1998030566A11998-07-16
WO1990005719A11990-05-31
WO1999052910A11999-10-21
WO1999052889A11999-10-21
WO1999029667A11999-06-17
WO1999007675A11999-02-18
Foreign References:
US20170204187A12017-07-20
US6352694B12002-03-05
US6534055B12003-03-18
US6905680B22005-06-14
US6692964B12004-02-17
US5858358A1999-01-12
US6887466B22005-05-03
US6905681B12005-06-14
US7144575B22006-12-05
US7067318B22006-06-27
US7172869B22007-02-06
US7232566B22007-06-19
US7175843B22007-02-13
US7572631B22009-08-11
US5883223A1999-03-16
US6905874B22005-06-14
US6797514B22004-09-28
US6867041B22005-03-15
US6258812B12001-07-10
US6630500B22003-10-07
US6515004B12003-02-04
US6713485B22004-03-30
US5521184A1996-05-28
US5770599A1998-06-23
US5747498A1998-05-05
US5990141A1999-11-23
US8623885B22014-01-07
EP0520722A11992-12-30
EP0566226A11993-10-20
EP0787772A21997-08-06
EP0837063A11998-04-22
DE19629652A11998-01-29
EP0682027A11995-11-15
US5789427A1998-08-04
US5650415A1997-07-22
US5656643A1997-08-12
US6656963B22003-12-02
US20210085677A12021-03-25
US10988466B22021-04-27
US10858359B22020-12-08
US10934302B12021-03-02
US10954243B22021-03-23
US6111090A2000-08-29
US8586023B22013-11-19
US7025962B12006-04-11
EP1947183A12008-07-23
US7812135B22010-10-12
US8388967B22013-03-05
US8591886B22013-11-26
US7618632B22009-11-17
EP1866339A22007-12-19
EP0606046A11994-07-13
EP0780386A11997-06-25
EP1786785A22007-05-23
EP1181017A12002-02-27
EP0818442A21998-01-14
EP1004578A22000-05-31
US20090012085A12009-01-08
US5863949A1999-01-26
US5861510A1999-01-19
US20030162712A12003-08-28
US6413932B12002-07-02
US6727225B22004-04-27
US20020042368A12002-04-11
US5981245A1999-11-09
US5728813A1998-03-17
US5969110A1999-10-19
US6596852B22003-07-22
US6232447B12001-05-15
US6057124A2000-05-02
US5712291A1998-01-27
US5892112A1999-04-06
US5792783A1998-08-11
EP0970070A12000-01-12
US5262564A1993-11-16
US5624677A1997-04-29
Other References:
JIANG J ET AL: "Optimal therapeutic positioning of a selective bi-steric inhibitor of MTORC1 in genetically defined cancers", EUROPEAN JOURNAL OF CANCER, ELSEVIER, AMSTERDAM NL, vol. 138, 1 October 2020 (2020-10-01), XP086321483, ISSN: 0959-8049, [retrieved on 20201026], DOI: 10.1016/S0959-8049(20)31217-X
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 2001, COLD SPRING HARBOR PRESS
PAEZ ET AL.: "EGFR Mutations In Lung Cancer Correlation With Clinical Response To Gefitinib Therapy", SCIENCE, vol. 304, no. 5676, 2004, pages 1497 - 500, XP002359959, DOI: 10.1126/science.1099314
"Handbook of Experimental Immunology", 1987, article "Gene Transfer Vectors for Mammalian Cells"
"Current Protocols in Molecular Biology", 1987, ACADEMIC PRESS, INC
PCR: THE POLYMERASE CHAIN REACTION,, 1994
CURRENT PROTOCOLS IN IMMUNOLOGY, 1991
SALTZ ET AL., PROC. AM. SOC. CLIN. ONCOL., vol. 18, 1999, pages 233a
CHEN ET AL., MOLPHARMACOL, vol. 70, 2006, pages 562
"Immunochemical Protocols", 2003
GOLDBERG ET AL., BLOOD, vol. 110, no. 1, 2007, pages 186 - 192
TERAMOTO ET AL., CANCER, vol. 77, 1996, pages 639 - 645
"Encyclopedia of Pharmaceutical Technology", 1988, MARCEL DEKKER
BLACK ET AL., NEUROLOGY, vol. 65, 2005, pages S3 - S6
PREUSSER, M ET AL., NAT. REV. NEUROL., 2015
DOUILLARD ET AL., LANCET, vol. 355, no. 9209, 2000, pages 1041 - 1047
AGNEW, CHEM. INTL. EDENGL., vol. 33, 1994, pages 183 - 186
CANCERS (BASEL, vol. 7, no. 3, September 2015 (2015-09-01), pages 1758 - 1784
ROCHE, PLOS ONE., vol. 9, no. 11, 25 November 2014 (2014-11-25)
CLIN CANCER RES., vol. 17, no. 5, 1 March 2011 (2011-03-01), pages 989 - 1000
CANCERS, vol. 7, no. 3, September 2015 (2015-09-01), pages 1758 - 1784
MODJTAHEDI ET AL., BR. J. CANCER, vol. 67, 1993, pages 247 - 253
GOLDSTEIN ET AL., CLIN. CANCER RES., vol. 1, 1995, pages 1311 - 1318
HUANG ET AL., CANCER RES., vol. 59, no. 8, 1999, pages 1236 - 1243
YAN ET AL.: "Pharmacogenetics and Pharmacogenomics In Oncology Therapeutic Antibody Development", BIOTECHNIQUES, vol. 39, no. 4, 2005, pages 565 - 8, XP001245630, DOI: 10.2144/000112043
TRAXLER ET AL., EXP. OPIN. THER.PATENTS, vol. 8, no. 12, 1998, pages 1599 - 1625
BARNETT ET AL., BIOCHEM. J., vol. 385, 2005, pages 399 - 408
JIN ET AL., BR. J. CANCER, vol. 91, 2004, pages 1808 - 12
SARKARLI, J NUTR, vol. 134, 2004, pages 3493S - 3498S
DASMAHAPATRA ET AL., CLIN. CANCER RES., vol. 10, no. 15, 2004, pages 5242 - 52
GILLSDENNIS EXPERT, OPIN. INVESTIG. DRUGS, vol. 13, 2004, pages 787 - 97
YANG ET AL., CANCER RES., vol. 64, 2004, pages 4394 - 9
SARVER ET AL., J. MED. CHEM., vol. 60, 2017, pages 113734
IGBE ET AL., ONCOTARGET, vol. 8, 2017, pages 113734
HALLIN ET AL., CANCER DISCOVERY, 28 October 2019 (2019-10-28)
CANON ET AL., NATURE, vol. 575, 2019, pages 217
THOMPSON ET AL., CLIN. CANCER RES., vol. 13, no. 6, 2007, pages 1757 - 1761
"Remington's Pharmaceutical Sciences", 1995, MACK PUBLISHING COMPANY
DOMAGALA ET AL., POL J PATHOL, vol. 3, 2012, pages 145 - 164
HANAHAN DWEINBERG RA: "The hallmarks of cancer", CELL, vol. 100, 2000, pages 57 - 70, XP055447752
GERL RVAUX DL: "Apoptosis in the development and treatment of cancer", CARCINOGENESIS, vol. 26, no. 2, 2005, pages 263 - 270, XP055715320, DOI: 10.1093/carcin/bgh283
LONG JRYAN, K: "New frontiers in promoting tumor cell death: targeting apoptosis, necroptosis and autophagy", ONCOGENE, vol. 31, 2012, pages 5045 - 5060
BOJADZICBUCHWALD, CURR TOP MED CHEM, vol. 18, 2019, pages 674 - 699
BERGE ET AL., J. PHARMACEUTICAL SCIENCES, vol. 66, no. 1-19, 1977
"Pharmaceutical Salts: Properties, Selection, and Use", 2008, WILEY-VCH
"Remington: The Science and Practice of Pharmacy", 2005, LIPPINCOTT WILLIAMS & WILKINS
Attorney, Agent or Firm:
WHITTLE, James et al. (US)
Download PDF:
Claims:
Claims

1. A method for delaying or preventing acquired resistance to a RAS inhibitor in a subj ect in need thereof, comprising administering to the subject an effective amount of a bi-steric inhibitor of mTOR, wherein the subject has already received or will receive administration of the RAS inhibitor, wherein the effective amount is an amount effective to delay or prevent acquired resistance to the RAS inhibitor in a subject in need thereof.

2. A method of treating acquired resistance to a RAS inhibitor in a subject in need thereof, comprising administering to the subject an effective amount of a bi-steric inhibitor of mTOR, wherein the effective amount is an amount effective to treat acquired resistance to the RAS inhibitor in a subject in need thereof.

3. The method of a claim 1 or claim 2, further comprising administering to the subject an effective amount of the RAS inhibitor.

4. The method of any one of claims 1-3, wherein the RAS inhibitor targets a specific RAS mutation.

5. The method of any one of claims 1-4, wherein the RAS inhibitor targets a KRAS mutation.

6. The method of any one of claims 1-5, wherein the RAS inhibitor targets the KRASG12C mutation.

7. The method of any one of claims 1-6, wherein the RAS inhibitor is a KRAS(OFF) inhibitor.

8. The method of claim 7, wherein the KRAS(OFF) inhibitor is selected from AMG 510, MRTX849, JDQ443 and MRTX1133, or a pharmaceutically acceptable salt thereof.

9. The method of any one of claims 1-8, wherein the bi-steric inhibitor of mTOR is RM- 006, also known as RMC-6272, or RMC-5552, or a pharmaceutically acceptable salt thereof.

10. The method of any one of claims 1-9, wherein the bi-steric inhibitor of mTOR is a compound having the formula or a stereoisomer or tautomer thereof.

11. The method of any one of claims 1-6 or 9-10, wherein the RAS inhibitor is a KRAS(ON) inhibitor.

12. The method of claim 11, wherein the KRAS(ON) inhibitor is a KRASG12C(ON) inhibitor.

13. The method of any one of claims 1-12, wherein the subject is administered the RAS inhibitor to treat or prevent a cancer.

14. The method of 13, wherein the cancer comprises a KRASG12C mutation.

15. The method of claim 13 or 14, wherein the cancer comprises co-occurring KRASG12C and

STK11 mutations.

16. The method of any one of claims 13-15, wherein the cancer is a Non-Small Cell Lung Cancer (NSCLC) or a colorectal cancer.

17. The method of any one of claims 13-16, wherein the cancer comprises co-occurring KRASG12C and PIK3CAE545K mutations.

18. The method of any one of claims 13-17, wherein the cancer is a colorectal cancer.

19. The method of any one of claims 1-18, wherein the method results in tumor regression.

20. The method of any one of claims 1-18, wherein the method results in tumor apoptosis.

21. A method of treating a subject having a cancer comprising administering to the subject an effective amount of a bi-steric inhibitor of mTOR in combination with a RAS inhibitor.

22. The method of claim 21, wherein the RAS inhibitor targets a specific RAS mutation.

23. The method of claim 21 or 22, wherein the RAS inhibitor targets a KRAS mutation.

24. The method of any one of claims 21-23, wherein the RAS inhibitor targets the KRASG12C mutation.

25. The method of any one of claims 21-24, wherein the RAS inhibitor is a KRAS(OFF) inhibitor.

26. The method of claim 25, wherein the KRAS(OFF) inhibitor is selected from AMG 510, MRTX849, JDQ443 and MRTX1133, or a pharmaceutically acceptable salt thereof.

27. The method of any one of claims 21-26, wherein the bi-steric inhibitor of mTOR is RM- 006, also known as RMC-6272, or RMC-5552, or a pharmaceutically acceptable salt thereof.

28. The method of any one of claims 21-26, wherein the bi-steric inhibitor of mTOR is a compound having the formula or a stereoisomer or tautomer thereof.

29. The method of any one of claims 21-24, 27 or 28, wherein the RAS inhibitor is a KRAS(ON) inhibitor.

30. The method of claim 29, wherein the KRAS(ON) inhibitor is a KRASG12C(ON) inhibitor.

31. The method of any one of claims 21-30, wherein the cancer comprises a KRASG12C mutation.

32. The method of any one of claims 21-31, wherein the cancer comprises co-occurring KRASG12C and STK11 mutations.

33. The method of any one of claims 21-32, wherein the cancer is a Non-Small Cell Lung Cancer (NSCLC).

34. The method of any one of claims 21-33, wherein the cancer comprises co-occurring KRASG12C and PIK3CAE545K mutations.

35. The method of any one of claims 21-32 or 34, wherein the cancer is a colorectal cancer.

36. The method of any one of claims 21-35, wherein the method results in tumor regression.

37. The method of any one of claims 21-36, wherein the method results in tumor apoptosis.

38. A method of inducing apoptosis of a tumor cell comprising contacting the tumor cell with an effective amount of a bi-steric inhibitor of mTOR in combination with a RAS inhibitor, wherein the effective amount is an amount effective to induce apoptosis of the tumor cell.

39. The method of any one of claims 1-38, wherein the method results in an improved lifespan for the subject as compared to the lifespan of a similar subject that has not received a treatment with the RAS inhibitor and the bi-steric mTOR inhibitor.

Description:
METHODS FOR DELAYING, PREVENTING, AND TREATING ACQUIRED RESISTANCE TO RAS INHIBITORS

CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This application claims the benefit of U.S. Provisional Application No. 63/041,071, filed June 18, 2020 and U.S. Provisional Application No. 63/062,973, filed August 7, 2020 and U.S. Provisional Application No. 63/117,417, filed November 23, 2020, and U.S. Provisional Application No. 63/134,128, filed January 5, 2021 and U.S. Provisional Application No. 63/192,976, filed May 25, 2021, the contents of each of which are incorporated herein by reference in their entireties.

FIELD OF THE INVENTION

[0002] The present disclosure relates to compositions and methods for the treatment of diseases or disorders (e.g ., cancer) with bi-steric mTOR inhibitors in combination with RAS inhibitors. Specifically, in some embodiments this disclosure includes compositions and methods for delaying, preventing, or treating acquired resistance to KRAS inhibitors using bi- steric mTOR inhibitors. In some embodiments, this disclosure includes compositions and methods for inducing apoptosis of a cell (e.g., a tumor cell) by contacting the cell with a RAS inhibitor (e.g, a KRAS(OFF) inhibitor such as a KRAS(OFF) G12C inhibitor) in combination with a bi-steric mTOR inhibitor. In some particular embodiments, the present disclosure includes methods for inducing apoptosis of a cell (e.g, a tumor cell) by contacting the cell with a RAS inhibitor (e.g, a RAS(ON) inhibitor such as a KRAS(ON) G12C inhibitor) in combination with a bi-steric mTOR inhibitor.

BACKGROUND OF THE INVENTION

[0003] Cancer remains one of the most deadly threats to human health. In the U.S., cancer affects nearly 1.3 million new patients each year, and is the second leading cause of death after heart disease, accounting for approximately 1 in 4 deaths (US20170204187).

[0004] It has been well established in literature that RAS proteins (KRAS, HRAS and NRAS) play an essential role in various human cancers and are therefore appropriate targets for anticancer therapy. Dysregulation of RAS proteins by activating mutations, overexpression or upstream activation is common in human tumors, and activating mutations in RAS are found in approximately 30% of human cancer. Of the RAS proteins, KRAS is the most frequently mutated and is therefore an important target for cancer therapy. RAS oscillates between GDP- bound “off’ (“RAS(OFF)”) and GTP -bound “on” (“RAS(ON)”)states, facilitated by interplay between a GEF protein (e.g ., SOS1), which loads RAS with GTP, and a GAP protein (e.g, NF1), which hydrolyzes GTP, thereby inactivating RAS. Additionally, the SH2 domain- containing protein tyrosine phosphatase-2 (SHP2) associates with the receptor signaling apparatus and becomes active upon RTK activation, and then promotes RAS activation. Mutations in RAS proteins can lock the protein in the “on” state resulting in a constituitively active signaling pathway that leads to uncontrolled cell growth.

[0005] First-in-class covalent inhibitors of the “off’ form of KRAS G12C have demonstrated promising anti-tumor activity in cancer patients with KRAS G12C mutations, albeit not in all. Further, therapeutic inhibition of the RAS pathway, although often initially efficacious, can ultimately prove ineffective as it may lead to over-activation of RAS pathway signaling via a number of mechanisms including, e.g. , reactivation of the pathway via relief of the negative feedback machineries that naturally operate in these pathways. For example, in various cancers, MEK inhibition results in increased ErbB signaling due to its relief of MEK / ERK-mediated feedback inhibition of RTK activation. As a result, cells that were initially sensitive to such inhibitors may become resistant. Thus, a need exists for methods of effectively inhibiting RAS pathway signaling without inducing activation of resistance mechanisms, or by minimizing resistance mechanism effects.

SUMMARY OF THE INVENTION

[0006] The present disclosure relates to compositions and methods for the treatment of diseases or disorders (e.g, cancer) with bi-steric inhibitors mTOR in combination with RAS inhibitors (e.g, KRAS(OFF) inhibitors such as KRAS(OFF) G12C -selective inhibitors or KRAS(ON) inhibitors). Surprisingly, it has been found that such combinations can delay, prevent or treat acquired resistance to a RAS inhibitor. Specifically, in some embodiments this disclosure relates, in part, to compositions and methods for delaying, preventing, or treating acquired resistance to KRAS(OFF) inhibitors using bi-steric mTOR inhibitors. In some embodiments this disclosure relates to compositions and methods for delaying, preventing, or treating acquired resistance to KRAS(ON) inhibitors using bi-steric mTOR inhibitors. Moreover, it has been surprisingly found that apoptosis occurs in the presence of such combinations. Accordingly, in some embodiments, the disclosure relates to compositions and methods for inducing apoptosis of tumor cells using one or more bi-steric mTOR inhibitor in combination with one or more KRAS(OFF) inhibitor. In some embodiments, the disclosure relates to compositions and methods for inducing apoptosis of tumor cells using one or more bi-steric mTOR inhibitor in combination with one or more KRAS(ON) inhibitor.

[0007] In some embodiments, the present disclosure includes a method for delaying or preventing acquired resistance to a RAS inhibitor in a subject, comprising administering to the subject an effective amount of a bi-steric inhibitor of mTOR, wherein the subject has already received or will receive administration of the RAS inhibitor. In some embodiments, the RAS is selected from KRAS, NRAS, and HRAS. In some embodiments, the method further comprises administering to the subject an effective amount of the RAS inhibitor. In some embodiments, the RAS inhibitor targets a specific RAS mutation. In some embodiments, the RAS inhibitor targets a KRAS mutation. In some embodiments, the RAS inhibitor targets a G12C mutation. In some embodiments, the RAS inhibitor targets the KRAS G12C mutation. In some embodiments, the RAS inhibitor binds the RAS in its “off’ position. In some embodiments, the RAS inhibitor binds the RAS in its “on” position. In some embodiments, the RAS inhibitor is a KRAS(OFF) inhibitor. In some embodiments, the RAS inhibitor is a KRAS(ON) inhibitor. In some embodiments, the RAS inhibitor is selected from the inhibitors disclosed in any one of Appendices A-1, B-1, and C-1, or a RAS inhibitor of WO 2020132597 (wherein WO 2020132597 is incorporated by reference in its entirety), or a combination of two or more of such inhibitors. In some embodiments, the RAS inhibitor targets a KRAS mutation selected from a KRAS G12A mutation, a KRAS G12D mutation, a KRAS G12F mutation, a KRAS G12I mutation, a KRAS G12L mutation, a KRAS G12R mutation, a KRAS G12S mutation, a KRAS G12V mutation, and a KRAS G12Y mutation. In some embodiments, the KRAS inhibitor is selected from AMG 510, MRTX849, JDQ443 and MRTX1133. In some embodiments, the KRAS inhibitor is selected from AMG 510 and MRTX849. In some embodiments, the KRAS inhibitor is AMG 510. In some embodiments, the KRAS inhibitor is MRTX849. In some embodiments, the inhibitor of mTOR is RM-006, also known as RMC-6272, or RMC-5552. In some embodiments, the subject is administered the RAS inhibitor to treat or prevent a cancer. In some embodiments, the cancer is a G12C cancer. In some embodiments, the cancer comprises a KRAS G12C mutation. In some embodiments, the cancer comprises co-occurring KRAS G12C and STK11 mutations. In some embodiments, the cancer is a Non-Small Cell Lung Cancer (NSCLC). In some embodiments, the cancer is a colorectal cancer. In some embodiments, the cancer is selected from pancreatic cancer, colorectal cancer, non-small cell lung cancer, squamous cell lung carcinoma, thyroid gland adenocarcinoma, and a hematological cancer (e.g ., blood (myeloid leukemia (acute and chronic), acute lymphoblastic leukemia, chronic lymphocytic leukemia); myeloproliferative diseases (e g., myelofibrosis and myeloproliferative neoplasms); multiple myeloma; myelodysplastic syndromes). In some embodiments, the cancer comprises co-occurring KRAS G12C and PIK3CA E545K mutations. In some embodiments, the cancer is a colorectal cancer. In some embodiments, the method results in tumor regression. In some embodiments, the method results in tumor apoptosis.

[0008] In some embodiments, the present disclosure includes a method of treating acquired resistance to a RAS inhibitor in a subject, comprising administering to the subject an effective amount of a bi-steric inhibitor of mTOR. In some embodiments, the RAS is selected from KRAS, NRAS, and HRAS. In some embodiments, the method further comprises administering to the subject an effective amount of the RAS inhibitor. In some embodiments, the RAS inhibitor targets a specific RAS mutation. In some embodiments, the RAS inhibitor targets a KRAS mutation. In some embodiments, the RAS inhibitor targets a G12C mutation. In some embodiments, the RAS inhibitor targets the KRAS G12C mutation. In some embodiments, the RAS inhibitor binds the RAS in its “off’ position. In some embodiments, the RAS inhibitor binds the RAS in its “on” position. In some embodiments, the RAS inhibitor is a KRAS(OFF) inhibitor. In some embodiments, the RAS inhibitor is a KRAS(ON) inhibitor. In some embodiments, the RAS inhibitor is selected from the inhibitors disclosed in any one of Appendices A-1, B-1, and C-1, or a RAS inhibitor of WO 2020132597 (wherein WO 2020132597 is incorporated by reference in its entirety), or a combination of two or more of such inhibitors. In some embodiments, the RAS inhibitor targets a KRAS mutation selected from a KRAS G12A mutation, a KRAS G12D mutation, a KRAS G12F mutation, a KRAS G12I mutation, a KRAS G12L mutation, a KRAS G12R mutation, a KRAS G12S mutation, a KRAS G12V mutation, and a KRAS G12Y mutation. In some embodiments, the KRAS inhibitor is selected from AMG 510, MRTX849, JDQ443 and MRTX1133. In some embodiments, the KRAS inhibitor is selected from AMG 510 and MRTX849. In some embodiments, the KRAS inhibitor is AMG 510. In some embodiments, the KRAS inhibitor is MRTX849. In some embodiments, the inhibitor of mTOR is RM-006, also known as RMC-6272, or RMC-5552. In some embodiments, the subject is administered the RAS inhibitor to treat or prevent a cancer. In some embodiments, the cancer is a G12C cancer. In some embodiments, the cancer comprises a KRAS G12C mutation. In some embodiments, the cancer comprises co-occurring KRAS G12C and STK11 mutations. In some embodiments, the cancer is a Non-Small Cell Lung Cancer (NSCLC). In some embodiments, the cancer is a colorectal cancer. In some embodiments, the cancer is selected from pancreatic cancer, colorectal cancer, non-small cell lung cancer, squamous cell lung carcinoma, thyroid gland adenocarcinoma, and a hematological cancer (e.g, blood (myeloid leukemia (acute and chronic), acute lymphoblastic leukemia, chronic lymphocytic leukemia); myeloproliferative diseases (e.g., myelofibrosis and myeloproliferative neoplasms); multiple myeloma; myelodysplastic syndromes). In some embodiments, the cancer comprises co-occurring KRAS G12C and PIK3CA E545K mutations. In some embodiments, the cancer is a colorectal cancer. In some embodiments, the method results in tumor regression. In some embodiments, the method results in tumor apoptosis.

[0009] In some embodiments, the present disclosure includes a method of treating a subj ect having a cancer comprising administering to the subject a bi-steric inhibitor of mTOR in combination with a RAS inhibitor. In some embodiments, the RAS is selected from KRAS, NRAS, and HRAS. In some embodiments, the RAS inhibitor targets a specific RAS mutation. In some embodiments, the RAS inhibitor targets a KRAS mutation. In some embodiments, the RAS inhibitor targets a G12C mutation. In some embodiments, the RAS inhibitor targets the KRAS G12C mutation. In some embodiments, the RAS inhibitor binds the RAS in its “off’ position. In some embodiments, the RAS inhibitor is a KRAS(OFF) inhibitor. In some embodiments, the RAS inhibitor is a KRAS(ON) inhibitor. In some embodiments, the RAS inhibitor is selected from the inhibitors disclosed in any one of Appendices A-1, B-1, and C-1, or a RAS inhibitor of WO 2020132597 (wherein WO 2020132597 is incorporated by reference in its entirety), or a combination of two or more of such inhibitors. In some embodiments, the KRAS inhibitor targets a KRAS mutation selected from a KRAS G12A mutation, a KRAS G12D mutation, a KRAS G12F mutation, a KRAS G12I mutation, a KRAS G12L mutation, a KRAS G12R mutation, a KRAS G12S mutation, a KRAS G12V mutation, and a KRAS G12Y mutation. In some embodiments, the KRAS inhibitor is selected from AMG 510, MRTX849, JDQ443 and MRTX1133. In some embodiments, the KRAS inhibitor is selected from AMG 510 and MRTX849. In some embodiments, the KRAS inhibitor is AMG 510. In some embodiments, the KRAS inhibitor is MRTX849. In some embodiments, the bi-steric inhibitor of mTOR is RM-006, also known as RMC-6272, or RMC-5552. In some embodiments, the cancer is a G12C cancer. In some embodiments, the cancer comprises a KRAS G12C mutation. In some embodiments, the cancer comprises co-occurring KRAS G12C and STK11 mutations. In some embodiments, the cancer is a Non-Small Cell Lung Cancer (NSCLC). In some embodiments, the cancer is a colorectal cancer. In some embodiments, the cancer is selected from pancreatic cancer, colorectal cancer, non-small cell lung cancer, squamous cell lung carcinoma, thyroid gland adenocarcinoma, and a hematological cancer (e.g, blood (myeloid leukemia (acute and chronic), acute lymphoblastic leukemia, chronic lymphocytic leukemia); myeloproliferative diseases (e.g., myelofibrosis and myeloproliferative neoplasms); multiple myeloma; myelodysplastic syndromes). In some embodiments, the cancer comprises co-occurring KRAS G12C and PIK3CA E545K mutations. In some embodiments, the cancer is a colorectal cancer. In some embodiments, the method results in tumor regression. In some embodiments, the method results in tumor apoptosis.

[0010] In some embodiments, the present disclosure includes a method of inducing apoptosis of a tumor cell comprising contacting the tumor cell with a bi-steric inhibitor of mTOR in combination with a RAS inhibitor. In some embodiments, the RAS is selected from KRAS, NRAS, and HRAS. In some embodiments, the RAS inhibitor targets a specific RAS mutation. In some embodiments, the RAS inhibitor targets a KRAS mutation. In some embodiments, the RAS inhibitor targets a G12C mutation. In some embodiments, the RAS inhibitor targets the KRAS G12C mutation. In some embodiments, the RAS inhibitor binds the RAS in its “off’ position. In some embodiments, the RAS inhibitor is a KRAS(OFF) inhibitor. In some embodiments, the RAS inhibitor is a KRAS(ON) inhibitor. In some embodiments, the RAS inhibitor is selected from the inhibitors disclosed in any one of Appendices A-1, B-1, and C-1, or a RAS inhibitor of WO 2020132597 (wherein WO 2020132597 is incorporated by reference in its entirety), or a combination of two or more of such inhibitors. In some embodiments, the KRAS inhibitor targets a KRAS mutation selected from a KRAS G12A mutation, a KRAS G12D mutation, a KRAS G12F mutation, a KRAS G12I mutation, a KRAS G12L mutation, a KRAS G12R mutation, a KRAS G12S mutation, a KRAS G12V mutation, and a KRAS G12Y mutation. In some embodiments, the KRAS inhibitor is selected from AMG 510, MRTX849, JDQ443 and MRTX1133. In some embodiments, the KRAS inhibitor is selected from AMG 510 and MRTX849. In some embodiments, the KRAS inhibitor is AMG 510. In some embodiments, the KRAS inhibitor is MRTX849. In some embodiments, the inhibitor of mTOR is RM-006, also known as RMC-6272, or RMC-5552. In some embodiments, the tumor is caused by a cancer. In some embodiments, the cancer is a G12C cancer. In some embodiments, the cancer comprises a KRAS G12C mutation. In some embodiments, the cancer comprises co-occurring KRAS G12C and STK11 mutations. In some embodiments, the cancer is a Non-Small Cell Lung Cancer (NSCLC). In some embodiments, the cancer is a colorectal cancer. In some embodiments, the cancer is selected from pancreatic cancer, colorectal cancer, non-small cell lung cancer, squamous cell lung carcinoma, thyroid gland adenocarcinoma, and a hematological cancer (e.g., blood (myeloid leukemia (acute and chronic), acute lymphoblastic leukemia, chronic lymphocytic leukemia); myeloproliferative diseases (e g., myelofibrosis and myeloproliferative neoplasms); multiple myeloma; myelodysplastic syndromes). In some embodiments, the cancer comprises co-occurring KRAS G12C and PIK3CA E545K mutations. In some embodiments, the cancer is a colorectal cancer. In some embodiments, the method results in tumor regression. In some embodiments, the method results in tumor apoptosis. In some embodiments, the method results in an improved lifespan for the subject as compared to the lifespan of a similar subject that has not received a treatment with the RAS inhibitor and the bi-steric mTOR inhibitor.

BRIEF DESCRIPTION OF THE DRAWINGS

[0011] Figure 1 shows the combinatorial anti-proliferative activity of RM-006 (also known as RMC-6272) and the KRAS G12C (OFF) inhibitor AMG 510 in the NSCLC cells lines NCI- H2122 and NCI-H2030, which each have with RAS and mTOR signaling co-activation. FIG. 1A shows the anti-proliferative activity that resulted from varying concentrations of AMG 510 in presence of constant RM-006 (also known as RMC-6272) (3 nM in H2122, left panel, and 10 nM in H2030, right panel). FIG. 1B shows the anti-proliferative activity that resulted from varying concentrations of RM-006 (also known as RMC-6272) in presence of constant AMG 510 (90 nM in H2122, left panel, or 10 nM in H2030, right panel).

[0012] Figure 2 shows that RM-006 (also known as RMC-6272) enhances the in vivo anti- tumor activity of a KRAS G12C (OFF) inhibitor, and the combination of these compounds delays tumor regrowth. FIG. 2A shows a tumor volume plot demonstrating the combinatorial effects of RM-006 (also known as RMC-6272) with AMG 510 on in vivo tumor growth in the human non-small cell lung cancer NCI-H358 KRAS G12C xenograft model. FIG. 2B shows a waterfall plot presenting the end of study responses of each mouse tested in FIG.2A. FIG. 2C shows a tumor volume plot demonstrating the combinatorial effects of RM-006 (also known as RMC- 6272) with AMG 510 on in vivo tumor growth delay following treatment cessation. FIG. 2D shows Kaplan-Meier analysis demonstrating a significant delay in tumor regrowth back to 500 mm3 after treatment cessation caused by the combination of AMG 510 with RM-006 (also known as RMC-6272) as compared to single-agent AMG 510, and as assessed by Log-rank (Mantel-Cox) test with p = 0.0395.

[0013] Figure 3 shows that the combination of RM-006 (also known as RMC-6272) and KRAS G12C (OFF) inhibition drives tumor regression in the NCI-H2122 NSCLC model, which has co-activation of RAS and mTOR signaling. FIG. 3A shows a tumor volume plot demonstrating the in vivo tumor growth inhibition induced by RM-006 (also known as RMC- 6272) and AMG 510 alone or in combination in the NCI-H2122 NSCLC CDX model. *** = p<0.001, assessed by an ordinary one-way ANOVA of tumor volumes along with multiple comparisons via a post-hoc Tukey’s test in GraphPad Prism software. FIG. 3B shows a waterfall plot demonstrating individual tumor responses at the end of the study.

[0014] Figure 4 shows results of a single-dose PKPD study using NCI-H2122 NSCLC CDX. Pathway modulation was assessed by quantitative image analyses of IHC staining of tumor sections for pS6RP (S235) (FIG. 4A); p4EBP1 (FIG. 4B); pERK (FIG. 4C); and by qPCR assay for human DUSP6 (FIG. 4D). FIG. 4E shows representative IHC staining images for pS6RP and FIG. 4F shows representative IHC staining images for p4EBP1.

[0015] Figure 5 shows synergistic in vivo induction of apoptosis in human non-small cell lung cancer NCI-H2122 KRAS G12C ; STK11del tumors induced by a single dose of the RM- 006 (also known as RMC-6272) in combination with AMG 510. FIG. 5A shows quantification of IHC staining for cleaved caspase 3 (CC3). FIG. 5B shows representative CC3 staining 24 (top row images) and 48 hrs. (bottom row images) post-treatment with the indicated amounts of RM-006 (also known as RMC-6272) and AMG 510 alone and in combination.

[0016] Figure 6 shows that the combination of RM-006 (also known as RMC-6272) and a KRAS G12C (OFF) inhibitor significantly delays on-treatment resistance in a NSCLC model with RAS and mTOR signaling co-activation. FIG. 6A shows a mean tumor volume plot demonstrating significant delay in on-treatment resistance induced by co-treatment with RM- 006 (also known as RMC-6272) and AMG 510 as compared to single agent treatment. FIG. 6B shows Kaplan-Meier analysis of tumors reaching baseline volume while on treatment, and the results demonstrate the combination significantly prolonged the time for tumors to develop resistance, as assessed by Log-rank (Mantel-Cox) test.

[0017] Figure 7 shows tumor volume plots of four mice demonstrating attenuation of AMG 510 xenograft tumor resistance in the NCI-H2030 model by treatment with RM-006 (also known as RMC-6272). N=4.

[0018] Figure 8 shows combinatorial activity of RM-006 (also known as RMC-6272) and a KRAS G12C (OFF) inhibitor in the ST3235 (KRAS G12C PIK3CA E545K ) CRC PDX model.

[0019] Figure 9 shows combinatorial activity of RM-006 (also known as RMC-6272) and a RAS(ON) inhibitor of the disclosure, Compound A, on tumor cell growth in vivo were evaluated in the human lung cancer ST 1989 KRAS G12C patient-derived xenograft model using female athymic nude mice (6-12 weeks old).

[0020] Figure 10 shows combinatorial effects of RMC-6272 (also known as RMC-006) with Compound B in a NSCLC CDX Model.

[0021] Figure 11 shows combinatorial effects of RMC-5552 with Compound B in a NSCLC CDX Model.

DETAILED DESCRIPTION OF THE INVENTION

[0022] The details of the invention are set forth in the accompanying description below. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, illustrative methods and materials are now described. Other features, objects, and advantages of the invention will be apparent from the description and from the claims. In the specification and the appended claims, the singular forms also include the plural unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All patents and publications cited in this specification are incorporated herein by reference in their entireties.

General Methods

[0023] The practice of the present invention will employ, unless otherwise indicated, conventional techniques of cell culturing, molecular biology (including recombinant techniques), microbiology, cell biology, biochemistry and immunology, which are within the skill of the art. Such techniques are explained fully in the literature, such as, Molecular Cloning: A Laboratory Manual , third edition (Sambrook et al., 2001) Cold Spring Harbor Press; Oligonucleotide Synthesis (P. Herdewijn, ed., 2004); Animal Cell Culture (R. I. Freshney), ed., 1987); Methods in Enzymology (Academic Press, Inc.); Handbook of Experimental Immunology (D. M. Weir & C. C. Blackwell, eds.); Gene Transfer Vectors for Mammalian Cells (J. M. Miller & M. P. Calos, eds., 1987); Current Protocols in Molecular Biology (F. M. Ausubel et al., eds., 1987); PCR: The Polymerase Chain Reaction, (Mullis et al., eds., 1994); Current Protocols in Immunology (J. E. Coligan et al., eds., 1991); Short Protocols in Molecular Biology (Wiley and Sons, 1999); Manual of Clinical Laboratory Immunology (B. Detrick, N. R. Rose, and J. D. Folds eds., 2006); Immunochemical Protocols

(J. Pound, ed., 2003); Lab Manual in Biochemistry: Immunology and Biotechnology (A. Nigam and A. Ayyagari, eds. 2007); Immunology Methods Manual: The Comprehensive Sourcebook of Techniques (Ivan Lefkovits, ed., 1996); Using Antibodies: A Laboratory Manual (E. Harlow and D. Lane, eds., 1988); and others.

Definitions

[0024] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by those of ordinary skill in the art to which the invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, preferred methods and materials are described. For the purposes of the present invention, the following terms are defined below.

[0025] The articles “a” and “an” are used in this disclosure to refer to one or more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element.

[0026] The term “or” is used to mean "and/or" unless explicitly indicated to refer to alternatives only or the alternative are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and "and/or”. The term “and/or” is used in this disclosure to mean either “and” or “or” unless indicated otherwise.

[0027] Throughout this specification, unless the context requires otherwise, the words “comprise,” “comprises,” and “comprising” will be understood to imply the inclusion of a stated step or element or group of steps or elements but not the exclusion of any other step or element or group of steps or elements. By “consisting of’ is meant including, and limited to, whatever follows the phrase “consisting of.” Thus, the phrase “consisting of’ indicates that the listed elements are required or mandatory, and that no other elements may be present. By “consisting essentially of’ is meant including any elements listed after the phrase, and limited to other elements that do not interfere with or contribute to the activity or action specified in the disclosure for the listed elements. Thus, the phrase “consisting essentially of’ indicates that the listed elements are required or mandatory, but that other elements are optional and may or may not be present depending upon whether or not they materially affect the activity or action of the listed elements.

[0028] The term “e.g.” is used herein to mean “for example,” and will be understood to imply the inclusion of a stated step or element or group of steps or elements but not the exclusion of any other step or element or group of steps or elements. [0029] By “optional” or “optionally,” it is meant that the subsequently described event or circumstance may or may not occur, and that the description includes instances where the event or circumstance occurs and instances in which it does not. For example, “optionally substituted aryl” encompasses both “aryl” and “substituted aryl” as defined herein. It will be understood by those ordinarily skilled in the art, with respect to any group containing one or more substituents, that such groups are not intended to introduce any substitution or substitution patterns that are sterically impractical, synthetically non-feasible, and/or inherently unstable.

[0030] The term “administer”, “administering”, or “administration” as used in this disclosure refers to either directly administering a disclosed compound or pharmaceutically acceptable salt of the disclosed compound or a composition to a subject, or administering a prodrug derivative or analog of the compound or pharmaceutically acceptable salt of the compound or composition to the subject, which can form an equivalent amount of active compound within the subject’s body.

[0031] The terms “bi-steric mTOR inhibitor” and “bi-steric inhibitor of mTOR” are used interchangeably in this disclosure to refer to two pharmacophores in a single compound. One pharmacophore binds to the well-known FRB (FKBP12-rapamycin binding) site on mTORC1 and the other binds to the mTOR kinase active site. As a result of these two binding interactions, such compounds exhibit two biologically useful features: (1) selectivity for mTORC1 over mTORC2, which is characteristic of the natural compound rapamycin, and (2) deep inhibition of mTORC1 , which is characteristic of known active site inhibitors. These properties enable selective inhibition of phosphorylation of mTORC1 substrates, including 4EBP1. In some embodiments, a bi-steric mTOR inhibitor has a molecular weight of between 1600 and 2100 Da, inclusive, and exhibits selective (> 10-fold) inhibition of mTORC1 over mTORC2.

[0032] The term “carrier”, as used in this disclosure, encompasses excipients, and diluents and means a material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting a pharmaceutical agent from one organ, or portion of the body, to another organ, or portion of the body of a subject.

[0033] The term “combination therapy” refers to a method of treatment comprising administering to a subject at least two therapeutic agents, optionally as one or more pharmaceutical compositions. For example, a combination therapy may comprise administration of a single pharmaceutical composition comprising at least two therapeutic agents and one or more pharmaceutically acceptable carrier, excipient, diluent, and/or surfactant. A combination therapy may comprise administration of two or more pharmaceutical compositions, each composition comprising one or more therapeutic agent and one or more pharmaceutically acceptable carrier, excipient, diluent, and/or surfactant. In various embodiments, at least one of the therapeutic agents is a bi-steric mTOR inhibitor (e.g., any one or more such bi-steric mTOR inhibitor disclosed herein or known in the art). In various embodiments, at least one of the therapeutic agents is a KRAS(OFF) inhibitor (e.g, any one or more KRAS(OFF) inhibitor disclosed herein or known in the art). In some particular embodiments, at least one of the therapeutic agents is a KRAS G12C inhibitor (e.g, any one or more of the KRAS G12C inhibitors disclosed herein or known in the art). In some particular embodiments, at least one of the therapeutic agents is AMG 510, MRTX849, JDQ443or MRTX1133. In some embodiments, the at least one of the therapeutic agents is selected from AMG 510 and MRTX849. In some embodiments, the therapeutic agent is AMG 510. In some embodiments, the therapeutic agent is MRTX849. In various embodiments, at least one of the therapeutic agents is a bi-steric mTOR inhibitor and one of the therapeutic agents is a KRAS G12C inhibitor. The two agents may optionally be administered simultaneously (as a single or as separate compositions) or sequentially (as separate compositions). The therapeutic agents may be administered in an effective amount. The therapeutic agent may be administered in a therapeutically effective amount. In some embodiments, the effective amount of one or more of the therapeutic agents may be lower when used in a combination therapy than the therapeutic amount of the same therapeutic agent when it is used as a monotherapy, e.g, due an additive or synergistic effect of combining the two or more therapeutics.

[0034] The term “disorder” is used in this disclosure to mean, and is used interchangeably with, the terms disease, condition, or illness, unless otherwise indicated.

[0035] An “effective amount” when used in connection with a compound is an amount effective for treating or preventing a disease or disorder in a subject as described herein.

[0036] The term “inhibitor” means a compound that prevents a biomolecule, (e.g, a protein, nucleic acid) from completing or initiating a reaction. An inhibitor can inhibit a reaction by competitive, uncompetitive, or non-competitive means. Exemplary inhibitors include, but are not limited to, nucleic acids, DNA, RNA, shRNA, siRNA, proteins, protein mimetics, peptides, peptidomimetics, antibodies, small molecules, chemicals, analogs that mimic the binding site of an enzyme, receptor, or other protein, e.g, that is involved in signal transduction, therapeutic agents, pharmaceutical compositions, drugs, and combinations of these. In some embodiments, the inhibitor can be nucleic acid molecules including, but not limited to, siRNA that reduce the amount of functional protein in a cell. Accordingly, compounds said to be “capable of inhibiting” a particular protein, e.g ., mTOR or RAS, comprise any such inhibitor.

[0037] As used herein, the term “RAS(OFF) inhibitor” refers to an inhibitor that targets, that is, selectively binds to or inhibits the GDP -bound, inactive state of RAS (e.g., selective over the GTP -bound, active state of RAS). Inhibition of the GDP -bound, inactive state of RAS includes, for example, sequestering the inactive state by inhibiting the exchange of GDP for GTP, thereby inhibiting RAS from adopting the active conformation. In certain embodiments, RAS(OFF) inhibitors may also bind to or inhibit the GTP -bound, active state of RAS (e.g., with a lower affinity or inhibition constant than for the GDP-bound, inactive state of RAS). In some embodiments, a RAS(OFF) inhibitor has a molecular weight of under 700 Da. The term “KRAS(OFF) inhibitor” refers to any inhibitor that binds to KRAS in its GDP -bound “OFF” position. Reference to the term KRAS(OFF) inhibitor includes, for example, AMG 510, MRTX849, JDQ443 and MRTX1133. In some embodiments, the KRAS(OFF) inhibitor is selected from AMG 510 and MRTX849. In some embodiments, the KRAS(OFF) inhibitor is AMG 510. In some embodiments, the KRAS(OFF) inhibitor is MRTX849. In some embodiments, the KRAS(OFF) inhibitor is selected from BPI-421286, JNJ-74699157 (ARS- 3248), LY3537982, MRTX1257, ARS853, ARS1620, or GDC-6036. In some embodiments, reference to the term KRAS(OFF) inhibitor includes any such KRAS(OFF) inhibitor disclosed in any one of the following patent applications: WO 2021113595, WO 2021107160, WO 2021106231, WO 2021088458, WO 2021086833, WO 2021085653, WO 2021081212, WO 2021058018, WO 2021057832, WO 2021055728, WO 2021031952, WO 2021027911, WO 2021023247, WO 2020259513, WO 2020259432, WO 2020234103, WO 2020233592, WO 2020216190, WO 2020178282, WO 2020146613, WO 2020118066, WO 2020113071, WO 2020106647, WO 2020102730, WO 2020101736, WO 2020097537, WO 2020086739, WO 2020081282, WO 2020050890, WO 2020047192, WO 2020035031, WO 2020028706, WO 2019241157, WO 2019232419, WO 2019217691, WO 2019217307, WO 2019215203, WO 2019213526, WO 2019213516, WO 2019155399, WO 2019150305, WO 2019110751, WO 2019099524, WO 2019051291, WO 2018218070, WO 2018218071, WO 2018218069, WO 2018217651, WO 2018206539, WO 2018143315, WO 2018140600, WO 2018140599, WO 2018140598, WO 2018140514, WO 2018140513, WO 2018140512, WO 2018119183, WO 2018112420, WO 2018068017, WO 2018064510, WO 2017201161, WO 2017172979, WO 2017100546, WO 2017087528, WO 2017058807, WO 2017058805, WO 2017058728, WO 2017058902, WO 2017058792, WO 2017058768, WO 2017058915, WO 2017015562, WO 2016168540, WO 2016164675, WO 2016049568, WO 2016049524, WO 2015054572, WO 2014152588, WO 2014143659 and WO 2013155223,, each of which are incorporated herein by reference in its entirety. Reference to “AMG 510” and “MRTX849” herein means the following compounds:

AMG 510

MRTX849

[0038] As used herein, the term “RAS(ON) inhibitor” refers to an inhibitor that targets, that is, selectively binds to or inhibits, the GTP -bound, active state of RAS (e.g., selective over the GDP -bound, inactive state of RAS). Inhibition of the GTP -bound, active state of RAS includes, for example, the inhibition of oncogenic signaling from the GTP -bound, active state of RAS. In some embodiments, the RAS(ON) inhibitor is an inhibitor that selectively binds to and inhibits the GTP-bound, active state of RAS. In certain embodiments, RAS(ON) inhibitors may also bind to or inhibit the GDP -bound, inactive state of RAS (e.g., with a lower affinity or inhibition constant than for the GTP-bound, active state of RAS). In some embodiments, a RAS(ON) inhibitor has a molecular weight of between 800 and 1100 Da, inclusive. The term “KRAS(ON) inhibitor” refers to any inhibitor that binds to KRAS in its GDP-bound “ON” position. Reference to the term KRAS(ON) inhibitor includes, without limitation, any one or more KRAS(ON) inhibitor selected from the KRAS(ON) inhibitors disclosed in Appendix A- 1, Appendix B-1, and Appendix C-1, or a RAS inhibitor of WO 2020132597 (wherein WO 2020132597 is incorporated by reference in its entirety), or a combination of any such KRAS(ON) inhibitors.

[0039] As used herein, “Compound A” and “Compound B” are each distinct KRAS G12C (ON) inhibitors disclosed in Appendix B-1, and encompass pharmaceutically acceptable salts thereof unless otherwise explicitly indicated otherwise.

[0040] The term “monotherapy” refers to a method of treatment comprising administering to a subject a single therapeutic agent, optionally as a pharmaceutical composition. For example, a monotherapy may comprise administration of a pharmaceutical composition comprising a therapeutic agent and one or more pharmaceutically acceptable carrier, excipient, diluent, and/or surfactant. The therapeutic agent may be administered in an effective amount. The therapeutic agent may be administered in a therapeutically effective amount.

[0041] The term “mutation” as used herein indicates any modification of a nucleic acid and/or polypeptide which results in an altered nucleic acid or polypeptide. The term “mutation” may include, for example, point mutations, deletions or insertions of single or multiple residues in a polynucleotide, which includes alterations arising within a protein-encoding region of a gene as well as alterations in regions outside of a protein-encoding sequence, such as, but not limited to, regulatory or promoter sequences, as well as amplifications and/or chromosomal breaks or translocations.

[0042] A “patient” or “subject” is a mammal, e.g ., a human, mouse, rat, guinea pig, dog, cat, horse, cow, pig, or non-human primate, such as a monkey, chimpanzee, baboon or rhesus.

[0043] The term “prevent” or “preventing” with regard to a subject refers to keeping a disease or disorder from afflicting the subject. Preventing includes prophylactic treatment. For instance, preventing can include administering to the subject a compound disclosed herein before a subject is afflicted with a disease and the administration will keep the subject from being afflicted with the disease.

[0044] The term “preventing acquired resistance,” as used herein, means avoiding the occurrence of acquired or adaptive resistance. Thus, e.g. , the use of a bi-steric mTOR inhibitor described herein in preventing acquired/adaptive resistance to a KRAS G12C inhibitor means that the bi-steric mTOR inhibitor is administered prior to any detectable existence of resistance to the KRAS G12C inhibitor and the result of such administration of the bi-steric mTOR inhibitor is that no resistance to the KRAS G12C inhibitor occurs.

[0045] The term “providing to a/the subject” a therapeutic agent, e.g ., a bi-steric mTOR inhibitor, includes administering such an agent.

[0046] The terms “RAS inhibitor” and “inhibitor of [a] RAS” are used interchangeably to refer to any inhibitor that targets a RAS protein. In various embodiments, these terms include RAS(OFF) and RAS(ON) inhibitors such as, e.g., the KRAS(OFF) and KRAS(ON) inhibitors disclosed herein. The term “RAS(OFF) inhibitor” refers to any inhibitor that binds to a RAS protein in its GDP -bound “OFF” position, as further defined herein. The term “RAS(ON) inhibitor” refers to any inhibitor that binds to a RAS protein in its GDP -bound “ON” position, as further defined herein. In some embodiments, a RAS inhibitor has a molecular weight of under 700 Da. In some embodiments, the RAS inhibitor is selected from the group consisting of AMG 510, MRTX1257, JNJ-74699157 (ARS-3248), LY3537982, ARS-853, ARS-1620, GDC-6036, BPI-421286, JDQ443, JAB-21000, JAB-22000, and JAB-23000. A RAS inhibitor may be a RAS vaccine, or another therapeutic modality designed to directly or indirectly decrease the oncogenic activity of RAS.

[0047] The terms “RAS pathway” and “RAS/MAPK pathway” are used interchangeably herein to refer to a signal transduction cascade downstream of various cell surface growth factor receptors in which activation of RAS (and its various isoforms and alleotypes) is a central event that drives a variety of cellular effector events that determine the proliferation, activation, differentiation, mobilization, and other functional properties of the cell. SHP2 conveys positive signals from growth factor receptors to the RAS activation/deactivation cycle, which is modulated by guanine nucleotide exchange factors (GEFs, such as SOS1) that load GTP onto RAS to produce functionally active GTP -bound RAS as well as GTP-accelerating proteins (GAPs, such as NF1) that facilitate termination of the signals by conversion of GTP to GDP. GTP-bound RAS produced by this cycle conveys essential positive signals to a series of serine/threonine kinases including RAF and MAP kinases, from which emanate additional signals to various cellular effector functions.

[0048] The term RM-006 (also known as RMC-6272) refers to a bi-steric mTOR inhibitor (also termed an mTORC1 -selective inhibitor), which has the following structure:

[0049] The term RMC-5552 refers to a bi-steric mTOR inhibitor (also termed an mTORC1 -selective inhibitor), found in Appendix D-1 and in WO 2019212990, wherein WO 2019212990 is incorporated herein by reference in its entirety, which has the following structure:

[0050] Reference to a “subtype” of a cell (e.g ., a KRAS G12C subtype, a KRAS G12S subtype, a KRAS G12D subtype, a KRAS G12V subtype) means that the cell contains a gene mutation encoding a change in the protein of the type indicated. For example, a cell classified as a “KRAS G12C subtype” contains at least one KRAS allele that encodes an amino acid substitution of cysteine for glycine at position 12 ( G12C ); and, similarly, other cells of a particular subtype (e.g., KRAS G12D , KRAS G12S and KRAS G12V subtypes) contain at least one allele with the indicated mutation (e.g. , a KRAS G12D mutation, a KRAS G12S mutation or a KRAS G12V mutation, respectively). Unless otherwise noted, all amino acid position substitutions referenced herein (such as, e.g, “ G12C ” in KRAS G12C ) correspond to substitutions in the human version of the referenced protein, i.e., KRAS G12C refers to a G→C substitution in position 12 of human KRAS.

[0051] A “therapeutic agent” is any substance, e.g, a compound or composition, capable of treating a disease or disorder. In some embodiments, therapeutic agents that are useful in connection with the present disclosure include without limitation mTOR inhibitors, RAS inhibitors such as, e.g., KRAS inhibitors (e.g, KRAS G12C inhibitors), and cancer chemotherapeutics. Many such inhibitors are known in the art and are disclosed herein. [0052] The terms “therapeutically effective amount”, “therapeutic dose”, “prophylactically effective amount”, or “diagnostically effective amount” is the amount of the drug, e.g., a bi- steric mTOR inhibitor, needed to elicit the desired biological response following administration.

[0053] The term “treatment” or “treating” with regard to a subject, refers to improving at least one symptom, pathology or marker of the subject’s disease or disorder, either directly or by enhancing the effect of another treatment. Treating includes curing, improving, or at least partially ameliorating the disorder, and may include even minimal changes or improvements in one or more measurable markers of the disease or condition being treated. “Treatment” or “treating” does not necessarily indicate complete eradication or cure of the disease or condition, or associated symptoms thereof. The subject receiving this treatment is any subject in need thereof. Exemplary markers of clinical improvement will be apparent to persons skilled in the art.

Overview

[0054] The present disclosure relates to, inter alia, compositions, methods, and kits for treating or preventing a disease or disorder (e.g, cancer) with a RAS inhibitor (e.g, a KRAS G12C inhibitor) in combination with a bi-steric mTOR inhibitor. In some particular embodiments, the present disclosure includes methods for delaying, preventing, or treating acquired resistance to a RAS inhibitor (e.g, a KRAS G12C inhibitor) by administering the RAS inhibitor (e.g, a KRAS G12C inhibitor) in combination with a bi-steric mTOR inhibitor. In some particular embodiments, the present disclosure includes methods for inducing apoptosis of a cell (e.g. , a tumor cell) by contacting the cell with a RAS inhibitor (e.g. , a KRAS(OFF) inhibitor such as a KRAS G12C inhibitor) in combination with a bi-steric mTOR inhibitor. In some particular embodiments, the present disclosure includes methods for inducing apoptosis of a cell (e.g, a tumor cell) by contacting the cell with a RAS inhibitor (e.g, a RAS(ON) inhibitor such as a KRAS(ON) G12C inhibitor) in combination with a bi-steric mTOR inhibitor.

[0055] The mammalian target of rapamycin (mTOR) is a serine-threonine kinase related to the lipid kinases of the phosphoinositide 3-kinase (PI3K) family. mTOR exists in two complexes, mTORC1 and mTORC2, which are differentially regulated, have distinct substrate specificities, and are differentially sensitive to rapamycin. mTORC1 integrates signals from growth factor receptors with cellular nutritional status and controls the level of cap-dependent mRNA translation by modulating the activity of key translational components such as the cap- binding protein and oncogene eIF4E. Hyperactivation of the PBK/mTOR pathway occurs frequently in human cancer, via mutation or deletion of different components.

[0056] Various inhibitors of mTOR exist and have differential specificity for the two mTOR complexes. However, despite clear biological rationale, PBK/mTOR pathway inhibitors have been largely unsuccessful in “all-comers” clinical trials, attributed to the lack of biomarker-guided patient stratification. The present inventors have developed a class of selective mTORC1 inhibitors, termed ‘bi-steric’, which comprise a rapamycin-like core moiety covalently linked to an mTOR active-site inhibitor. Bi-steric mTORC1 inhibitors exhibit potent and selective (> 10-fold) inhibition of mTORC1 over mTORC2, durably suppress S6K and 4EBP1 phosphorylation, and induce growth suppression and apoptosis in multiple cancer cell lines. These inhibitors provide the mTORC1 selectivity of rapalogs and potently inhibit translation initiation by the 4EBP1-eIF4E axis while sparing mTORC2. In various embodiments, any one or more of these bi-steric mTOR inhibitors may utilized in any of methods disclosed herein.

[0057] Accordingly, in some embodiments, the present disclosure relates to the unexpected discovery that acquired resistance to KRAS inhibitors, and in particular KRAS G12C inhibitors, can be delayed and even arrested or reversed by coadministration of a bi-steric mTOR inhibitor (e.g., such as RM-006, also known as RMC-6272, or RMC-5552). Moreover, in some embodiments, the present disclosure relates to the unexpected discovery that the combination of KRAS inhibitors, and in particular KRAS G12C inhibitors with a bi-steric mTOR inhibitor (e.g, such as RM-006, also known as RMC-6272, or RMC-5552) results in synergistic apoptosis of tumor cells. Thus, in some embodiments, the present disclosure includes compositions, methods, and kits for the treatment of a disease or condition (e.g, a cancer or tumor) with a RAS inhibitor in combination with a bi-steric mTOR inhibitor. In particular embodiments, the RAS inhibitor targets KRAS, NRAS, or HRAS. In particular embodiments the RAS inhibitor is a RAS mutant specific inhibitor. In certain embodiments, RAS mutant is selected from:

(a) the following K-Ras mutants: G12D, G12V, G12C, G13D, G12R, G12A, Q61H, G12S, A146T, G13C, Q61L, Q61R, K117N, A146V, G12F, Q61K, L19F, Q22K, V14I, A59T, A146P, GBR, G12L, or G13V, and combinations thereof;

(b) the following H-Ras mutants: Q61R, GBR, Q61K, G12S, Q61L, G12D, G13V, G13D, G12C, K117N, A59T, G12V, G13C, Q61H, G13S, A18V, D119N, G13N, A146T, A66T, G12A, A146V, G12N, or GBR, and combinations thereof; and (c) the following N-Ras mutants: Q61R, Q61K, G12D, Q61L, Q61H, G13R, G13D,

G12S, G12C, G12V, G12A, G13V, G12R, P185S, G13C, A146T, G60E, Q61P,

A59D, E132K, E49K, T50I, A146V, or A59T, and combinations thereof.

Mutations at these positions may result in RAS-driven tumors. In some particular embodiments, the RAS inhibitor is a KRAS(OFF) inhibitor known in the art or disclosed herein. The KRAS(OFF) inhibitor may be any one or more of the KRAS(OFF) inhibitors disclosed in any one of WO 2020118066, WO 2020113071, WO 2020106647, WO

2020106640, WO 2020102730, WO 2020101736, WO 2020097537, WO 2020086739, WO

2020018282, WO 2020050890, WO 2020047192, WO 2020035031, WO 2020033413, WO

2020028706, WO 2019241157, WO 2019234405, WO 2019232419, WO 2019227040, WO

2019217933, WO 2019217691, WO 2019217307, WO 2019215203, WO 2019213526, WO

2019213516, WO 2019204442, WO 2019204449, WO 2019204505, WO 2019155399, WO

2019150305, WO 2019137985, WO 2019110751, WO 2019099524, WO 2019055540, WO

2019051291, WO 2018237084, WO 2018218070, WO 2018217651, WO 2018218071, WO

2018218069, WO 2018212774, WO 2018206539, WO 2018195439, WO 2018143315, WO

2018140600, WO 2018140599, WO 2018140598, WO 2018140514, WO 2018140513, WO

2018140512, WO 2018119183, WO 2018112420, WO 2018068017, WO 2018064510, WO

2018011351, WO 2018005678, WO 2017201161, WO 20171937370, WO 2017172979, WO

2017112777, WO 2017106520, WO 2017096045, WO 2017100546, WO 2017087528, WO

2017079864, WO 2017058807, WO 2017058805, WO 2017058728, WO 2017058902, WO

2017058792, WO 2017058768, WO 2017058915, WO 2017015562, WO 2016179558, WO

2016176338, WO 2016168540, WO 2016164675, WO 2016100546, WO 2016049568, WO

2016049524, WO 2015054572, WO 2014152588, WO 2014143659 and WO 2013155223, each of which is incorporated herein by reference in its entirety. In one such embodiment, the disclosure includes compositions, methods, and kits for the treatment of a disease or condition (e.g ., a cancer or tumor) with a bi-steric mTOR inhibitor and KRAS(OFF) inhibitor selected from AMG 510, MRTX849, JDQ443 and MRTX1133. In some embodiments, the

KRAS(OFF) inhibitor is selected from AMG 510 and MRTX849. In some embodiments, the

KRAS(OFF) inhibitor is AMG 510. In some embodiments, the KRAS(OFF) inhibitor is

MRTX849. In some particular embodiments, the RAS inhibitor is a KRAS(ON) inhibitor known in the art or disclosed herein. The KRAS(ON) inhibitor may be any one or more of the

KRAS(ON) inhibitors disclosed in any one of Appendices A-1, B-1, and C-1, or a RAS inhibitor of WO 2020132597 (wherein WO 2020132597 is incorporated by reference in its entirety). The bi-steric mTOR inhibitor utilized in any such methods may in some embodiments be any bi-steric mTOR inhibitor known in the art or disclosed herein. In some embodiments, the bi-steric mTOR inhibitor is selected from any one of more of the bi-steric mTOR inhibitors disclosed in WO 2016/040806, WO 2018/204416, WO 2019/212990, or WO 2019/212991, each of which is incorporated herein by reference in its entirety. In some embodiments, the bi- steric mTOR inhibitor may be any one or more bi-steric mTOR inhibitors disclosed in Appendix D-1.

[0058] In some embodiments, the mTOR inhibitor is RM-006 (also known as RMC-6272). In some embodiments, the mTOR inhibitor is RMC-5552. In some embodiments, the bi-steric mTOR inhibitor is or a stereoisomer thereof. In some embodiments, the bi-steric mTOR inhibitor is or a tautomer thereof. In some embodiments, the bi-steric mTOR inhibitor is or an oxepane isomer thereof, such as described in WO 2019212990, incorporated herein by reference in its entirety. In some embodiments, the bi-steric mTOR inhibitor is or a stereoisomer thereof. In some embodiments, the bi-steric mTOR inhibitor is or a tautomer thereof. In some embodiments, the bi-steric mTOR inhibitor is

In some embodiments, the bi-steric mTOR inhibitor is

In some embodiments, a composition is provided comprising or a stereoisomer or tautomer thereof and

or a stereoisomer or tautomer thereof. The composition may further comprise a pharmaceutically acceptable excipient. In some embodiments, a composition is provided comprising and

The composition may further comprise a pharmaceutically acceptable excipient.

[0059] Any disease or condition treatable with a RAS inhibitor may be treated according to the present disclosure. The treatment may be in a subject in need thereof. The compounds (e.g ., bi-steric mTOR inhibitor and/or RAS inhibitor, such as a KRAS G12C inhibitor) may be administered to treat the disease or condition (e.g., cancer or a tumor) in an effective amount. In particular embodiments, the disease or condition that is treated according to the methods disclosed herein is a cancer. The cancer may form a tumor. For example, the present disclosure provides a method of treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of the present invention (e.g ., a bi-steric mTOR inhibitor disclosed herein or known in the art and/or RAS inhibitor, such as a KRAS G12C inhibitor disclosed herein or known in the art), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising such a compound or salt.

[0060] In some embodiments, the cancer comprises a RAS mutation. In some embodiments, the cancer is colorectal cancer, non-small cell lung cancer, small-cell lung cancer, pancreatic cancer, appendiceal cancer, melanoma, acute myeloid leukemia, small bowel cancer, ampullary cancer, germ cell cancer, cervical cancer, cancer of unknown primary origin, endometrial cancer, esophagogastric cancer, GI neuroendocrine cancer, ovarian cancer, sex cord stromal tumor cancer, hepatobiliary cancer, or bladder cancer. Also provided is a method of treating a Ras protein-related disorder in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of the present invention (e.g., a bi-steric mTOR inhibitor disclosed herein or known in the art and/or RAS inhibitor, such as a KRAS G12C inhibitor disclosed herein or known in the art), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising such a compound or salt.

[0061] In some embodiments, the compounds of the present invention or pharmaceutically acceptable salts thereof, pharmaceutical compositions comprising such compounds or salts, and methods provided herein may be used for the treatment of a wide variety of cancers including tumors such as lung, prostate, breast, brain, skin, cervical carcinomas, testicular carcinomas, etc. More particularly, cancers that may be treated by the compounds or salts thereof, pharmaceutical compositions comprising such compounds or salts, and methods of the invention include, but are not limited to tumor types such as astrocytic, breast, cervical, colorectal, endometrial, esophageal, gastric, head and neck, hepatocellular, laryngeal, lung, oral, ovarian, prostate and thyroid carcinomas and sarcomas. Other cancers include, for example:

[0062] Cardiac, for example: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma, and teratoma; [0063] Lung, for example: bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma;

[0064] Gastrointestinal, for example: esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Kaposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma);

[0065] Genitourinary tract, for example: kidney (adenocarcinoma, Wilm's tumor (nephroblastoma), lymphoma, leukemia), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma);

[0066] Liver, for example: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma;

[0067] Biliary tract, for example: gall bladder carcinoma, ampullary carcinoma, cholangiocarcinoma;

[0068] Bone, for example: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma, and giant cell tumors;

[0069] Nervous system, for example: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinoma (pinealoma), glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, neurofibromatosis type 1, meningioma, glioma, sarcoma);

[0070] Gynecological, for example: uterus (endometrial carcinoma, uterine carcinoma, uterine corpus endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian carcinoma (serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma), granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), fallopian tubes (carcinoma);

[0071] Hematologic, for example: blood (myeloid leukemia (acute and chronic), acute lymphoblastic leukemia, chronic lymphocytic leukemia), myeloproliferative diseases (e.g., myelofibrosis and myeloproliferative neoplasms): multiple myeloma; myelodysplastic syndromes, Hodgkin's disease, non-Hodgkin's lymphoma (malignant lymphoma);

[0072] Skin, for example: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Kaposi's sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis; and

[0073] Adrenal glands, for example: neuroblastoma.

[0074] In some embodiments, the disease or condition that is treated according to the methods disclosed herein is a RAS G12C cancer. As used herein, the term “G12C cancer” means a cancer that comprises one or more G12C mutation. Such mutations can occur in HRAS, NRAS, and KRAS.

[0075] In some embodiments, the disease or condition that is treated according to the methods disclosed herein is pancreatic cancer, colorectal cancer, non-small cell lung cancer, squamous cell lung carcinoma, thyroid gland adenocarcinoma, or a hematological cancer.

[0076] In some embodiments, the present disclosure includes a method of delaying or preventing acquired resistance to a RAS inhibitor in a subject, comprising administering to the subject a bi-steric inhibitor of mTOR, wherein the subject has already received or will receive administration of the RAS inhibitor. In particular embodiments, the RAS inhibitor targets KRAS, NRAS, or HRAS. In particular embodiments the RAS inhibitor is a RAS mutant specific inhibitor. In certain embodiments, RAS mutant is selected from

(a) the following K-Ras mutants: G12D, G12V, G12C, G13D, G12R, G12A, Q61H, G12S, A146T, G13C, Q61L, Q61R, K117N, A146V, G12F, Q61K, L19F, Q22K, VI 41, A59T, A146P, G13R, G12L, or G13V, and combinations thereof;

(b) the following H-Ras mutants: Q61R, G13R, Q61K, G12S, Q61L, G12D, G13V, G13D, G12C, K117N, A59T, G12V, G13C, Q61H, G13S, A18V, D119N, G13N, A146T, A66T, G12A, A146V, G12N, or G12R, and combinations thereof; and (c) the following N-Ras mutants: Q61R, Q61K, G12D, Q61L, Q61H, G13R, G13D, G12S, G12C, G12V, G12A, G13V, G12R, P185S, G13C, A146T, G60E, Q61P, A59D, E132K, E49K, T50I, A146V, or A59T, and combinations thereof.

In some particular embodiments, the RAS inhibitor is a KRAS(OFF) inhibitor known in the art or disclosed herein. In some embodiments, the disclosure includes compositions, methods, and kits for the delaying or preventing acquired resistance to a KRAS(OFF) inhibitor selected from AMG 510, MRTX849, JDQ443 and MRTX1133, the method comprising administering to the subject a bi-steric mTOR inhibitor. In some embodiments, the KRAS(OFF) inhibitor is selected from AMG 510 and MRTX849. In some embodiments, the KRAS(OFF) inhibitor is AMG 510. In some embodiments, the KRAS(OFF) inhibitor is MRTX849. In some particular embodiments, the RAS inhibitor is a KRAS(ON) inhibitor known in the art or disclosed herein. The KRAS(ON) inhibitor may be any one or more of the KRAS(ON) inhibitors disclosed in any one of Appendices A-1, B-1, and C-1, or a RAS inhibitor of WO 2020132597 (wherein WO 2020132597 is incorporated by reference in its entirety). The bi-steric mTOR inhibitor utilized in any such methods may in some embodiments be any bi-steric mTOR inhibitor known in the art or disclosed herein. In some embodiments, the bi-steric mTOR inhibitor is selected from any one of more of the bi-steric mTOR inhibitors disclosed in WO 2016/040806, WO 2018/204416, WO 2019/212990, or WO 2019/212991, each of which is incorporated herein by reference in its entirety. In some embodiments, the bi-steric mTOR inhibitor is RM- 006 (also known as RMC-6272). In some embodiments, the bi-steric mTOR inhibitor is RMC- 5552. The subject may have a cancer, e.g ., any one of more of the cancers disclosed herein. The cancer may be a G12C cancer.

[0077] In some embodiments, the present disclosure includes a method of treating acquired resistance to a RAS inhibitor in a subject, comprising administering to the subject a bi-steric inhibitor of mTOR, wherein the subject has already received administration of the RAS inhibitor and developed resistance to the RAS inhibitor. In particular embodiments, the RAS inhibitor targets KRAS, NRAS, or HRAS. In particular embodiments the RAS inhibitor is a RAS mutant specific inhibitor. In certain embodiments, RAS mutant is selected from

(a) the following K-Ras mutants: G12D, G12V, G12C, G13D, G12R, G12A, Q61H, G12S, A146T, G13C, Q61L, Q61R, K117N, A146V, G12F, Q61K, L19F, Q22K, VI 41, A59T, A146P, G13R, G12L, or G13V, and combinations thereof;

(b) the following H-Ras mutants: Q61R, G13R, Q61K, G12S, Q61L, G12D, G13V, G13D, G12C, K117N, A59T, G12V, G13C, Q61H, G13S, A18V, D119N, G13N, A146T, A66T, G12A, A146V, G12N, or G12R, and combinations thereof; and

(c) the following N-Ras mutants: Q61R, Q61K, G12D, Q61L, Q61H, G13R, G13D, G12S, G12C, G12V, G12A, G13V, G12R, P185S, G13C, A146T, G60E, Q61P, A59D, E132K, E49K, T50I, A146V, or A59T, and combinations thereof.

In some embodiments, the disclosure includes compositions, methods, and kits for treating acquired resistance to a KRAS(OFF) inhibitor selected from AMG 510, MRTX849, JDQ443, and MRTX1133, the method comprising administering to the subject a bi-steric mTOR inhibitor, wherein the subject has already received administration of the RAS inhibitor and developed resistance to the RAS inhibitor. In some embodiments, the KRAS(OFF) inhibitor is selected from AMG 510 and MRTX849. In some embodiments, the KRAS(OFF) inhibitor is AMG 510. In some embodiments, the KRAS(OFF) inhibitor is MRTX849. In some particular embodiments, the RAS inhibitor is a KRAS(ON) inhibitor known in the art or disclosed herein. The KRAS(ON) inhibitor may be any one or more of the KRAS(ON) inhibitors disclosed in any one of Appendices A-1, B-1, and C-1, or a RAS inhibitor of WO 2020132597 (wherein WO 2020132597 is incorporated by reference in its entirety). The bi- steric mTOR inhibitor utilized in any such methods may in some embodiments be any bi-steric mTOR inhibitor known in the art or disclosed herein. In some embodiments, the bi-steric mTOR inhibitor is selected from any one of more of the bi-steric mTOR inhibitors disclosed in WO 2016/040806, WO 2018/204416, WO 2019/212990, or WO 2019/212991, each of which is incorporated herein by reference in its entirety. In some embodiments, bi-steric mTOR inhibitor is RM-006 (also known as RMC-6272). In some embodiments, the bi-steric inhibitor of mTOR is RMC-5552. The subject may have a cancer, e.g, any one of more of the cancers disclosed herein. The cancer may be a G12C cancer.

[0078] In various embodiments, the methods described herein for treating such diseases or conditions, and for treating, delaying or preventing acquired resistance to a RAS inhibitor in a subject, comprising administering to the subject a bi-steric inhibitor of mTOR, involve administering to a subject an effective amount of a bi-steric mTOR inhibitor, a RAS inhibitor (e.g., a KRAS G12C inhibitor), or a composition (e.g. , a pharmaceutical composition) comprising such a bi-steric mTOR inhibitor, a RAS inhibitor (e.g. , a KRAS G12C inhibitor), or a combination thereof. In some such embodiments, the RAS inhibitor is a KRAS(OFF) inhibitor known in the art or disclosed herein. In some such embodiments, the RAS inhibitor is a KRAS(ON) inhibitor known in the art or disclosed herein. [0079] Any compound or substance capable of inhibiting RAS may be utilized in application with the present disclosure to inhibit RAS. Non-limiting examples of such RAS inhibitors are known in the art and are disclosed herein. For example, the compositions and methods described herein may utilize one or more RAS inhibitor selected from, but not limited to, any KRAS(OFF) inhibitor disclosed herein or known in the art. The KRAS(OFF) inhibitor may be any one or more KRAS(OFF) inhibitor disclosed in any one of WO 2020118066, WO

2020113071, WO 2020106647, WO 2020106640, WO 2020102730, WO 2020101736, WO

2020097537, WO 2020086739, WO 2020018282, WO 2020050890, WO 2020047192, WO

2020035031, WO 2020033413, WO 2020028706, WO 2019241157, WO 2019234405, WO

2019232419, WO 2019227040, WO 2019217933, WO 2019217691, WO 2019217307, WO

2019215203, WO 2019213526, WO 2019213516, WO 2019204442, WO 2019204449, WO

2019204505, WO 2019155399, WO 2019150305, WO 2019137985, WO 2019110751, WO

2019099524, WO 2019055540, WO 2019051291, WO 2018237084, WO 2018218070, WO

2018217651, WO 2018218071, WO 2018218069, WO 2018212774, WO 2018206539, WO

2018195439, WO 2018143315, WO 2018140600, WO 2018140599, WO 2018140598, WO

2018140514, WO 2018140513, WO 2018140512, WO 2018119183, WO 2018112420, WO

2018068017, WO 2018064510, WO 2018011351, WO 2018005678, WO 2017201161, WO

20171937370, WO 2017172979, WO 2017112777, WO 2017106520, WO 2017096045, WO

2017100546, WO 2017087528, WO 2017079864, WO 2017058807, WO 2017058805, WO

2017058728, WO 2017058902, WO 2017058792, WO 2017058768, WO 2017058915, WO

2017015562, WO 2016179558, WO 2016176338, WO 2016168540, WO 2016164675, WO

2016100546, WO 2016049568, WO 2016049524, WO 2015054572, WO 2014152588, WO

2014143659 and WO 2013155223, each of which is incorporated herein by reference in its entirety. In various embodiments, the compositions and methods described herein utilize the

KRAS(OFF) inhibitor AMG 510. In various embodiments, the compositions and methods described herein utilize the KRAS(OFF) inhibitor MRTX849. In various embodiments, the compositions and methods described herein utilize the KRAS(OFF) inhibitor JDQ443. In various embodiments, the compositions and methods described herein utilize the KRAS(OFF) inhibitor MRTX1133. In some embodiments, the compositions and methods described herein utilize a RAS inhibitor that is a KRAS(ON) inhibitor known in the art or disclosed herein. The

KRAS(ON) inhibitor may be any one or more of the KRAS(ON) inhibitors disclosed in any one of Appendices A-1, B-1, and C-1, or a RAS inhibitor of WO 2020132597 (wherein WO

2020132597 is incorporated by reference in its entirety). The compositions and methods described herein may utilize one or more bi-steric mTOR inhibitor selected from, but not limited to any bi-steric mTOR inhibitor disclosed in WO 2016/040806, WO 2018/204416, WO 2019/212990, and WO 2019/212991, each of which is incorporated herein by reference in its entirety.

[0080] The bi-steric mTOR inhibitor may be administered alone as a monotherapy or in combination with one or more other therapeutic agent (e.g ., a RAS inhibitor such as a KRAS(OFF) inhibitor a KRAS(ON) inhibitor and/or an anti-cancer therapeutic agent) as a combination therapy. The bi-steric mTOR inhibitor and/or the RAS inhibitor (e.g., KRAS(OFF) inhibitor or KRAS(ON) inhibitor) may be administered as a pharmaceutical composition. The bi-steric mTOR inhibitor may be administered before, after, and/or concurrently with the one or more other therapeutic agent (e.g, a RAS inhibitor and/or an anti- cancer therapeutic agent). For example, the bi-steric mTOR inhibitor may be administered before, after, and/or concurrently with a KRAS G12C inhibitor. The bi-steric mTOR inhibitor may be administered before, after, and/or concurrently with AMG 510. The bi-steric mTOR inhibitor may be administered before, after, and/or concurrently with MRTX849. The bi-steric mTOR inhibitor may be administered before, after, and/or concurrently with JDQ443. The bi- steric mTOR inhibitor may be administered before, after, and/or concurrently with MRTX1133. The bi-steric mTOR inhibitor may be administered before, after, and/or concurrently with a RAS(ON) inhibitor (e.g., a KRAS(ON) inhibitor). The bi-steric mTOR inhibitor may be administered before, after, and/or concurrently with a RAS(ON) inhibitor disclosed in any one of Appendices A-1, B-1, and C-1, or a RAS inhibitor of WO 2020132597 (wherein WO 2020132597 is incorporated by reference in its entirety). If the bi-steric mTOR inhibitor is administered concurrently with the one or more other therapeutic agent, such administration may be simultaneous (e.g, in a single composition) or may be via two or more separate compositions, optionally via the same or different modes of administration (e.g. , local, systemic, oral, intravenous, etc.).

[0081] In certain embodiments, the bi-steric mTOR inhibitor is administered to the subject as a monotherapy for the treatment of a cancer associated with a mutation in a RAS gene. The RAS gene mutation may be a KRAS, NRAS, or HRAS mutation. Oncogenic RAS mutations, such as KRAS mutations, shift the RAS equilibrium to the GTP -bound “on” state, driving signaling to RAS effectors and oncogene addiction. As used herein, “oncogene addiction” refers to the phenomenon whereby a tumor cell exhibits apparent dependence on a single oncogenic pathway or protein for sustained proliferation and/or survival, despite its myriad of genetic alterations. In certain embodiments, the bi-steric mTOR inhibitor is administered to the subject as a monotherapy for the treatment of a cancer associated with a KRAS G12C mutation. In certain embodiments, the bi-steric mTOR inhibitor is administered to the subject as a monotherapy for the treatment of a cancer associated with a KRAS G12A ; a KRAS G12D , a KRAS G12S , or a KRAS G12V mutation, or any other RAS mutation described herein.

[0082] In certain embodiments, the bi-steric mTOR inhibitor is administered to the subject in combination with one or more other therapeutic agent (e.g ., a RAS inhibitor) as a combination therapy for the treatment of a cancer associated with a mutation in a RAS gene. The mutation may be in KRAS, NRAS or HRAS. The mutation may comprise one or more of a KRAS mutation selected from a KRAS G12A mutation; a KRAS G12C mutation; a KRAS G12D mutation; a KRAS G12S mutation; and a KRAS G12V mutation. The combination therapy may comprise administration of a bi-steric mTOR inhibitor and any RAS inhibitor known in the art or disclosed herein. For example, the bi-steric mTOR inhibitor may be administered to the subject in combination with a KRAS(OFF) inhibitor known in the art or disclosed herein. The bi-steric mTOR inhibitor may be administered to the subject in combination with AMG 510. The bi-steric mTOR inhibitor may be administered to the subject in combination with MRTX849. The bi-steric mTOR inhibitor may be administered to the subject in combination with JDQ443. The bi-steric mTOR inhibitor may be administered to the subject in combination with MRTX1133. The bi-steric mTOR inhibitor may be administered to the subject in combination with a RAS(ON) inhibitor (e.g., a KRAS(ON) inhibitor). The bi-steric mTOR inhibitor may be administered to the subject in combination with a RAS(ON) inhibitor disclosed any one or more of Appendices A-1, B-1, and C-1, or a RAS inhibitor of WO 2020132597 (wherein WO 2020132597 is incorporated by reference in its entirety). The mTOR inhibitor and optionally the RAS inhibitor may also be administered in combination with one or more other therapeutic agent. In some embodiments, the other therapeutic agent used in combination is selected from JNJ-74699157; LY3499446; MRTX1257; ARS 1620; and a combination thereof. MRTX1257 and ARS 1620 have the following structures, respectively:

MRTX1257

ARS 1620

Combination Therapy

[0083] The methods of the invention may include a compound of the invention used alone or in combination with one or more additional therapies (e.g., non-drug treatments or therapeutic agents). In various embodiments, “compound of the invention” refers to any of the compounds described herein. For example, in particular embodiments, the term “compound of the invention” includes any one of more of the RAS inhibitors (e.g., KRAS inhibitors) disclosed herein and any one or more of the bi-steric mTOR inhibitors disclosed herein. In various embodiments, it is contemplated that reference to any one of the compounds disclosed herein (e.g., any one of more of the RAS inhibitors (e.g., KRAS inhibitors) disclosed herein and any one or more of the bi-steric mTOR inhibitors disclosed herein, as well as any other therapeutic agents described herein) also may include a salt of such a compound, such as a pharmaceutically acceptable salt. The dosages of one or more of the additional therapies (e.g., non-drug treatments or therapeutic agents) may be reduced from standard dosages when administered alone. For example, doses may be determined empirically from drug combinations and permutations or may be deduced by i sob olographic analysis (e.g., Black et al., Neurology 65:S3-S6 (2005)). [0084] A compound of the present invention may be administered before, after, or concurrently with one or more of such additional therapies. When combined, dosages of a compound of the invention and dosages of the one or more additional therapies (e.g., non-drug treatment or therapeutic agent) provide a therapeutic effect (e.g., synergistic or additive therapeutic effect). A compound of the present invention and an additional therapy, such as an anti-cancer agent, may be administered together, such as in a unitary pharmaceutical composition, or separately and, when administered separately, this may occur simultaneously or sequentially. Such sequential administration may be close or remote in time.

[0085] In some embodiments, the additional therapy is the administration of side-effect limiting agents (e.g., agents intended to lessen the occurrence or severity of side effects of treatment). For example, in some embodiments, the compounds of the present invention can also be used in combination with a therapeutic agent that treats nausea. Examples of agents that can be used to treat nausea include: dronabinol, granisetron, metoclopramide, ondansetron, and prochlorperazine, or pharmaceutically acceptable salts thereof.

[0086] In some embodiments, the one or more additional therapies includes a non-drug treatment (e.g., surgery or radiation therapy). In some embodiments, the one or more additional therapies includes a therapeutic agent (e.g., a compound or biologic that is an anti-angiogenic agent, signal transduction inhibitor, antiproliferative agent, glycolysis inhibitor, or autophagy inhibitor). In some embodiments, the one or more additional therapies includes a non-drug treatment (e.g., surgery or radiation therapy) and a therapeutic agent (e.g., a compound or biologic that is an anti-angiogenic agent, signal transduction inhibitor, antiproliferative agent, glycolysis inhibitor, or autophagy inhibitor). In other embodiments, the one or more additional therapies includes two therapeutic agents. In still other embodiments, the one or more additional therapies includes three therapeutic agents. In some embodiments, the one or more additional therapies includes four or more therapeutic agents.

[0087] In this Combination Therapy section, all references are incorporated by reference for the agents described, whether explicitly stated as such or not.

Non-drug therapies

[0088] Examples of non-drug treatments include, but are not limited to, radiation therapy, cryotherapy, hyperthermia, surgery (e.g., surgical excision of tumor tissue), and T cell adoptive transfer (ACT) therapy. [0089] In some embodiments, the compounds of the invention may be used as an adjuvant therapy after surgery. In some embodiments, the compounds of the invention may be used as a neo-adjuvant therapy prior to surgery.

[0090] Radiation therapy may be used for inhibiting abnormal cell growth or treating a hyperproliferative disorder, such as cancer, in a subject (e.g., mammal (e.g., human)). Techniques for administering radiation therapy are known in the art. Radiation therapy can be administered through one of several methods, or a combination of methods, including, without limitation, external-beam therapy, internal radiation therapy, implant radiation, stereotactic radiosurgery, systemic radiation therapy, radiotherapy, and permanent or temporary interstitial brachy therapy. The term "brachy therapy," as used herein, refers to radiation therapy delivered by a spatially confined radioactive material inserted into the body at or near a tumor or other proliferative tissue disease site. The term is intended, without limitation, to include exposure to radioactive isotopes (e.g., At-211, I-131, I-125, Y-90, Re-186, Re-188, Sm-153, Bi-212, P- 32, and radioactive isotopes of Lu). Suitable radiation sources for use as a cell conditioner of the present invention include both solids and liquids. By way of non-limiting example, the radiation source can be a radionuclide, such as I-125, I-131, Yb-169, Ir-192 as a solid source, I-125 as a solid source, or other radionuclides that emit photons, beta particles, gamma radiation, or other therapeutic rays. The radioactive material can also be a fluid made from any solution of radionuclide(s), e.g., a solution of I-125 or I-131, or a radioactive fluid can be produced using a slurry of a suitable fluid containing small particles of solid radionuclides, such as Au-198, or Y-90. Moreover, the radionuclide(s) can be embodied in a gel or radioactive micro spheres.

[0091] In some embodiments, the compounds of the present invention can render abnormal cells more sensitive to treatment with radiation for purposes of killing or inhibiting the growth of such cells. Accordingly, this invention further relates to a method for sensitizing abnormal cells in a mammal to treatment with radiation which comprises administering to the mammal an amount of a compound of the present invention, which amount is effective to sensitize abnormal cells to treatment with radiation. The amount of the compound in this method can be determined according to the means for ascertaining effective amounts of such compounds described herein. In some embodiments, the compounds of the present invention may be used as an adjuvant therapy after radiation therapy or as a neo-adjuvant therapy prior to radiation therapy. [0092] In some embodiments, the non-drug treatment is a T cell adoptive transfer (ACT) therapy. In some embodiments, the T cell is an activated T cell. The T cell may be modified to express a chimeric antigen receptor (CAR). CAR modified T (CAR-T) cells can be generated by any method known in the art. For example, the CAR-T cells can be generated by introducing a suitable expression vector encoding the CAR to a T cell. Prior to expansion and genetic modification of the T cells, a source of T cells is obtained from a subject. T cells can be obtained from a number of sources, including peripheral blood mononuclear cells, bone marrow, lymph node tissue, cord blood, thymus tissue, tissue from a site of infection, ascites, pleural effusion, spleen tissue, and tumors. In certain embodiments of the present invention, any number of T cell lines available in the art may be used. In some embodiments, the T cell is an autologous T cell. Whether prior to or after genetic modification of the T cells to express a desirable protein (e.g., a CAR), the T cells can be activated and expanded generally using methods as described, for example, in U.S. Patents 6,352,694; 6,534,055; 6,905,680; 6,692,964; 5,858,358; 6,887,466; 6,905,681; 7,144,575; 7,067,318; 7,172,869; 7,232,566; 7,175,843; 7,572,631; 5,883,223; 6,905,874; 6,797,514; and 6,867,041.

Therapeutic agents

[0093] A therapeutic agent may be a compound used in the treatment of cancer or symptoms associated therewith.

[0094] For example, a therapeutic agent may be a steroid. Accordingly, in some embodiments, the one or more additional therapies includes a steroid. Suitable steroids may include, but are not limited to, 21-acetoxypregnenolone, alclometasone, algestone, amcinonide, beclomethasone, betamethasone, budesonide, chloroprednisone, clobetasol, clocortolone, cloprednol, corticosterone, cortisone, cortivazol, deflazacort, desonide, desoximetasone, dexamethasone, diflorasone, diflucortolone, difuprednate, enoxolone, fluazacort, fiucloronide, flumethasone, flunisolide, fluocinolone acetonide, fluocinonide, fluocortin butyl, fluocortolone, fluorometholone, fluperolone acetate, fluprednidene acetate, fluprednisolone, flurandrenolide, fluticasone propionate, formocortal, halcinonide, halobetasol propionate, halometasone, hydrocortisone, loteprednol etabonate, mazipredone, medrysone, meprednisone, methylprednisolone, mometasone furoate, paramethasone, prednicarbate, prednisolone, prednisolone 25-diethylaminoacetate, prednisolone sodium phosphate, prednisone, prednival, prednylidene, rimexolone, tixocortol, triamcinolone, triamcinolone acetonide, triamcinolone benetonide, triamcinolone hexacetonide, and salts or derivatives thereof. [0095] Further examples of therapeutic agents that may be used in combination therapy with a compound of the present invention include compounds described in the following patents: U.S. Patent Nos. 6,258,812, 6,630,500, 6,515,004, 6,713,485, 5,521,184, 5,770,599, 5,747,498, 5,990,141, 6,235,764, and 8,623,885, and International Patent Applications W001/37820, WOOl/32651, W002/68406, W002/66470, W002/55501, W004/05279, W004/07481, W004/07458, W004/09784, W002/59110, W099/45009, WO00/59509, W099/61422, WO00/12089, and WO00/02871.

[0096] A therapeutic agent may be a biologic (e.g., cytokine (e.g., interferon or an interleukin such as IL-2)) used in treatment of cancer or symptoms associated therewith. In some embodiments, the biologic is an immunoglobulin-based biologic, e.g., a monoclonal antibody (e.g., a humanized antibody, a fully human antibody, an Fc fusion protein, or a functional fragment thereof) that agonizes a target to stimulate an anti-cancer response or antagonizes an antigen important for cancer. Also included are antibody-drug conjugates.

[0097] A therapeutic agent may be a T-cell checkpoint inhibitor. In some embodiments, the checkpoint inhibitor is an inhibitory antibody (e.g., a monospecific antibody such as a monoclonal antibody). The antibody may be, e.g., humanized or fully human. In some embodiments, the checkpoint inhibitor is a fusion protein, e.g., an Fc-receptor fusion protein. In some embodiments, the checkpoint inhibitor is an agent, such as an antibody, that interacts with a checkpoint protein. In some embodiments, the checkpoint inhibitor is an agent, such as an antibody, that interacts with the ligand of a checkpoint protein. In some embodiments, the checkpoint inhibitor is an inhibitor (e.g., an inhibitory antibody or small molecule inhibitor) of CTLA-4 (e.g., an anti-CTLA-4 antibody or fusion a protein). In some embodiments, the checkpoint inhibitor is an inhibitor or antagonist (e.g., an inhibitory antibody or small molecule inhibitor) of PD-1. In some embodiments, the checkpoint inhibitor is an inhibitor or antagonist (e.g., an inhibitory antibody or small molecule inhibitor) of PDL-1. In some embodiments, the checkpoint inhibitor is an inhibitor or antagonist (e.g., an inhibitory antibody or Fc fusion or small molecule inhibitor) of PDL-2 (e.g., a PDL-2/Ig fusion protein). In some embodiments, the checkpoint inhibitor is an inhibitor or antagonist (e.g., an inhibitory antibody or small molecule inhibitor) of B7-H3, B7-H4, BTLA, HVEM, TIM3, GAIN, LAG3, VISTA, KIR, 2B4, CD 160, CGEN-15049, CHK 1, CHK2, A2aR, B-7 family ligands, or a combination thereof. In some embodiments, the checkpoint inhibitor is pembrolizumab, nivolumab, PDR001 (NVS), REGN2810 (Sanofi/Regeneron), a PD-L1 antibody such as, e.g., avelumab, durvalumab, atezolizumab, pidilizumab, JNJ-63723283 (JNJ), BGB-A317 (BeiGene & Celgene) or a checkpoint inhibitor disclosed in Preusser, M. et al. (2015) Nat. Rev. Neurol., including, without limitation, ipilimumab, tremelimumab, nivolumab, pembrolizumab, AMP224, AMP514/ MEDI0680, BMS936559, MED14736, MPDL3280A, MSB0010718C, BMS986016, IMP321, lirilumab, IPH2101, 1-7F9, and KW-6002.

[0098] A therapeutic agent may be an anti-TIGIT antibody, such as MBSA43, BMS- 986207, MK-7684, COM902, AB154, MTIG7192A or OMP-313M32 (etigilimab).

[0099] A therapeutic agent may be an agent that treats cancer or symptoms associated therewith (e.g., a cytotoxic agent, non-peptide small molecules, or other compound useful in the treatment of cancer or symptoms associated therewith, collectively, an “anti-cancer agent”). Anti-cancer agents can be, e.g., chemotherapeutics or targeted therapy agents.

[00100] Anti-cancer agents include mitotic inhibitors, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, alkylating agents, antimetabolites, folic acid analogs, pyrimidine analogs, purine analogs and related inhibitors, vinca alkaloids, epipodopyyllotoxins, antibiotics, L- Asparaginase, topoisomerase inhibitors, interferons, platinum coordination complexes, anthracenedione substituted urea, methyl hydrazine derivatives, adrenocortical suppressant, adrenocorticosteroides, progestins, estrogens, antiestrogen, androgens, antiandrogen, and gonadotropin-releasing hormone analog. Further anti-cancer agents include leucovorin (LV), irenotecan, oxaliplatin, capecitabine, paclitaxel, and doxetaxel. In some embodiments, the one or more additional therapies includes two or more anti-cancer agents. The two or more anti- cancer agents can be used in a cocktail to be administered in combination or administered separately. Suitable dosing regimens of combination anti-cancer agents are known in the art and described in, for example, Saltz et al., Proc. Am. Soc. Clin. Oncol. 18:233a (1999), and Douillard et al., Lancet 355(9209): 1041-1047 (2000).

[00101] Other non-limiting examples of anti-cancer agents include Gleevec® (Imatinib Mesylate); Kyprolis® (carfilzomib); Velcade® (bortezomib); Casodex (bicalutamide); Iressa® (gefitinib); alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, triethylenephosphoramide, triethiylenethiophosphoramide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analogue topotecan); bryostatin; cally statin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CB1-TM1); eleutherobin; pancrati statin; sarcodictyin A; spongistatin; nitrogen mustards such as chlorambucil, chlomaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimustine; antibiotics such as the enediyne antibiotics (e.g., calicheamicin, such as calicheamicin gammall and calicheamicin omegall (see, e.g., Agnew, Chem. Inti. EdEngl. 33:183-186 (1994)); dynemicin such as dynemicin A; bisphosphonates such as clodronate; an esperamicin; neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromophores, aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, calicheamicin, carabicin, caminomycin, carminomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo- 5-oxo-L-norleucine, adriamycin (doxorubicin), morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin, deoxydoxorubicin, epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenishers such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfomithine; elliptinium acetate; an epothilone such as epothilone B; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidamine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; phenamet; pirarubicin; losoxantrone; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK® polysaccharide complex (JHS Natural Products, Eugene, OR); razoxane; rhizoxin; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2,2',2"-trichlorotriethylamine; trichothecenes such as T- 2 toxin, verracurin A, roridin A and anguidine; urethane; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C"); cyclophosphamide; thiotepa; taxoids, e.g., Taxol® (paclitaxel), Abraxane® (cremophor-free, albumin-engineered nanoparticle formulation of paclitaxel), and Taxotere® (doxetaxel); chloranbucil; tamoxifen (Nolvadex™); raloxifene, aromatase inhibiting 4(5)-imidazoles; 4-hydroxytamoxifen; trioxifene; keoxifene; LY 117018; onapristone; toremifene (Fareston®); flutamide, nilutamide, bicalutamide, leuprolide, goserelin; chlorambucil; Gemzar® gemcitabine; 6-thioguanine; mercaptopurine; platinum coordination complexes such as cisplatin, oxaliplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitoxantrone; vincristine; Navelbine® (vinorelbine); novantrone; teniposide; edatrexate; daunomycin; aminopterin; ibandronate; irinotecan (e.g., CPT-11); topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoids such as retinoic acid; esperamicins; capecitabine (e.g., Xeloda®); and pharmaceutically acceptable salts of any of the above.

[00102] Additional non-limiting examples of anti-cancer agents include trastuzumab (Herceptin®), bevacizumab (Avastin®), cetuximab (Erbitux®), rituximab (Rituxan®), Taxol®, Arimidex®, ABVD, avicine, abagovomab, acridine carboxamide, adecatumumab, 17- N-allylamino-17-demethoxygeldanamycin, alpharadin, alvocidib, 3-aminopyridine-2- carboxaldehyde thiosemicarbazone, amonafide, anthracenedione, anti-CD22 immunotoxins, antineoplastics (e.g., cell-cycle nonspecific antineoplastic agents, and other antineoplastics described herein), antitumorigenic herbs, apaziquone, atiprimod, azathioprine, belotecan, bendamustine, BIBW 2992, biricodar, brostallicin, bryostatin, buthionine sulfoximine, CBV (chemotherapy), calyculin, dichloroacetic acid, discodermolide, elsamitrucin, enocitabine, eribulin, exatecan, exisulind, ferruginol, forodesine, fosfestrol, ICE chemotherapy regimen, IT- 101, imexon, imiquimod, indolocarbazole, irofulven, laniquidar, larotaxel, lenalidomide, lucanthone, lurtotecan, mafosfamide, mitozolomide, nafoxidine, nedaplatin, olaparib, ortataxel, PAC-1, pawpaw, pixantrone, proteasome inhibitors, rebeccamycin, resiquimod, rubitecan, SN-38, salinosporamide A, sapacitabine, Stanford V, swainsonine, talaporfm, tariquidar, tegafur-uracil, temodar, tesetaxel, triplatin tetranitrate, tris(2-chloroethyl)amine, troxacitabine, uramustine, vadimezan, vinflunine, ZD6126, and zosuquidar.

[00103] Further non-limiting examples of anti-cancer agents include natural products such as vinca alkaloids (e.g., vinblastine, vincristine, and vinorelbine), epidipodophyllotoxins (e.g., etoposide and teniposide), antibiotics (e.g., dactinomycin (actinomycin D), daunorubicin, and idarubicin), anthracyclines, mitoxantrone, bleomycins, plicamycin (mithramycin), mitomycin, enzymes (e.g., L-asparaginase which systemically metabolizes L-asparagine and deprives cells which do not have the capacity to synthesize their own asparagine), antiplatelet agents, antiproliferative/antimitotic alkylating agents such as nitrogen mustards (e.g., mechlorethamine, cyclophosphamide and analogs, melphalan, and chlorambucil), ethylenimines and methylmelamines (e.g., hexaamethylmelaamine and thiotepa), CDK inhibitors (e.g., a CDK4/6 inhibitor such as abemaciclib, ribociclib, palbociclib; seliciclib, UCN-01, P1446A-05, PD-0332991, dinaciclib, P27-00, AT-7519, RGB286638, and SCH727965), alkyl sulfonates (e.g., busulfan), nitrosoureas (e.g., carmustine (BCNU) and analogs, and streptozocin), trazenes-dacarbazinine (DTIC), antiproliferative/antimitotic antimetabolites such as folic acid analogs, pyrimidine analogs (e.g., fluorouracil, floxuridine, and cytarabine), purine analogs and related inhibitors (e.g., mercaptopurine, thioguanine, pentostatin, and 2-chlorodeoxyadenosine), aromatase inhibitors (e.g., anastrozole, exemestane, and letrozole), and platinum coordination complexes (e.g., cisplatin and carboplatin), procarbazine, hydroxyurea, mitotane, aminoglutethimide, histone deacetylase (HDAC) inhibitors (e.g., trichostatin, sodium butyrate, apicidan, suberoyl anilide hydroamic acid, vorinostat, LBH 589, romidepsin, ACY-1215, and panobinostat), KSP(Eg5) inhibitors (e.g., Array 520), DNA binding agents (e.g., Zalypsis®), PI3K inhibitors such as PI3K delta inhibitor (e.g., GS-1101 and TGR-1202), PI3K delta and gamma inhibitor (e.g., CAL-130), copanlisib, alpelisib and idelalisib; multi-kinase inhibitor (e.g., TG02 and sorafenib), hormones (e.g., estrogen) and hormone agonists such as leutinizing hormone releasing hormone (LHRH) agonists (e.g., goserelin, leuprolide and triptorelin), BAFF-neutralizing antibody (e.g., LY2127399), IKK inhibitors, p38MAPK inhibitors, anti-IL-6 (e.g., CNT0328), telomerase inhibitors (e.g., GRN 163L), aurora kinase inhibitors (e.g., MLN8237), cell surface monoclonal antibodies (e.g., anti-CD38 (HUMAX-CD38), anti-CS1 (e.g., elotuzumab), HSP90 inhibitors (e.g., 17 AAG and KOS 953), P13K / Akt inhibitors (e.g., perifosine), Akt inhibitors (e.g., GSK-2141795), PKC inhibitors (e.g., enzastaurin), FTIs (e.g., Zarnestra™), anti-CD138 (e.g., BT062), Torcl/2 specific kinase inhibitors (e.g., INK128), ER/UPR targeting agents (e.g., MKC-3946), cFMS inhibitors (e.g., ARRY-382), JAKl/2 inhibitors (e.g., CYT387), PARP inhibitors (e.g., olaparib and veliparib (ABT-888)), and BCL-2 antagonists.

[00104] In some embodiments, an anti-cancer agent is selected from mechlorethamine, camptothecin, ifosfamide, tamoxifen, raloxifene, gemcitabine, Navelbine®, sorafenib, or any analog or derivative variant of the foregoing.

[00105] In some embodiments, the anti-cancer agent is a HER2 inhibitor. Non-limiting examples of HER2 inhibitors include monoclonal antibodies such as trastuzumab (Herceptin®) and pertuzumab (Perjeta®); small molecule tyrosine kinase inhibitors such as gefitinib (Iressa®), erlotinib (Tarceva®), pilitinib, CP-654577, CP-724714, canertinib (Cl 1033), HKI- 272, lapatinib (GW-572016; Tykerb®), PKI-166, AEE788, BMS-599626, HKI-357, BIBW 2992, ARRY-334543, and JNJ-26483327.

[00106] In some embodiments, an anti-cancer agent is an ALK inhibitor. Non-limiting examples of ALK inhibitors include ceritinib, TAE-684 (NVP-TAE694), PF02341066 (crizotinib or 1066), alectinib; brigatinib; entrectinib; ensartinib (X-396); lorlatinib; ASP3026; CEP-37440; 4SC-203; TL-398; PLB1003; TSR-011; CT-707; TPX-0005, and AP26113. Additional examples of ALK kinase inhibitors are described in examples 3-39 of W005016894.

[00107] In some embodiments, an anti-cancer agent is an inhibitor of a member downstream of a Receptor Tyrosine Kinase (RTK)/Growth Factor Receptor (e.g., a SHP2 inhibitor (e.g., SHP099, TN0155, RMC-4550, RMC-4630, JAB-3068, JAB-3312, RLY-1971, ERAS-601, or BBP-398), an SOS1 inhibitor (e.g., BI-1701963, BI-3406), a Raf inhibitor, a MEK inhibitor, an ERK inhibitor, a PI3K inhibitor, a PTEN inhibitor, or an AKT inhibitor. In some embodiments, the anti-cancer agent is JAB-3312.

[00108] In some embodiments, a therapeutic agent that may be combined with a compound of the present invention is an inhibitor of the MAP kinase (MAPK) pathway (or “MAPK inhibitor”). MAPK inhibitors include, but are not limited to, one or more MAPK inhibitor described in Cancers (Basel) 2015 Sep; 7(3): 1758-1784. For example, the MAPK inhibitor may be selected from one or more of trametinib, binimetinib, selumetinib, cobimetinib, LErafAON (NeoPharm), ISIS 5132; vemurafenib, pimasertib, TAK733, R04987655 (CH4987655); CI-1040; PD-0325901; CH5126766; MAP855; AZD6244; refametinib (RDEA 119/BAY 86-9766); GDC-0973/XL581; AZD8330 (ARRY-

424704/ ARRY-704); R05126766 (Roche, described in PLoS One. 2014 Nov 25;9(11)); and GSK1120212 (or JTP-74057, described in Clin Cancer Res. 2011 Mar 1;17(5):989-1000). The MAPK inhibitor may be PLX8394, LXH254, GDC-5573, or LY3009120.

[00109] In some embodiments, an anti-cancer agent is a disrupter or inhibitor of the RAS- RAF-ERK or PI3K-AKT-TOR or PI3K-AKT signaling pathways. The PI3K/AKT inhibitor may include, but is not limited to, one or more PI3K/AKT inhibitor described in Cancers (Basel) 2015 Sep; 7(3): 1758-1784. For example, the PI3K/AKT inhibitor may be selected from one or more of NVP-BEZ235; BGT226; XL765/SAR245409; SF1126; GDC-0980; PI- 103; PF-04691502; PKI-587; GSK2126458. [00110] In some embodiments, an anti-cancer agent is a PD-1 or PD-L1 antagonist.

[00111] In some embodiments, additional therapeutic agents include ALK inhibitors, HER2 inhibitors, EGFR inhibitors, IGF-1R inhibitors, MEK inhibitors, PI3K inhibitors, AKT inhibitors, TOR inhibitors, MCL-1 inhibitors, BCL-2 inhibitors, SHP2 inhibitors, proteasome inhibitors, and immune therapies. In some embodiments, a therapeutic agent may be a pan- RTK inhibitor, such as afatinib.

[00112] IGF-1R inhibitors include linsitinib, or a pharmaceutically acceptable salt thereof.

[00113] EGFR inhibitors include, but are not limited to, small molecule antagonists, antibody inhibitors, or specific antisense nucleotide or siRNA. Useful antibody inhibitors of EGFR include cetuximab (Erbitux®), panitumumab (Vectibix®), zalutumumab, nimotuzumab, and matuzumab. Further antibody -based EGFR inhibitors include any anti- EGFR antibody or antibody fragment that can partially or completely block EGFR activation by its natural ligand. Non-limiting examples of antibody -based EGFR inhibitors include those described in Modjtahedi et al., Br. J. Cancer 1993, 67:247-253; Teramoto et al., Cancer 1996, 77:639-645; Goldstein et al., Clin. Cancer Res. 1995, 1:1311-1318; Huang et al., 1999, Cancer Res. 15:59(8):1935-40; and Yang et al., Cancer Res.1999, 59:1236-1243. The EGFR inhibitor can be monoclonal antibody Mab E7.6.3 (Yang, 1999 supra), or Mab C225 (ATCC Accession No. HB-8508), or an antibody or antibody fragment having the binding specificity thereof.

[00114] Small molecule antagonists of EGFR include gefitinib (Iressa®), erlotinib (Tarceva®), and lapatinib (TykerB®). See, e.g., Yan et al., Pharmacogenetics and Pharmacogenomics In Oncology Therapeutic Antibody Development, BioTechniques 2005, 39(4):565-8; and Paez et al., EGFR Mutations In Lung Cancer Correlation With Clinical Response To Gefitinib Therapy, Science 2004, 304(5676): 1497-500. In some embodiments, the EGFR inhibitor is osimertinib (Tagrisso®). Further non-limiting examples of small molecule EGFR inhibitors include any of the EGFR inhibitors described in the following patent publications, and all pharmaceutically acceptable salts of such EGFR inhibitors: EP 0520722; EP 0566226; WO96/33980; U.S. Pat. No. 5,747,498; WO96/30347; EP 0787772;

W097/30034; W097/30044; W097/38994; W097/49688; EP 837063; WO98/02434;

W097/38983; W095/19774; WO95/19970; W097/13771; WO98/02437; WO98/02438; W097/32881; DE 19629652; W098/33798; WO97/32880; WO97/32880; EP 682027; WO97/02266; W097/27199; WO98/07726; W097/34895; WO96/31510; W098/14449; WO98/14450; W098/14451; WO95/09847; WO97/19065; W098/17662; U.S. Pat. No. 5,789,427; U.S. Pat. No. 5,650,415; U.S. Pat. No. 5,656,643; W099/35146; W099/35132; W099/07701; and WO92/20642. Additional non-limiting examples of small molecule EGFR inhibitors include any of the EGFR inhibitors described in Traxler et al., Exp. Opin. Ther. Patents 1998, 8(12): 1599-1625.

[00115] MEK inhibitors include, but are not limited to, pimasertib, selumetinib, cobimetinib (Cotellic®), trametinib (Mekinist®), and binimetinib (Mektovi®). In some embodiments, a MEK inhibitor targets a MEK mutation that is a Class I MEKl mutation selected from D67N; P124L; P124S; and L177V. In some embodiments, the MEK mutation is a Class II MEKl mutation selected from ΔE51-Q58; ΔF53-Q58; E203K; L177M; C121S; F53L; K57E; Q56P; and K57N.

[00116] PI3K inhibitors include, but are not limited to, wortmannin; 17-hydroxy wortmannin analogs described in WO06/044453; 4-[2-(1H-Indazol-4-yl)-6-[[4-(methylsulfonyl)piperazin- 1-yl]methyl]thieno[3,2-d]pyrimidin-4-yl]morpholine (also known as pictilisib or GDC-0941 and described in W009/036082 and W009/055730); 2-methyl-2-[4-[3-methyl-2-oxo-8- (quinolin-3-yl)-2,3-dihydroimidazo[4,5-c]quinolin-1-yl]pheny l]propionitrile (also known as BEZ 235 or NVP-BEZ 235, and described in W006/122806); (S)-1-(4-((2-(2-aminopyrimidin- 5-yl)-7-methyl-4-morpholinothieno[3,2-d]pyrimidin-6-yl)methy l)piperazin-1-yl)-2- hydroxypropan-1-one (described in W008/070740); LY294002 (2-(4-morpholinyl)-8-phenyl- 4H-1-benzopyran-4-one (available from Axon Medchem); PI 103 hydrochloride (3-[4-(4- morpholinylpyrido-[3',2':4,5]furo[3,2-d]pyrimidin-2-yl] phenol hydrochloride (available from Axon Medchem); PIK 75 (2-methyl-5-nitro-2-[(6-bromoimidazo[1,2-a]pyridin-3- yl)methylene]-1-methylhydrazide-benzenesulfonic acid, monohydrochloride) (available from Axon Medchem); PIK 90 (N-(7,8-dimethoxy-2,3-dihydro-imidazo[1,2-c]quinazolin-5-yl) - nicotinamide (available from Axon Medchem); AS-252424 (5-[1-[5-(4-fluoro-2-hydroxy- phenyl)-furan-2-yl]-meth-(Z)-ylidene]-thiazolidine-2,4-dione (available from Axon Medchem); TGX-221 (7-methyl-2-(4-morpholinyl)-9-[1-(phenylamino)ethyl]-4H-pyri do- [1,2-a]pyrirnidin-4-one (available from Axon Medchem); XL-765; and XL-147. Other PI3K inhibitors include demethoxyviridin, perifosine, CALIOI, PX-866, BEZ235, SF1126, INK1117, IPI-145, BKM120, XL147, XL765, Palomid 529, GSK1059615, ZSTK474, PWT33597, IC87114, TGI 00-115, CAL263, PI-103, GNE-477, CUDC-907, and AEZS-136.

[00117] AKT inhibitors include, but are not limited to, Akt-1-1 (inhibits Aktl) (Barnett et al., Biochem. J. 2005, 385(Pt. 2): 399-408); Akt-1-1, 2 (inhibits Akl and 2) (Barnett et al., Biochem. J. 2005, 385(Pt. 2): 399-408); API-59CJ-Ome (e.g., Jin et al., Br. J. Cancer 2004, 91:1808-12); 1-H-imidazo[4,5-c]pyridinyl compounds (e.g., WO 05/011700); indole-3- carbinol and derivatives thereof (e.g., U.S. Pat. No. 6,656,963; Sarkar and Li J Nutr. 2004, 134(12 Suppl):3493S-3498S); perifosine (e.g., interferes with Akt membrane localization; Dasmahapatra et al. Clin. Cancer Res. 2004, 10(15):5242-52); phosphatidylinositol ether lipid analogues (e.g., Gills and Dennis Expert. Opin. Investig. Drugs 2004, 13:787-97); and triciribine (TCN or API-2 or NCI identifier: NSC 154020; Yang et al., Cancer Res. 2004, 64:4394-9).

[00118] BRAF inhibitors that may be used in combination with compounds of the invention include, for example, vemurafenib, dabrafenib, and encorafenib. A BRAF may comprise a Class 3 BRAF mutation. In some embodiments, the Class 3 BRAF mutation is selected from one or more of the following amino acid substitutions in human BRAF: D287H; P367R; V459L; G466V; G466E; G466A; S467L; G469E; N581S; N581I; D594N; D594G; D594A; D594H; F595L; G596D; G596R and A762E.

[00119] MCL-1 inhibitors include, but are not limited to, AMG-176, MIK665, and S63845. The myeloid cell leukemia-1 (MCL-1) protein is one of the key anti-apoptotic members of the B-cell lymphoma-2 (BCL-2) protein family. Over-expression of MCL-1 has been closely related to tumor progression as well as to resistance, not only to traditional chemotherapies but also to targeted therapeutics including BCL-2 inhibitors such as ABT-263.

[00120] In some embodiments, the additional therapeutic agent is a SHP2 inhibitor. SHP2 is a non-receptor protein tyrosine phosphatase encoded by the PTPN11 gene that contributes to multiple cellular functions including proliferation, differentiation, cell cycle maintenance and migration. SHP2 has two N-terminal Src homology 2 domains (N-SH2 and C-SH2), a catalytic domain (PTP), and a C-terminal tail. The two SH2 domains control the subcellular localization and functional regulation of SHP2. The molecule exists in an inactive, self- inhibited conformation stabilized by a binding network involving residues from both the N- SH2 and PTP domains. Stimulation by, for example, cytokines or growth factors acting through receptor tyrosine kinases (RTKs) leads to exposure of the catalytic site resulting in enzymatic activation of SHP2.

[00121] SHP2 is involved in signaling through the RAS-mitogen-activated protein kinase (MAPK), the JAK-STAT or the phosphoinositol 3-kinase-AKT pathways. Mutations in the PTPN11 gene and subsequently in SHP2 have been identified in several human developmental diseases, such as Noonan Syndrome and Leopard Syndrome, as well as human cancers, such as juvenile myelomonocytic leukemia, neuroblastoma, melanoma, acute myeloid leukemia and cancers of the breast, lung and colon. Some of these mutations destabilize the auto-inhibited conformation of SHP2 and promote autoactivation or enhanced growth factor driven activation of SHP2. SHP2, therefore, represents a highly attractive target for the development of novel therapies for the treatment of various diseases including cancer. A SHP2 inhibitor (e.g., RMC- 4550 or SHP099) in combination with a RAS pathway inhibitor (e.g., a MEK inhibitor) have been shown to inhibit the proliferation of multiple cancer cell lines in vitro (e.g., pancreas, lung, ovarian and breast cancer). Thus, combination therapy involving a SHP2 inhibitor with a RAS pathway inhibitor could be a general strategy for preventing tumor resistance in a wide range of malignancies.

[00122] Non-limiting examples of such SHP2 inhibitors that are known in the art, include: Chen etal. Mol Pharmacol . 2006, 70, 562; Sarver et al., J. Med. Chem. 2017, 62, 1793; Xie et al., J. Med. Chem. 2017, 60, 113734; and Igbe et al., Oncotarget, 2017, 8, 113734; and applications: WO 2021110796; WO 2021088945; WO 2021073439, WO 2021061706, WO

2021061515, WO 2021043077, WO 2021033153, WO 2021028362, WO 2021033153, WO

2021028362, WO 2021018287, WO 2020259679, WO 2020249079, WO 2020210384, WO

2020201991, WO 2020181283, WO 2020177653, WO 2020165734, WO 2020165733, WO

2020165732, WO 2020156243, WO 2020156242, WO 2020108590, WO 2020104635, WO

2020094104, WO 2020094018, WO 2020081848, WO 2020073949, WO 2020073945, WO

2020072656, WO 2020065453, WO 2020065452, WO 2020063760, WO 2020061103, WO

2020061101, WO 2020033828, WO 2020033286, WO 2020022323, WO 2019233810, WO

2019213318, WO 2019183367, WO 2019183364, WO 2019182960, WO 2019167000, WO

2019165073, WO 2019158019, WO 2019152454, WO 2019051469, WO 2019051084, WO

2018218133, WO 2018172984, WO 2018160731, WO 2018136265, WO 2018136264, WO

2018130928, WO 2018129402, WO 2018081091, WO 2018057884, WO 2018013597, WO

2017216706, WO 2017211303, WO 2017210134, WO 2017156397, WO 2017100279, WO

2017079723, WO 2017078499, WO 2016203406, WO 2016203405, WO 2016203404, WO

2016196591, WO 2016191328, WO 2015107495, WO 2015107494, WO 2015107493, WO

2014176488, WO 2014113584, US 20210085677, US 10988466, US 10858359, US 10934302 and US 10954243, each of which is incorporated herein by reference in its entirety.

[00123] In some embodiments, a SHP2 inhibitor binds in the active site. In some embodiments, a SHP2 inhibitor is a mixed-type irreversible inhibitor. In some embodiments, a SHP2 inhibitor binds an allosteric site e.g., a non-covalent allosteric inhibitor. In some embodiments, a SHP2 inhibitor is a covalent SHP2 inhibitor, such as an inhibitor that targets the cysteine residue (C333) that lies outside the phosphatase’s active site. In some embodiments a SHP2 inhibitor is a reversible inhibitor. In some embodiments, a SHP2 inhibitor is an irreversible inhibitor. In some embodiments, the SHP2 inhibitor is SHP099. In some embodiments, the SHP2 inhibitor is TN0155. In some embodiments, the SHP2 inhibitor is RMC-4550. In some embodiments, the SHP2 inhibitor is RMC-4630, whose structure is shown below:

[00124] In some embodiments, the SHP2 inhibitor is JAB-3068.

[00125] In some embodiments, the additional therapeutic agent is selected from the group consisting of a HER2 inhibitor, a SHP2 inhibitor, a CDK4/6 inhibitor, an SO SI inhibitor, and a PD-L1 inhibitor. See, e.g., Hallin et al., Cancer Discovery, DOI: 10.1158/2159-8290 (October 28, 2019) and Canon et al., Nature, 575:217 (2019).

[00126] Proteasome inhibitors include, but are not limited to, carfilzomib (Kyprolis®), bortezomib (Velcade®), and oprozomib.

[00127] Immune therapies include, but are not limited to, monoclonal antibodies, immunomodulatory imides (IMiDs), GITR agonists, genetically engineered T-cells (e.g., CAR-T cells), bispecific antibodies (e.g., BiTEs), and anti-PD-1, anti-PDL-1, anti-CTLA4, anti-LAG1, and anti-OX40 agents).

[00128] Immunomodulatory agents (IMiDs) are a class of immunomodulatory drugs (drugs that adjust immune responses) containing an imide group. The IMiD class includes thalidomide and its analogues (lenalidomide, pomalidomide, and apremilast). [00129] Exemplary anti -PD-1 antibodies and methods for their use are described by Goldberg et al., Blood 2007, 110(1): 186-192; Thompson et al., Clin. Cancer Res. 2007, 13(6): 1757-1761; and WO06/121168 Al), as well as described elsewhere herein.

[00130] GITR agonists include, but are not limited to, GITR fusion proteins and anti-GITR antibodies (e.g., bivalent anti-GITR antibodies), such as, a GITR fusion protein described in U.S. Pat. No. 6,111,090, U.S. Pat. No. 8,586,023, W02010/003118 and WO2011/090754; or an anti-GITR antibody described, e.g., in U.S. Pat. No. 7,025,962, EP 1947183, U.S. Pat. No. 7,812,135, U.S. Pat. No. 8,388,967, U.S. Pat. No. 8,591,886, U.S. Pat. No. 7,618,632, EP 1866339, and WO2011/028683, WO2013/039954, W005/007190, WO07/133822,

W005/055808, WO99/40196, W001/03720, WO99/20758, WO06/083289, WO05/115451, and WO2011/051726.

[00131] Another example of a therapeutic agent that may be used in combination with the compounds of the invention is an anti-angiogenic agent. Anti -angiogenic agents are inclusive of, but not limited to, in vitro synthetically prepared chemical compositions, antibodies, antigen binding regions, radionuclides, and combinations and conjugates thereof. An anti -angiogenic agent can be an agonist, antagonist, allosteric modulator, toxin or, more generally, may act to inhibit or stimulate its target (e.g., receptor or enzyme activation or inhibition), and thereby promote cell death or arrest cell growth. In some embodiments, the one or more additional therapies include an anti -angiogenic agent.

[00132] Anti-angiogenic agents can be MMP-2 (matrix-metalloproteinase 2) inhibitors, MMP-9 (matrix-metalloprotienase 9) inhibitors, and COX-II (cyclooxygenase 11) inhibitors. Non-limiting examples of anti-angiogenic agents include rapamycin, temsirolimus (CCI-779), everolimus (RAD001), sorafenib, sunitinib, and bevacizumab. Examples of useful COX-II inhibitors include alecoxib, valdecoxib, and rofecoxib. Examples of useful matrix metalloproteinase inhibitors are described in W096/33172, W096/27583, WO98/07697, WO98/03516, W098/34918, W098/34915, W098/33768, WO98/30566, W090/05719, WO99/52910, W099/52889, W099/29667, WO99007675, EP0606046, EP0780386, EP1786785, EP1181017, EP0818442, EP1004578, andUS20090012085, and U.S. Patent Nos. 5,863,949 and 5,861,510. Preferred MMP-2 and MMP-9 inhibitors are those that have little or no activity inhibiting MMP-1. More preferred, are those that selectively inhibit MMP-2 or AMP-9 relative to the other matrix- metalloproteinases (i.e., MAP-1, MMP-3, MMP-4, MMP- 5, MMP-6, MMP- 7, MMP- 8, MMP-10, MMP-11, MMP-12, and MMP-13). Some specific examples of MMP inhibitors are AG-3340, RO 32-3555, and RS 13-0830. [00133] Further exemplary anti-angiogenic agents include KDR (kinase domain receptor) inhibitory agents (e.g., antibodies and antigen binding regions that specifically bind to the kinase domain receptor), anti-VEGF agents (e.g., antibodies or antigen binding regions that specifically bind VEGF (e.g., bevacizumab), or soluble VEGF receptors or a ligand binding region thereof) such as VEGF-TRAP™, and anti-VEGF receptor agents (e.g., antibodies or antigen binding regions that specifically bind thereto), EGFR inhibitory agents (e.g., antibodies or antigen binding regions that specifically bind thereto) such as Vectibix® (panitumumab), erlotinib (Tarceva®), anti-Angl and anti-Ang2 agents (e.g., antibodies or antigen binding regions specifically binding thereto or to their receptors, e.g., Tie2/Tek), and anti-Tie2 kinase inhibitory agents (e.g., antibodies or antigen binding regions that specifically bind thereto). Other anti-angiogenic agents include Campath, IL-8, B-FGF, Tek antagonists (US2003/0162712; US6,413,932), anti-TWEAK agents (e.g., specifically binding antibodies or antigen binding regions, or soluble TWEAK receptor antagonists; see US6, 727,225), ADAM distintegrin domain to antagonize the binding of integrin to its ligands (US 2002/0042368), specifically binding anti-eph receptor or anti-ephrin antibodies or antigen binding regions (U.S. Patent Nos. 5,981,245; 5,728,813; 5,969,110; 6,596,852; 6,232,447; 6,057,124 and patent family members thereof), and anti-PDGF-BB antagonists (e.g., specifically binding antibodies or antigen binding regions) as well as antibodies or antigen binding regions specifically binding to PDGF-BB ligands, and PDGFR kinase inhibitory agents (e.g., antibodies or antigen binding regions that specifically bind thereto). Additional anti- angiogenic agents include: SD-7784 (Pfizer, USA); cilengitide (Merck KGaA, Germany, EPO 0770622); pegaptanib octasodium, (Gilead Sciences, USA); Alphastatin, (BioActa, UK); M- PGA, (Celgene, USA, US 5712291); ilomastat, (Arriva, USA, US5892112); emaxanib, (Pfizer, USA, US 5792783); vatalanib, (Novartis, Switzerland); 2-methoxyestradiol (EntreMed, USA); TLC ELL-12 (Elan, Ireland); anecortave acetate (Alcon, USA); alpha-D148 Mab (Amgen, USA); CEP-7055 (Cephalon, USA); anti-Vn Mab (Crucell, Netherlands), DACantiangiogenic (ConjuChem, Canada); Angiocidin (InKine Pharmaceutical, USA); KM-2550 (Kyowa Hakko, Japan); SU-0879 (Pfizer, USA); CGP-79787 (Novartis, Switzerland, EP 0970070); ARGENT technology (Ariad, USA); YIGSR-Stealth (Johnson & Johnson, USA); fibrinogen-E fragment (BioActa, UK); angiogenic inhibitor (Trigen, UK); TBC-1635 (Encysive Pharmaceuticals, USA); SC-236 (Pfizer, USA); ABT-567 (Abbott, USA); Metastatin (EntreMed, USA); maspin (Sosei, Japan); 2-methoxyestradiol (Oncology Sciences Corporation, USA); ER-68203-00 (IV AX, USA); BeneFin (Lane Labs, USA); Tz-93 (Tsumura, Japan); TAN-1120 (Takeda, Japan);

FR-111142 (Fujisawa, Japan, JP 02233610); platelet factor 4 (RepliGen, USA, EP 407122); vascular endothelial growth factor antagonist (Borean, Denmark); bevacizumab (pINN)

(Genentech, USA); angiogenic inhibitors (SUGEN, USA); XL 784 (Exelixis, USA); XL 647

(Exelixis, USA); MAb, alpha5beta3 integrin, second generation (Applied Molecular Evolution,

USA and Medlmmune, USA); enzastaurin hydrochloride (Lilly, USA); CEP 7055 (Cephalon,

USA and Sanofi-Synthelabo, France); BC 1 (Genoa Institute of Cancer Research, Italy); rBPI

21 and BPI-derived antiangiogenic (XOMA, USA); PI 88 (Progen, Australia); cilengitide

(Merck KGaA, German; Munich Technical University, Germany, Scripps Clinic and Research

Foundation, USA); AVE 8062 (Ajinomoto, Japan); AS 1404 (Cancer Research Laboratory,

New Zealand); SG 292, (Telios, USA); Endostatin (Boston Childrens Hospital, USA); ATN

161 (Attenuon, USA); 2-methoxyestradiol (Boston Childrens Hospital, USA); ZD 6474,

(AstraZeneca, UK); ZD 6126, (Angiogene Pharmaceuticals, UK); PPI 2458, (Praecis, USA);

AZD 9935, (AstraZeneca, UK); AZD 2171, (AstraZeneca, UK); vatalanib (pINN), (Novartis,

Switzerland and Schering AG, Germany); tissue factor pathway inhibitors, (EntreMed, USA); pegaptanib (Pinn), (Gilead Sciences, USA); xanthorrhizol, (Yonsei University, South Korea); vaccine, gene-based, VEGF-2, (Scripps Clinic and Research Foundation, USA); SPV5.2,

(Supratek, Canada); SDX 103, (University of California at San Diego, USA); PX 478, (ProlX,

USA); METASTATIN, (EntreMed, USA); troponin I, (Harvard University, USA); SU 6668,

(SUGEN, USA); OXI 4503, (OXiGENE, USA); o-guanidines, (Dimensional Pharmaceuticals,

USA); motuporamine C, (British Columbia University, Canada); CDP 791, (Celltech Group,

UK); atiprimod (pINN), (GlaxoSmithKline, UK); E 7820, (Eisai, Japan); CYC 381, (Harvard

University, USA); AE 941, (Aeterna, Canada); vaccine, angiogenic, (EntreMed, USA); urokinase plasminogen activator inhibitor, (Dendreon, USA); oglufanide (pINN), (Melmotte,

USA); HIF-lalfa inhibitors, (Xenova, UK); CEP 5214, (Cephalon, USA); BAY RES 2622,

(Bayer, Germany); Angiocidin, (InKine, USA); A6, (Angstrom, USA); KR 31372, (Korea

Research Institute of Chemical Technology, South Korea); GW 2286, (GlaxoSmithKline, UK);

EHT 0101, (ExonHit, France); CP 868596, (Pfizer, USA); CP 564959, (OSI, USA); CP

547632, (Pfizer, USA); 786034, (GlaxoSmithKline, UK); KRN 633, (Kirin Brewery, Japan); drug delivery system, intraocular, 2-methoxyestradiol; anginex (Maastricht University,

Netherlands, and Minnesota University, USA); ABT 510 (Abbott, USA); AAL 993 (Novartis,

Switzerland); VEGI (ProteomTech, USA); tumor necrosis factor-alpha inhibitors; SU 11248

(Pfizer, USAand SUGENUSA); ABT 518, (Abbott, USA); YH16 (Yantai Rongchang, China);

S-3APG (Boston Childrens Hospital, USA and EntreMed, USA); MAb, KDR (ImClone

Systems, USA); MAb, alpha5 beta (Protein Design, USA); KDR kinase inhibitor (Celltech

Group, UK, and Johnson & Johnson, USA); GFB 116 (South Florida University, USA and Yale University, USA); CS 706 (Sankyo, Japan); combretastatin A4 prodrug (Arizona State University, USA); chondroitinase AC (IBEX, Canada); BAY RES 2690 (Bayer, Germany); AGM 1470 (Harvard University, USA, Takeda, Japan, and TAP, USA); AG 13925 (Agouron, USA); Tetrathiomolybdate (University of Michigan, USA); GCS 100 (Wayne State University, USA) CV 247 (Ivy Medical, UK); CKD 732 (Chong Kun Dang, South Korea); irsogladine, (Nippon Shinyaku, Japan); RG 13577 (Aventis, France); WX 360 (Wilex, Germany); squalamine, (Genaera, USA); RPI 4610 (Sirna, USA); heparanase inhibitors (InSight, Israel); KL 3106 (Kolon, South Korea); Honokiol (Emory University, USA); ZK CDK (Schering AG, Germany); ZK Angio (Schering AG, Germany); ZK 229561 (Novartis, Switzerland, and Schering AG, Germany); XMP 300 (XOMA, USA); VGA 1102 (Taisho, Japan); VE-cadherin- 2 antagonists(ImClone Systems, USA); Vasostatin (National Institutes of Health, USA); Flk-1 (ImClone Systems, USA); TZ 93 (Tsumura, Japan); TumStatin (Beth Israel Hospital, USA); truncated soluble FLT 1 (vascular endothelial growth factor receptor 1) (Merck & Co, USA); Tie-2 ligands (Regeneron, USA); and thrombospondin 1 inhibitor (Allegheny Health, Education and Research Foundation, USA).

[00134] Further examples of therapeutic agents that may be used in combination with compounds of the invention include agents (e.g., antibodies, antigen binding regions, or soluble receptors) that specifically bind and inhibit the activity of growth factors, such as antagonists of hepatocyte growth factor (HGF, also known as Scatter Factor), and antibodies or antigen binding regions that specifically bind its receptor, c-Met.

[00135] Another example of a therapeutic agent that may be used in combination with compounds of the invention is an autophagy inhibitor. Autophagy inhibitors include, but are not limited to chloroquine, 3- methyladenine, hydroxychloroquine (Plaquenil™), bafilomycin A1, 5-amino-4-imidazole carboxamide riboside (AICAR), okadaic acid, autophagy - suppressive algal toxins which inhibit protein phosphatases of type 2 A or type 1, analogues of cAMP, and drugs which elevate cAMP levels such as adenosine, LY204002, N6- mercaptopurine riboside, and vinblastine. In addition, antisense or siRNA that inhibits expression of proteins including but not limited to ATG5 (which are implicated in autophagy), may also be used. In some embodiments, the one or more additional therapies include an autophagy inhibitor.

[00136] Another example of a therapeutic agent that may be used in combination with compounds of the invention is an anti -neoplastic agent. In some embodiments, the one or more additional therapies include an anti-neoplastic agent. Non-limiting examples of anti -neoplastic agents include acemannan, aclarubicin, aldesleukin, alemtuzumab, alitretinoin, altretamine, amifostine, aminolevulinic acid, amrubicin, amsacrine, anagrelide, anastrozole, ancer, ancestim, arglabin, arsenic trioxide, BAM-002 (Novelos), bexarotene, bicalutamide, broxuridine, capecitabine, celmoleukin, cetrorelix, cladribine, clotrimazole, cytarabine ocfosfate, DA 3030 (Dong- A), daclizumab, denileukin diftitox, deslorelin, dexrazoxane, dilazep, docetaxel, docosanol, doxercalciferol, doxifluridine, doxorubicin, bromocriptine, carmustine, cytarabine, fluorouracil, HIT diclofenac, interferon alfa, daunorubicin, doxorubicin, tretinoin, edelfosine, edrecolomab, eflornithine, emitefur, epirubicin, epoetin beta, etoposide phosphate, exemestane, exisulind, fadrozole, filgrastim, finasteride, fludarabine phosphate, formestane, fotemustine, gallium nitrate, gemcitabine, gemtuzumab zogamicin, gimeracil/oteracil/tegafur combination, glycopine, goserelin, heptaplatin, human chorionic gonadotropin, human fetal alpha fetoprotein, ibandronic acid, idarubicin, (imiquimod, interferon alfa, interferon alfa, natural, interferon alfa-2, interferon alfa-2a, interferon alfa-2b, interferon alfa-N1, interferon alfa-n3, interferon alfacon-1, interferon alpha, natural, interferon beta, interferon beta-la, interferon beta-lb, interferon gamma, natural interferon gamma- la, interferon gamma-lb, interleukin-1 beta, iobenguane, irinotecan, irsogladine, lanreotide, LC

9018 (Yakult), leflunomide, lenograstim, lentinan sulfate, letrozole, leukocyte alpha interferon, leuprorelin, levamisole + fluorouracil, liarozole, lobaplatin, lonidamine, lovastatin, masoprocol, melarsoprol, metoclopramide, mifepristone, miltefosine, mirimostim, mismatched double stranded RNA, mitoguazone, mitolactol, mitoxantrone, molgramostim, nafarelin, naloxone + pentazocine, nartograstim, nedaplatin, nilutamide, noscapine, novel erythropoiesis stimulating protein, NSC 631570 octreotide, oprelvekin, osaterone, oxaliplatin, paclitaxel, pamidronic acid, pegaspargase, peginterferon alfa-2b, pentosan polysulfate sodium, pentostatin, picibanil, pirarubicin, rabbit antithymocyte polyclonal antibody, polyethylene glycol interferon alfa-2a, porfimer sodium, raloxifene, raltitrexed, rasburiembodiment, rhenium Re 186 etidronate, RII retinamide, rituximab, romurtide, samarium (153 Sm) lexidronam, sargramostim, sizofiran, sobuzoxane, sonermin, strontium-89 chloride, suramin, tasonermin, tazarotene, tegafur, temoporfm, temozolomide, teniposide, tetrachlorodecaoxide, thalidomide, thymalfasin, thyrotropin alfa, topotecan, toremifene, tositumomab-iodine 131, trastuzumab, treosulfan, tretinoin, trilostane, trimetrexate, triptorelin, tumor necrosis factor alpha, natural, ubenimex, bladder cancer vaccine, Maruyama vaccine, melanoma lysate vaccine, valrubicin, verteporfm, vinorelbine, virulizin, zinostatin stimalamer, or zoledronic acid; abarelix; AE 941 (Aetema), ambamustine, antisense oligonucleotide, bcl-2 (Genta), APC

8015 (Dendreon), decitabine, dexaminoglutethimide, diaziquone, EL 532 (Elan), EM 800 (Endorecherche), eniluracil, etanidazole, fenretinide, filgrastim SD01 (Amgen), fulvestrant, galocitabine, gastrin 17 immunogen, HLA-B7 gene therapy (Vical), granulocyte macrophage colony stimulating factor, histamine dihydrochloride, ibritumomab tiuxetan, ilomastat, IM 862 (Cytran), interleukin-2, iproxifene, LDI 200 (Milkhaus), leridistim, lintuzumab, CA 125 MAb (Biomira), cancer MAb (Japan Pharmaceutical Development), HER-2 and Fc MAb (Medarex), idiotypic 105AD7 MAb (CRC Technology), idiotypic CEA MAb (Trilex), LYM-1-iodine 131 MAb (Techni clone), polymorphic epithelial mucin-yttrium 90 MAb (Antisoma), marimastat, menogaril, mitumomab, motexafm gadolinium, MX 6 (Galderma), nelarabine, nolatrexed, P 30 protein, pegvisomant, pemetrexed, porfiromycin, prinomastat, RL 0903 (Shire), rubitecan, satraplatin, sodium phenyl acetate, sparfosic acid, SRL 172 (SR Pharma), SU 5416 (SUGEN), TA 077 (Tanabe), tetrathiomolybdate, thaliblastine, thrombopoietin, tin ethyl etiopurpurin, tirapazamine, cancer vaccine (Biomira), melanoma vaccine (New York Einiversity), melanoma vaccine (Sloan Kettering Institute), melanoma oncolysate vaccine (New York Medical College), viral melanoma cell lysates vaccine (Royal Newcastle Hospital), or valspodar.

[00137] Additional examples of therapeutic agents that may be used in combination with compounds of the invention include ipilimumab (Yervoy®); tremelimumab; galiximab; nivolumab, also known as BMS-936558 (Opdivo®); pembrolizumab (Keytruda®); avelumab (Bavencio®); AMP224; BMS-936559; MPDL3280A, also known as RG7446; MEDI-570; AMG557; MGA271; IMP321; BMS-663513; PF-05082566; CDX-1127; anti-OX40 (Providence Health Services); huMAbOX40L; atacicept; CP-870893; lucatumumab; dacetuzumab; muromonab-CD3; ipilumumab; MEDI4736 (Imfinzi®); MSB0010718C; AMP 224; adalimumab (Humira®); ado-trastuzumab emtansine (Kadcyla®); aflibercept (Eylea®); alemtuzumab (Campath®); basiliximab (Simulect®); belimumab (Benlysta®); basiliximab (Simulect®); belimumab (Benlysta®); brentuximab vedotin (Adcetris®); canakinumab (Ilaris®); certolizumab pegol (Cimzia®); daclizumab (Zenapax®); daratumumab (Darzalex®); denosumab (Prolia®); eculizumab (Soliris®); efalizumab (Raptiva®); gemtuzumab ozogamicin (Mylotarg®); golimumab (Simponi®); ibritumomab tiuxetan (Zevalin®); infliximab (Remicade®); motavizumab (Numax®); natalizumab (Tysabri®); obinutuzumab (Gazyva®); ofatumumab (Arzerra®); omalizumab (Xolair®); palivizumab (Synagis®); pertuzumab (Perjeta®); pertuzumab (Perjeta®); ranibizumab (Lucentis®); raxibacumab (Abthrax®); tocilizumab (Actemra®); tositumomab; tositumomab-i-131; tositumomab and tositumomab-i-131 (Bexxar®); ustekinumab (Stelara®); AMG 102; AMG 386; AMG 479; AMG 655; AMG 706; AMG 745; and AMG 951. [00138] The compounds described herein can be used in combination with the agents disclosed herein or other suitable agents, depending on the condition being treated. Hence, in some embodiments the one or more compounds of the disclosure will be co-administered with other therapies as described herein. When used in combination therapy, the compounds described herein may be administered with the second agent simultaneously or separately. This administration in combination can include simultaneous administration of the two agents in the same dosage form, simultaneous administration in separate dosage forms, and separate administration. That is, a compound described herein and any of the agents described herein can be formulated together in the same dosage form and administered simultaneously. Alternatively, a compound of the invention and any of the therapies described herein can be simultaneously administered, wherein both the agents are present in separate formulations. In another alternative, a compound of the present disclosure can be administered and followed by any of the therapies described herein, or vice versa. In some embodiments of the separate administration protocol, a compound of the invention and any of the therapies described herein are administered a few minutes apart, or a few hours apart, or a few days apart.

[00139] In some embodiments of any of the methods described herein, the first therapy (e.g., a compound of the invention) and one or more additional therapies are administered simultaneously or sequentially, in either order. The first therapeutic agent may be administered immediately, up to 1 hour, up to 2 hours, up to 3 hours, up to 4 hours, up to 5 hours, up to 6 hours, up to 7 hours, up to, 8 hours, up to 9 hours, up to 10 hours, up to 11 hours, up to 12 hours, up to 13 hours, 14 hours, up to hours 16, up to 17 hours, up 18 hours, up to 19 hours up to 20 hours, up to 21 hours, up to 22 hours, up to 23 hours, up to 24 hours, or up to 1-7, 1-14, 1-21 or 1-30 days before or after the one or more additional therapies.

[00140] The invention also features kits including (a) a pharmaceutical composition including an agent (e.g., a compound of the invention) described herein, and (b) a package insert with instructions to perform any of the methods described herein. In some embodiments, the kit includes (a) a pharmaceutical composition including an agent (e.g., a compound of the invention) described herein, (b) one or more additional therapies (e.g., non-drug treatment or therapeutic agent), and (c) a package insert with instructions to perform any of the methods described herein.

[00141] As one aspect of the present invention contemplates the treatment of the disease or symptoms associated therewith with a combination of pharmaceutically active compounds that may be administered separately, the invention further relates to combining separate pharmaceutical compositions in kit form. The kit may comprise two separate pharmaceutical compositions: a compound of the present invention, and one or more additional therapies. The kit may comprise a container for containing the separate compositions such as a divided bottle or a divided foil packet. Additional examples of containers include syringes, boxes, and bags. In some embodiments, the kit may comprise directions for the use of the separate components. The kit form is particularly advantageous when the separate components are preferably administered in different dosage forms (e.g., oral and parenteral), are administered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing health care professional.

[00142] As one of ordinary skill in the art will appreciate, in various embodiments, all of the therapeutic agents disclosed herein, i.e. , the specific bi-steric mTOR inhibitors, RAS inhibitors (e.g., KRAS(OFF) inhibitors, KRAS G12C specific inhibitors, KRAS(ON) inhibitors), TKI inhibitors, MEK inhibitors, ALK inhibitors, SHP2 inhibitors, EGFR inhibitors, etc., may be used in any one or more of the embodiments disclosed herein that call for such an inhibitor, generally. Thus, for example, an embodiment comprising treatment with, e.g. , a “bi-steric mTOR inhibitor,” generally, or a “RAS inhibitor,” generally, may comprise treatment with any one or more bi-steric mTOR inhibitor or RAS inhibitor, respectively, that is disclosed herein (unless context requires otherwise).

[00143] Administration of the disclosed compositions and compounds (e.g. , bi-steric mTOR inhibitors, RAS inhibitors (e.g, KRAS(OFF) inhibitors and/or KRAS(ON) inhibitors) and/or other therapeutic agents) can be accomplished via any mode of administration for therapeutic agents. These modes include systemic or local administration such as oral, nasal, parenteral, transdermal, subcutaneous, vaginal, buccal, rectal or topical administration modes.

[00144] Depending on the intended mode of administration, the disclosed compounds or pharmaceutical compositions can be in solid, semi-solid or liquid dosage form, such as, for example, injectables, tablets, suppositories, pills, time-release capsules, elixirs, tinctures, emulsions, syrups, powders, liquids, suspensions, or the like, sometimes in unit dosages and consistent with conventional pharmaceutical practices. Likewise, they can also be administered in intravenous (both bolus and infusion), intraperitoneal, subcutaneous or intramuscular form, and all using forms well known to those skilled in the pharmaceutical arts. Pharmaceutical compositions suitable for the delivery of a bi-steric mTOR inhibitor and a RAS inhibitor (e.g, a KRAS(OFF) inhibitor or a KRAS(ON) inhibitor) (alone or, e.g., in combination with another therapeutic agent according to the present disclosure) and methods for their preparation will be readily apparent to those skilled in the art. Such compositions and methods for their preparation may be found, e.g ., in Remington’s Pharmaceutical Sciences, 19th Edition (Mack Publishing Company, 1995), incorporated herein in its entirety.

[00145] Illustrative pharmaceutical compositions are tablets and gelatin capsules comprising a bi-steric mTOR inhibitor, a RAS inhibitor (e.g, a KRAS(OFF) inhibitor and/or a KRAS(ON) inhibitor) alone or in combination with one another and/or in combination with another therapeutic agent according to the disclosure and a pharmaceutically acceptable carrier, such as a) a diluent, e.g, purified water, triglyceride oils, such as hydrogenated or partially hydrogenated vegetable oil, or mixtures thereof, com oil, olive oil, sunflower oil, safflower oil, fish oils, such as EPA or DHA, or their esters or triglycerides or mixtures thereof, omega-3 fatty acids or derivatives thereof, lactose, dextrose, sucrose, mannitol, sorbitol, cellulose, sodium, saccharin, glucose and/or glycine; b) a lubricant, e.g, silica, talcum, stearic acid, its magnesium or calcium salt, sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and/or polyethylene glycol; for tablets also; c) a binder, e.g, magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, magnesium carbonate, natural sugars such as glucose or beta- lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, waxes and/or polyvinylpyrrolidone, if desired; d) a disintegrant, e.g, starches, agar, methyl cellulose, bentonite, xanthan gum, algiic acid or its sodium salt, or effervescent mixtures; e) absorbent, colorant, flavorant and sweetener; f) an emulsifier or dispersing agent, such as Tween 80, Labrasol, HPMC, DOSS, caproyl 909, labrafac, labrafil, peceol, transcutol, capmul MCM, capmul PG-12, captex 355, gelucire, vitamin E TGPS or other acceptable emulsifier; and/or g) an agent that enhances absorption of the compound such as cyclodextrin, hydroxypropyl-cyclodextrin, PEG400, PEG200.

[00146] Liquid, particularly injectable, compositions can, for example, be prepared by dissolution, dispersion, etc. For example, a bi-steric mTOR inhibitor, a RAS inhibitor (e.g, a KRAS(OFF) inhibitor and/or a KRAS(ON) inhibitor) alone or in combination with one another and/or in combination with another therapeutic agent according to the disclosure) is dissolved in or mixed with a pharmaceutically acceptable solvent such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like, to thereby form an injectable isotonic solution or suspension. Proteins such as albumin, chylomicron particles, or serum proteins can be used to solubilize the SHP2 inhibitor (alone or in combination with another therapeutic agent according to the disclosure). [00147] A bi-steric mTOR inhibitor and/or a RAS inhibitor (e.g., a KRAS(OFF) inhibitor and/or a KRAS(ON) inhibitor) alone or in combination with one another and/or in combination with another therapeutic agent can be also formulated as a suppository, alone or in combination with another therapeutic agent according to the disclosure, which can be prepared from fatty emulsions or suspensions; using polyalkylene glycols such as propylene glycol, as the carrier.

[00148] A bi-steric mTOR inhibitor and/or a RAS inhibitor (e.g, a KRAS(OFF) inhibitor and/or a KRAS(ON) inhibitor) alone or in combination with one another and/or in combination with another therapeutic agent can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles, either alone or in combination with another therapeutic agent according to the disclosure. Liposomes can be formed from a variety of phospholipids, containing cholesterol, stearylamine or phosphatidylcholines. In some embodiments, a film of lipid components is hydrated with an aqueous solution of drug to a form lipid layer encapsulating the drug, as described for instance in U. S. Pat. No. 5,262,564, the contents of which are hereby incorporated by reference.

[00149] A bi-steric mTOR inhibitor and/or a RAS inhibitor (e.g, a KRAS(OFF) inhibitor and/or a KRAS(ON) inhibitor) alone or in combination with one another and/or in combination with another therapeutic agent inhibitors can also be delivered by the use of monoclonal antibodies as individual carriers to which the disclosed compounds are coupled. Bi-steric mTOR inhibitor and/or the RAS inhibitor (e.g., a KRAS(OFF) inhibitor) alone or in combination with one another and/or in combination with another therapeutic agent inhibitors can also be coupled with soluble polymers as targetable drug carriers. Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol, polyhydroxyethylaspanamidephenol, or polyethyleneoxidepolylysine substituted with palmitoyl residues. Furthermore, a bi-steric mTOR inhibitor and/or a RAS inhibitor (e.g, a KRAS(OFF) inhibitor and/or a KRAS(ON) inhibitor) can be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross-linked or amphipathic block copolymers of hydrogels. In some embodiments, disclosed compounds are not covalently bound to a polymer, e.g, a poly carboxylic acid polymer, or a polyacrylate.

[00150] Parental inj ectable administration is generally used for subcutaneous, intramuscular or intravenous injections and infusions. Injectables can be prepared in conventional forms, either as liquid solutions or suspensions or solid forms suitable for dissolving in liquid prior to injection.

[00151] Another aspect of the invention relates to a pharmaceutical composition comprising a bi-steric mTOR inhibitor and/or the RAS inhibitor (e.g., a KRAS(OFF) inhibitor) alone or in combination with one another and/or in combination with another therapeutic agent according to the present disclosure and a pharmaceutically acceptable carrier. The pharmaceutically acceptable carrier can further include an excipient, diluent, or surfactant.

[00152] Thus, the present disclosure provides compositions (e.g, pharmaceutical compositions) comprising one or more bi-steric mTOR inhibitors for use in a method disclosed herein. Such compositions may comprise a bi-steric mTOR inhibitors inhibitor and, e.g, one or more carrier, excipient, diluent, and/or surfactant. The present disclosure provides compositions (e.g, pharmaceutical compositions) comprising one or more RAS inhibitors (e.g, a KRAS(OFF) inhibitor) for use in a method disclosed herein. Such compositions may comprise a RAS inhibitor (e.g, a KRAS(OFF) inhibitor) and, e.g, one or more carrier, excipient, diluent, and/or surfactant. The present disclosure provides compositions (e.g, pharmaceutical compositions) comprising one or more bi-steric mTOR inhibitors and one or more RAS inhibitors (e.g, a KRAS(OFF) inhibitor) for use in a method disclosed herein. Such compositions may comprise one or more bi-steric mTOR inhibitors inhibitor and one or more RAS inhibitor (e.g, a KRAS(OFF) inhibitor) e.g, one or more carrier, excipient, diluent, and/or surfactant. Such compositions may also comprise one or more additional therapeutic agent for use in a method disclosed herein, such as, e.g, a SHP2 inhibitor, a TKI, a MAPK pathway inhibitor, an EGFR inhibitor, an ALK inhibitor, and or a MEK inhibitor and, e.g. , one or more carrier, excipient, diluent, and/or surfactant.

[00153] Compositions can be prepared according to conventional mixing, granulating or coating methods, respectively, and the present pharmaceutical compositions can contain from about 0.1% to about 99%, from about 5% to about 90%, or from about 1% to about 20% of the disclosed Compound By weight or volume.

[00154] The dosage regimen utilizing the disclosed compound is selected in accordance with a variety of factors including type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal or hepatic function of the patient; and the particular disclosed compound employed. A physician or veterinarian of ordinary skill in the art can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the condition.

[00155] Effective dosage amounts of a bi-steric mTOR inhibitor, when used for the indicated effects, range from about 0.1 mg to about 1000 mg as needed to treat the condition. Compositions for in vivo or in vitro use can contain about 0.1, 0.2, 0.3, 0.4, 0.5, 5, 20, 50, 75, 100, 150, 250, 500, 750, or 1000 mg of the disclosed compound, or, in a range of from one amount to another amount in the list of doses. In some embodiments, compositions for in vivo or in vitro use contain from 0.5 mg to 500 mg (e.g., from about 1 mg to about 400 mg). In some embodiments, the compositions are in the form of an intravenous solution.

[00156] Effective dosage amounts of an ALK inhibitor, when used for the indicated effects, range from about 0.5 mg to about 5000 mg as needed to treat the condition. Compositions for in vivo or in vitro use can contain about 0.5, 5, 20, 50, 75, 100, 150, 250, 500, 750, 1000, 1250, 2500, 3500, or 5000 mg of the disclosed compound, or, in a range of from one amount to another amount in the list of doses. In some embodiments, the compositions are in the form of a tablet that can be scored.

[00157] Effective dosage amounts of an EGFR inhibitor, when used for the indicated effects, range from about 0.5 mg to about 5000 mg as needed to treat the condition. Compositions for in vivo or in vitro use can contain about 0.5, 5, 20, 50, 75, 100, 150, 250, 500, 750, 1000, 1250, 2500, 3500, or 5000 mg of the disclosed compound, or, in a range of from one amount to another amount in the list of doses. In some embodiments, the compositions are in the form of a tablet that can be scored.

[00158] Effective dosage amounts of an MEK inhibitor, when used for the indicated effects, range from about 0.05 mg to about 5000 mg as needed to treat the condition. Compositions for in vivo or in vitro use can contain about 0.05, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 5, 20, 50, 75, 100, 150, 250, 500, 750, 1000, 1250, 2500, 3500, or 5000 mg of the disclosed compound, or, in a range of from one amount to another amount in the list of doses. In some embodiments, the compositions are in the form of a tablet that can be scored.

[00159] The present invention also provides kits for treating a disease or disorder with a bi- steric mTOR inhibitor and optionally a RAS inhibitor (e.g, a KRAS(OFF) inhibitor and/or a KRAS(ON) inhibitor), one or more carrier, excipient, diluent, and/or surfactant, and a means for determining whether a sample from a subject (e.g, a tumor sample) is likely to be sensitive to such a bi-steric mTOR and/or RAS inhibitor treatment. In some embodiments, the means for determining comprises a means for determining whether the sample comprises a RAS mutation, e.g ., a NRAS, KRAS, or HRAS mutation. Such mutations may comprise a G12C mutation. Such mutations may be selected from a KRAS G12C mutation, a KRAS G12D mutation, a KRAS G12S mutation, and/or a KRAS G12V mutation. Such means include, but are not limited to direct sequencing, and utilization of a high-sensitivity diagnostic assay (with CE-IVD mark), e.g., as described in Domagala, et al., Pol J Pathol 3: 145-164 (2012), incorporated herein by reference in its entirety, including TheraScreen PCR; AmoyDx; PNAClamp; RealQuality; EntroGen; LightMix; StripAssay; Hybcell plexA; Devyser; Surveyor; Cobas; and TheraScreen Pyro.

[00160] Methods for detecting a mutation in a KRAS, HRAS or NRAS nucleotide sequence are known by those of skill in the art. These methods include, but are not limited to, polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) assays, polymerase chain reaction-single strand conformation polymorphism (PCR-SSCP) assays, real-time PCR assays, PCR sequencing, mutant allele-specific PCR amplification (MASA) assays, direct sequencing, primer extension reactions, electrophoresis, oligonucleotide ligation assays, hybridization assays, TaqMan assays, SNP genotyping assays, high resolution melting assays and microarray analyses. In some embodiments, samples are evaluated for G12C KRAS, HRAS or NRAS mutations by real-time PCR. In real-time PCR, fluorescent probes specific for the KRAS, HRAS or NRAS G12C mutation are used. When a mutation is present, the probe binds and fluorescence is detected. In some embodiments, the KRAS, HRAS or NRAS G12C mutation is identified using a direct sequencing method of specific regions (e.g, exon 2 and/or exon 3) in the KRAS, HRAS or NRAS gene. This technique will identify all possible mutations in the region sequenced.

[00161] Methods for detecting a mutation in a KRAS, HRAS or NRAS protein are known by those of skill in the art. These methods include, but are not limited to, detection of a KRAS, HRAS or NRAS mutant using a binding agent (e.g, an antibody) specific for the mutant protein, protein electrophoresis and Western blotting, and direct peptide sequencing.

[00162] Methods for determining whether a tumor or cancer comprises a G12C or other KRAS, HRAS or NRAS mutation can use a variety of samples. In some embodiments, the sample is taken from a subject having a tumor or cancer. In some embodiments, the sample is a fresh tumor/cancer sample. In some embodiments, the sample is a frozen tumor/cancer sample. In some embodiments, the sample is a formalin-fixed paraffin-embedded sample. In some embodiments, the sample is a circulating tumor cell (CTC) sample. In some embodiments, the sample is processed to a cell lysate. In some embodiments, the sample is processed to DNA or RNA.

Exemplary Embodiments

Some embodiments of this disclosure are in the Embodiments, as follows:

Embodiment I-1. A method for delaying or preventing acquired resistance to a RAS inhibitor in a subject in need thereof, comprising administering to the subject an effective amount of a bi-steric inhibitor of mTOR, wherein the subject has already received or will receive administration of the RAS inhibitor, wherein the effective amount is an amount effective to delay or prevent acquired resistance to the RAS inhibitor in a subject in need thereof.

Embodiment I-2. A method of treating acquired resistance to a RAS inhibitor in a subject in need thereof, comprising administering to the subject an effective amount of a bi-steric inhibitor of mTOR, wherein the effective amount is an amount effective to treat acquired resistance to the RAS inhibitor in a subject in need thereof.

Embodiment I-3. The method of Embodiment I-1 or I-2, wherein the RAS is selected from KRAS, NRAS, and HRAS.

Embodiment I-4. The method of any one of Embodiments I-1 to I-3, further comprising administering to the subject an effective amount of the RAS inhibitor.

Embodiment I-5. The method of any one of Embodiments I-1 to I-4 wherein the RAS inhibitor targets a specific RAS mutation.

Embodiment I-6. The method of any one of Embodiments I-1 to I-5, wherein the RAS inhibitor targets a KRAS mutation.

Embodiment I-7. The method of any one of Embodiments I-1 to I-6, wherein the RAS inhibitor targets a G12C mutation.

Embodiment I-8. The method of any one of Embodiments I-1 to I-7, wherein the RAS inhibitor targets the KRAS G12C mutation.

Embodiment I-9. The method of any one of Embodiments I-1 to I-8, wherein the RAS inhibitor binds the RAS in its “off’ position. Embodiment I-10. The method of any one of Embodiments I-6 to I-9, wherein the RAS inhibitor is a KRAS(OFF) inhibitor.

Embodiment I-11. The method of any one of Embodiments I-1 to I-6 or Embodiments I-9 to I-10, wherein the RAS inhibitor targets a KRAS mutation selected from a KRAS G12A mutation, a KRAS G12D mutation, a KRAS G12F mutation, a KRAS G12I mutation, a KRAS G12L mutation, a KRAS G12R mutation, a KRAS G12S mutation, a KRAS G12V mutation, and a KRAS G12Y mutation.

Embodiment I-12. The method of any one of Embodiments I-1 to I-11, wherein the KRAS inhibitor is selected from AMG 510, MRTX849, JDQ443 and MRTX1133, or a pharmaceutically acceptable salt thereof.

Embodiment I-13. The method of any one of the preceding Embodiments, wherein the bi- steric inhibitor of mTOR is RM-006, also known as RMC-6272, or RMC-5552, or a pharmaceutically acceptable salt thereof.

Embodiment I-14. The method of any one of Embodiments I-1 to I-12, wherein the bi-steric inhibitor of mTOR is a compound having the formula or a stereoisomer thereof.

Embodiment I-15. The method of any one of Embodiments I-1 to I-12, wherein the bi-steric inhibitor of mTOR is a compound having the formula or a tautomer thereof.

Embodiment I-16. The method of any one of Embodiments I-1 to I-12, wherein the bi-steric inhibitor of mTOR is a compound having the formula or an oxepane isomer thereof.

Embodiment I-17. The method of any one of Embodiments I-1 to I-12, wherein the bi-steric inhibitor of mTOR is a compound having the formula or a stereoisomer thereof.

Embodiment I-18. The method of any one of Embodiments I-1 to I-12, wherein the bi-steric inhibitor of mTOR is a compound having the formula

or a tautomer thereof.

Embodiment I-19. The method of any one of Embodiments I-1 to I-12, wherein the bi-steric inhibitor of mTOR is a compound having the formula

Embodiment I-20. The method of any one of Embodiments I-1 to I-12, wherein the bi-steric inhibitor of mTOR is a compound having the formula

Embodiment I-21. The method of any one of Embodiments I-1 to I-12, wherein the bi-steric inhibitor of mTOR is comprised in a composition comprising a compound having the formula or a stereoisomer or tautomer thereof and a compound having the formula or a stereoisomer or tautomer thereof.

Embodiment I-22. The method of any one of Embodiments I-1 to I-12, wherein the bi-steric inhibitor of mTOR is comprised in a composition comprising a compound having the formula and

Embodiment I-23. The method of any one of Embodiments I-1 to I-8, Embodiment 11, or Embodiments I-13 to I-22, wherein the RAS inhibitor binds the RAS in its “on” position.

Embodiment I-24. The method of any one of Embodiments I-1 to I-8, Embodiment 11, or Embodiments I-13 to I-23, wherein the RAS inhibitor is a KRAS(ON) inhibitor.

Embodiment I-25. The method of Embodiment I-24, wherein the KRAS(ON) inhibitor is a KRAS G12C (ON) inhibitor.

Embodiment I-26. The method of any one of Embodiments I-1 to I-8, Embodiment I-11, or Embodiments I-13 to I-25, wherein the RAS inhibitor is selected from compounds A1-A741 of Appendix B-1, or a pharmaceutically acceptable salt thereof.

Embodiment I-27. The method of any one of Embodiments I-1 to I-8, Embodiment I-11, or Embodiments I-13 to I-25, wherein the RAS inhibitor is a compound, or a pharmaceutically acceptable salt thereof, of Appendix B-1, Formula VIb, Formula VIb wherein A is a 3 to 6-membered heterocycloalkylene, a phenylene, or a hydroxy-substituted phenylene; B is -CH(C 1 -C 6 alkyl)-; L is a linker selected from the following: ; and

W is a cross-linking group selected from the following:

Embodiment I-28. The method of any one of Embodiments I-1 to I-8, Embodiment I-11, or Embodiments I-13 to I-27, wherein the RAS inhibitor is selected from compounds A121, A131, A133, A145, A150, A173, A182, A191, A198, A199, A201, A244, A245, A246, A247, A248, A266, A290, A292, A310, A316, A317, A324, A325, A326, A337, A339, A351, A365, A377, A391, A402, A412, A413, A414, A426, A476, A487, A499, A508, A509, A526, A528, A532, A533, A534, A551, A559, A560, A565, A566, A567, A568, A569, A584, A585, A591, A592, A599, A601, A613, A614, A615, A616, A617, A643, A644, A646, A647, A648, A657, A663, A672, A699, A708, A715, A717 and A733 of Appendix B-1, or a pharmaceutically acceptable salt thereof.

Embodiment I-29. The method of any one of Embodiments I-1 to I-8, Embodiment I-11, or Embodiments I-13 to I-28, wherein the RAS inhibitor is Compound A, or a pharmaceutically acceptable salt thereof.

Embodiment I-30. The method of any one of Embodiments I-1 to I-8, Embodiment I-11, or Embodiments I-13 to I-28, wherein the RAS inhibitor is Compound B, or a pharmaceutically acceptable salt thereof. Embodiment I-31. The method of any one of the preceding Embodiments, wherein the subject is administered the RAS inhibitor to treat or prevent a cancer.

Embodiment I-32. The method of Embodiment I-31, wherein the cancer is a RAS G12C cancer.

Embodiment I-33. The method of Embodiment I-31 or Embodiment I-32, wherein the cancer comprises a KRAS G12C mutation.

Embodiment I-34. The method of any one of Embodiments I-31 to I-33, wherein the cancer comprises co-occurring KRAS G12C and STK11 mutations.

Embodiment I-35. The method of any one of Embodiments I-31 to I-34, wherein the cancer is a Non-Small Cell Lung Cancer (NSCLC).

Embodiment I-36. The method of any one of Embodiments I-31 to I-34, wherein the cancer is a colorectal cancer.

Embodiment I-37. The method of any one of Embodiments I-31 to I-36, wherein the cancer is selected from pancreatic cancer, colorectal cancer, non-small cell lung cancer, squamous cell lung carcinoma, thyroid gland adenocarcinoma, and a hematological cancer.

Embodiment I-38. The method of any one of Embodiments I-31 to I-37, wherein the cancer comprises co-occurring KRAS G12C and PIK3CA E545K mutations.

Embodiment I-39. The method of Embodiment I-37 or Embodiment I-38, wherein the cancer is a colorectal cancer.

Embodiment I-40. The method of any one of Embodiments I-31 to I-39, wherein the method results in tumor regression.

Embodiment I-41. The method of any one of Embodiments I-31 to I-40, wherein the method results in tumor apoptosis.

Embodiment I-42. A method of treating a subject having a cancer comprising administering to the subject an effective amount of a bi-steric inhibitor of mTOR in combination with a RAS inhibitor.

Embodiment I-43. The method of Embodiment I-42, wherein the RAS is selected from KRAS, NRAS, and HRAS. Embodiment I-44. The method of Embodiment I-42 or Embodiment I-43, wherein the RAS inhibitor targets a specific RAS mutation.

Embodiment I-45. The method of any one of Embodiments I-42 to I-44, wherein the RAS inhibitor targets a KRAS mutation.

Embodiment I-46. The method of any one of Embodiments I-42 to I-45, wherein the RAS inhibitor targets a RAS G12C mutation.

Embodiment I-47. The method of any one of Embodiments I-42 to I-46, wherein the RAS inhibitor targets the KRAS G12C mutation.

Embodiment I-48. The method of any one of Embodiments I-42 to I-47, wherein the RAS inhibitor binds the RAS in its “off’ position.

Embodiment I-49. The method of any one of Embodiments I-42 to I-48, wherein the RAS inhibitor is a KRAS(OFF) inhibitor.

Embodiment I-50. The method of any one of Embodiments I-42 to I-45 or Embodiments I- 48 or Embodiment I-49, wherein the KRAS inhibitor targets a KRAS mutation selected from a KRAS G12A mutation, a KRAS G12D mutation, a KRAS G12F mutation, a KRAS G12I mutation, a KRAS G12L mutation, a KRAS G12R mutation, a KRAS G12S mutation, a KRAS G12V mutation, and a KRAS G12Y mutation.

Embodiment I-51. The method of any one of Embodiments I-42 to I-50, wherein the KRAS inhibitor is selected from AMG 510, MRTX849, JDQ443 and MRTX1133, or a pharmaceutically acceptable salt thereof.

Embodiment I-52. The method of one of Embodiments I-42 to I-51, wherein the bi-steric inhibitor of mTOR is RM-006, also known as RMC-6272, or RMC-5552, or a pharmaceutically acceptable salt thereof.

Embodiment I-53. The method of any one of Embodiments I-42 to I-51, wherein the bi-steric inhibitor of mTOR is a compound having the formula or a stereoisomer thereof.

Embodiment I-54. The method of any one of Embodiments I-42 to I-51, wherein the bi-steric inhibitor of mTOR is a compound having the formula or a tautomer thereof.

Embodiment I-55. The method of any one of Embodiments I-42 to I-51, wherein the bi-steric inhibitor of mTOR is a compound having the formula or an oxepane isomer thereof. Embodiment I-56. The method of any one of Embodiments I-42 to I-51, wherein the bi-steric inhibitor of mTOR is a compound having the formula or a stereoisomer thereof.

Embodiment I-57. The method of any one of Embodiments I-42 to I-51, wherein the bi-steric inhibitor of mTOR is a compound having the formula or a tautomer thereof.

Embodiment I-58. The method of any one of Embodiments I-42 to I-51, wherein the bi- steric inhibitor of mTOR is a compound having the formula Embodiment I-59. The method of any one of Embodiments I-42 to I-51, wherein the bi- steric inhibitor of mTOR is a compound having the formula

Embodiment I-60. The method of any one of Embodiments I-42 to I-51, wherein the bi-steric inhibitor of mTOR is comprised in a composition comprising a compound having the formula or a stereoisomer or tautomer thereof and a compound having the formula or a stereoisomer or tautomer thereof. Embodiment I-61. The method of any one of Embodiments I-42 to I-51, wherein the bi-steric inhibitor of mTOR is comprised in a composition comprising a compound having the formula

Embodiment I-62. The method of any one of Embodiments I-42 to I-47, Embodiment I-50, or Embodiments I-52 to I-61, wherein the RAS inhibitor binds the RAS in its “on” position.

Embodiment I-63. The method of Embodiment I-62, wherein the RAS inhibitor is a KRAS(ON) inhibitor.

Embodiment I-64. The method of Embodiment I-63, wherein the KRAS(ON) inhibitor is a KRAS G12C (ON) inhibitor.

Embodiment I-65. The method of any one of Embodiments I-42 to I-47, Embodiment I-50, or Embodiments I-52 to I-64, wherein the RAS inhibitor is selected from compounds A1- A741 of Appendix B-1, or a pharmaceutically acceptable salt thereof.

Embodiment I-66. The method of any one of Embodiments I-42 to I-47, Embodiment I-50 or Embodiments I-52 to I-64, wherein the RAS inhibitor is a compound, or a pharmaceutically acceptable salt thereof, of Appendix B-1, Formula VIb,

Formula VIb wherein A is a 3 to 6-membered heterocycloalkylene, a phenylene, or a hydroxy-substituted phenylene; B is -CH(C 1 -C 6 alkyl)-; L is a linker selected from the following:

W is a cross-linking group selected from the following:

Embodiment I-67. The method of any one of Embodiments I-42 to I-47, Embodiment I-50 or Embodiments I-52 to I-66, wherein the RAS inhibitor is selected from compounds A121, A131, A133, A145, A150, A173, A182, A191, A198, A199, A201, A244, A245, A246,

A247, A248, A266, A290, A292, A310, A316, A317, A324, A325, A326, A337, A339,

A351, A365, A377, A391, A402, A412, A413, A414, A426, A476, A487, A499, A508,

A509, A526, A528, A532, A533, A534, A551, A559, A560, A565, A566, A567, A568,

A569, A584, A585, A591, A592, A599, A601, A613, A614, A615, A616, A617, A643,

A644, A646, A647, A648, A657, A663, A672, A699, A708, A715, A717 and A733 of

Appendix B-1, or a pharmaceutically acceptable salt thereof. Embodiment I-68. The method of any one of Embodiments I-42 to I-47, Embodiments I-50 or Embodiments I-52 to I-67, wherein the RAS inhibitor is Compound A, or a pharmaceutically acceptable salt thereof.

Embodiment I-69. The method of any one of Embodiments I-42 to I-47, Embodiments I-50 or Embodiments I-52 to I-67, wherein the RAS inhibitor is Compound B, or a pharmaceutically acceptable salt thereof.

Embodiment I-70. The method of any one of Embodiments I-42 to I-49 or Embodiments I- 51 to I-69, wherein the cancer is a RAS G12C cancer.

Embodiment I-71. The method of any one of Embodiments I-42 to I-70, wherein the cancer comprises a KRAS G12C mutation.

Embodiment I-72. The method of any one of Embodiments I-42 to I-71, wherein the cancer comprises co-occurring KRAS G12C and STK11 mutations.

Embodiment I-73. The method of any one of c Embodiments I-42 to I-71, wherein the cancer is a Non-Small Cell Lung Cancer (NSCLC).

Embodiment I-74. The method of any one of Embodiments I-42 to I-72, wherein the cancer is a colorectal cancer.

Embodiment I-75. The method of any one of Embodiments I-42 to I-74, wherein the cancer is selected from pancreatic cancer, colorectal cancer, non-small cell lung cancer, squamous cell lung carcinoma, thyroid gland adenocarcinoma, and a hematological cancer.

Embodiment I-76. The method of any one of Embodiments I-42 to I-75, wherein the cancer comprises co-occurring KRAS G12C and PIK3CA E545K mutations.

Embodiment I-77. The method of any one of Embodiments I-42 to I-72 or Embodiments I- 74 to I-76, wherein the cancer is a colorectal cancer.

Embodiment I-78. The method of any one of Embodiments I-42 to I-77, wherein the method results in tumor regression.

Embodiment I-79. The method of any one of Embodiments I-42 to I-78, wherein the method results in tumor apoptosis. Embodiment I-80. A method of inducing apoptosis of a tumor cell comprising contacting the tumor cell with an effective amount of a bi-steric inhibitor of mTOR in combination with a RAS inhibitor, wherein the effective amount is an amount effective to induce apoptosis of the tumor cell.

Embodiment I-81. The method of Embodiment I-80, wherein the RAS is selected from KRAS, NRAS, and HRAS.

Embodiment I-82. The method of Embodiment I-80 or Embodiment I-81, wherein the RAS inhibitor targets a specific RAS mutation.

Embodiment I-83. The method of any one of Embodiments I-80 to I-82, wherein the RAS inhibitor targets a KRAS mutation.

Embodiment I-84. The method of any one of Embodiments I-80 to I-83, wherein the RAS inhibitor targets a RAS G12C mutation.

Embodiment I-85. The method of any one of Embodiments I-80 to I-84, wherein the RAS inhibitor targets the KRAS G12C mutation.

Embodiment I-86. The method of any one of Embodiments I-80 to I-85, wherein the RAS inhibitor binds the RAS in its “off’ position.

Embodiment I-87. The method of any one of Embodiments I-80 to I-86, wherein the RAS inhibitor is a KRAS(OFF) inhibitor.

Embodiment I-88. The method of any one of Embodiments I-80 to I-84 or Embodiments I- 85 to I-87, wherein the KRAS inhibitor targets a KRAS mutation selected from a KRAS G12A mutation, a KRAS G12D mutation, a KRAS G12F mutation, a KRAS G12I mutation, a KRAS G12L mutation, a KRAS G12R mutation, a KRAS G12S mutation, a KRAS G12V mutation, and a KRAS G12Y mutation.

Embodiment I-89. The method of any one of Embodiments I-80 to I-88, wherein the KRAS inhibitor is selected from AMG 510, MRTX849, JDQ443 and MRTX1133, or a pharmaceutically acceptable salt thereof.

Embodiment I-90. The method of one of Embodiments I-80 to I-89, wherein the inhibitor of mTOR is RM-006, also known as RMC-6272, or RMC-5552, or a pharmaceutically acceptable salt thereof. Embodiment I-91. The method of any one of Embodiments I-80 to I-89, wherein the bi-steric inhibitor of mTOR is a compound having the formula or a stereoisomer thereof.

Embodiment I-92. The method of any one of Embodiments I-80 to I-88, wherein the bi-steric inhibitor of mTOR is a compound having the formula or a tautomer thereof.

Embodiment I-93. The method of any one of Embodiments I-80 to I-88, wherein the bi-steric inhibitor of mTOR is a compound having the formula or an oxepane isomer thereof.

Embodiment I-94. The method of any one of Embodiments I-80 to I-88, wherein the bi-steric inhibitor of mTOR is a compound having the formula or a stereoisomer thereof.

Embodiment I-95. The method of any one of Embodiments I-80 to I-88, wherein the bi-steric inhibitor of mTOR is a compound having the formula or a tautomer thereof.

Embodiment I-96. The method of any one of Embodiments I-80 to I-88, wherein the bi-steric inhibitor of mTOR is a compound having the formula Embodiment I-97. The method of any one of Embodiments I-80 to I-88, wherein the bi-steric inhibitor of mTOR is a compound having the formula

Embodiment I-98. The method of any one of Embodiments I-80 to I-88, wherein the bi-steric inhibitor of mTOR is comprised in a composition comprising a compound having the formula or a stereoisomer or tautomer thereof and a compound having the formula or a stereoisomer or tautomer thereof. Embodiment I-99. The method of any one of Embodiments I-80 to I-88, wherein the bi-steric inhibitor of mTOR is comprised in a composition comprising a compound having the formula

Embodiment I-100. The method of any one of Embodiments I-80 to I-85, Embodiment I-88 or Embodiments I-90 to I-99, wherein the RAS inhibitor binds the RAS in its “on” position.

Embodiment I-101. The method of Embodiment I-100, wherein the RAS inhibitor is a KRAS(ON) inhibitor.

Embodiment I-102. The method of Embodiment I-101 , wherein the KRAS(ON) inhibitor is a KRAS G12C (ON) inhibitor.

Embodiment I-103. The method of any one of Embodiments I-80 to I-85, Embodiment I-88 or Embodiments I-90 to I-102, wherein the RAS inhibitor is selected from compounds A1- A741 of Appendix B-1, or a pharmaceutically acceptable salt thereof.

Embodiment I-104. The method of any one of Embodiments I-80 to I-85, Embodiment I-88 or Embodiments I-90 to I-102, wherein the RAS inhibitor is a compound, or a pharmaceutically acceptable salt thereof, of Appendix B-1, Formula VIb,

Formula VIb wherein A is a 3 to 6-membered heterocycloalkylene, a phenylene, or a hydroxy-substituted phenylene; B is -CH(C 1 -C 6 alkyl)-; L is a linker selected from the following:

W is a cross-linking group selected from the following:

Embodiment I-105. The method of any one of Embodiments I-80 to I-85, Embodiment I-88 or Embodiments I-90 to I-104, wherein the RAS inhibitor is selected from compound A121, A131, A133, A145, A150, A173, A182, A191, A198, A199, A201, A244, A245, A246, A247, A248, A266, A290, A292, A310, A316, A317, A324, A325, A326, A337, A339, A351, A365, A377, A391, A402, A412, A413, A414, A426, A476, A487, A499, A508, A509, A526, A528, A532, A533, A534, A551, A559, A560, A565, A566, A567, A568, A569, A584, A585, A591, A592, A599, A601, A613, A614, A615, A616, A617, A643, A644, A646, A647, A648, A657, A663, A672, A699, A708, A715, A717 and A733 of

Appendix B-1, or a pharmaceutically acceptable salt thereof. Embodiment I-106. The method of any one of Embodiments I-80 to I-85, Embodiment I-88 or Embodiments I-90 to I-105, wherein the RAS inhibitor is Compound A, or a pharmaceutically acceptable salt thereof.

Embodiment I-107. The method of any one of Embodiments I-80 to I-85, Embodiment I-88 or Embodiments I-90 to I-107, wherein the RAS inhibitor is Compound B, or a pharmaceutically acceptable salt thereof.

Embodiment I-108. The method of any one of Embodiments I-80 to I-107, wherein the tumor is caused by a cancer.

Embodiment I-109. The method of any one of Embodiments I-80 to I-83, Embodiments I-86 to I-87, or Embodiments I-89 to I-107, wherein the cancer is a RAS G12C cancer.

Embodiment I-110. The method of any one of Embodiments I-80 to I-109, wherein the cancer comprises a KRAS G12C mutation.

Embodiment I-111. The method of any one of Embodiments I-80 to I-110, wherein the cancer comprises co-occurring KRAS G12C and STK11 mutations.

Embodiment I-112. The method of any one of Embodiments I-80 to I-110, wherein the cancer is a Non-Small Cell Lung Cancer (NSCLC).

Embodiment I-113. The method of any one of Embodiments I-80 to I-111, wherein the cancer is a colorectal cancer.

Embodiment I-114. The method of any one of Embodiments 1-80 to I-113, wherein the cancer is selected from pancreatic cancer, colorectal cancer, non-small cell lung cancer, squamous cell lung carcinoma, thyroid gland adenocarcinoma, and a hematological cancer.

Embodiment I-115. The method of any one of Embodiments 1-80 to I-114, wherein the cancer comprises co-occurring KRAS G12C and PIK3CA E545K mutations.

Embodiment I-116. The method of any one of Embodiments 1-80 to I-111 or Embodiments I-113 to I-115, wherein the cancer is a colorectal cancer.

Embodiment I-117. The method of any one of Embodiments 1-80 to I-116, wherein the method results in tumor regression. Embodiment I-118. The method of any one of Embodiments I-1 to I-117, wherein the method results in an improved lifespan for the subject as compared to the lifespan of a similar subject that has not received a treatment with the RAS inhibitor and the bi-steric mTOR inhibitor.

Embodiment II-1. A method for delaying or preventing acquired resistance to AMG 510 or MRTX849, or a pharmaceutically acceptable salt thereof, in a subject having a RAS G12C mutated NSCLC or colorectal cancer, comprising administering to the subject an effective amount of RMC-5552, or a stereoisomer or tautomer thereof, wherein the subject has already received or will receive administration of AMG 510 or MRTX849, or a pharmaceutically acceptable salt thereof, wherein the effective amount is an amount effective to delay or prevent acquired resistance to AMG 510 or MRTX849, or a pharmaceutically acceptable salt thereof, in the subject.

Embodiment II-2. A method for delaying or preventing acquired resistance to a compound of Formula IVb of Appendix B-1, or a pharmaceutically acceptable salt thereof: Formula VIb wherein A is a 3 to 6-membered heterocycloalkylene, a phenylene, or a hydroxy-substituted phenylene; B is -CH(C 1 -C 6 alkyl)-; L is a linker selected from the following:

W is a cross-linking group selected from the following:

RAS G12C mutated NSCLC or colorectal cancer, comprising administering to the subject an effective amount of RMC-5552, or a stereoisomer or tautomer thereof, wherein the subject has already received or will receive administration of the compound, or a pharmaceutically acceptable salt thereof, wherein the effective amount is an amount effective to delay or prevent acquired resistance to the compound, or a pharmaceutically acceptable salt thereof, in the subject.

Embodiment II-3. A method for delaying or preventing acquired resistance to a compound selected from compound A121, A131, A133, A145, A150, A173, A182, A191, A198, A199,

A201, A244, A245, A246, A247, A248, A266, A290, A292, A310, A316, A317, A324,

A325, A326, A337, A339, A351, A365, A377, A391, A402, A412, A413, A414, A426,

A476, A487, A499, A508, A509, A526, A528, A532, A533, A534, A551, A559, A560,

A565, A566, A567, A568, A569, A584, A585, A591, A592, A599, A601, A613, A614,

A615, A616, A617, A643, A644, A646, A647, A648, A657, A663, A672, A699, A708,

A715, A717 and A733 of Appendix B-1, or a pharmaceutically acceptable salt thereof, in a subject having a RAS G12C mutated NSCLC or colorectal cancer, comprising administering to the subject an effective amount of RMC-5552, or a stereoisomer or tautomer thereof, wherein the subject has already received or will receive administration of the compound, or a pharmaceutically acceptable salt thereof, wherein the effective amount is an amount effective to delay or prevent acquired resistance to the compound, or a pharmaceutically acceptable salt thereof, in the subject.

Embodiment II-4. A method for delaying or preventing acquired resistance to Compound A, or a pharmaceutically acceptable salt thereof, in a subject having a RAS G12C mutated NSCLC or colorectal cancer, comprising administering to the subject an effective amount of RMC- 5552, or a stereoisomer or tautomer thereof, wherein the subject has already received or will receive administration of Compound A, or a pharmaceutically acceptable salt thereof, wherein the effective amount is an amount effective to delay or prevent acquired resistance to Compound B, or a pharmaceutically acceptable salt thereof, in the subject.

Embodiment II-5. A method for delaying or preventing acquired resistance to Compound B, or a pharmaceutically acceptable salt thereof, in a subject having a RAS G12C mutated NSCLC or colorectal cancer, comprising administering to the subject an effective amount of RMC- 5552, or a stereoisomer or tautomer thereof, wherein the subject has already received or will receive administration of Compound B, or a pharmaceutically acceptable salt thereof, wherein the effective amount is an amount effective to delay or prevent acquired resistance to Compound B, or a pharmaceutically acceptable salt thereof, in the subject.

Embodiment III-1. A method of treating acquired resistance to AMG 510 or MRTX849, or a pharmaceutically acceptable salt thereof, in a subject having a RAS G12C mutated NSCLC or colorectal cancer, comprising administering to the subject an effective amount of RMC-5552, or a stereoisomer or tautomer thereof, wherein the effective amount is an amount effective to treat acquired resistance to AMG 510 or MRTX849, or a pharmaceutically acceptable salt thereof, in the subject.

Embodiment III-2. A method of treating acquired resistance to a compound of Formula IVb of Appendix B-1, or a pharmaceutically acceptable salt thereof:

Formula VIb wherein A is a 3 to 6-membered heterocycloalkylene, a phenylene, or a hydroxy-substituted phenylene; B is -CH(C 1 -C 6 alkyl)-; L is a linker selected from the following:

W is a cross-linking group selected from the following: a

RAS G12C mutated NSCLC or colorectal cancer, comprising administering to the subject an effective amount of RMC-5552, or a stereoisomer or tautomer thereof, wherein the effective amount is an amount effective to treat acquired resistance to the compound, or a pharmaceutically acceptable salt thereof, in the subject.

Embodiment III-3. A method of treating acquired resistance to a compound selected from compound A121, A131, A133, A145, A150, A173, A182, A191, A198, A199, A201, A244, A245, A246, A247, A248, A266, A290, A292, A310, A316, A317, A324, A325, A326,

A337, A339, A351, A365, A377, A391, A402, A412, A413, A414, A426, A476, A487,

A499, A508, A509, A526, A528, A532, A533, A534, A551, A559, A560, A565, A566, A567, A568, A569, A584, A585, A591, A592, A599, A601, A613, A614, A615, A616,

A617, A643, A644, A646, A647, A648, A657, A663, A672, A699, A708, A715, A717 and A733 of Appendix B-1, or a pharmaceutically acceptable salt thereof, in a subject having a RAS G12C mutated NSCLC or colorectal cancer, comprising administering to the subject an effective amount of RMC-5552, or a stereoisomer or tautomer thereof, wherein the effective amount is an amount effective to treat acquired resistance to the compound, or a pharmaceutically acceptable salt thereof, in the subject.

Embodiment III-4. A method of treating acquired resistance to Compound A, or a pharmaceutically acceptable salt thereof, in a subject having a RAS G12C mutated NSCLC or colorectal cancer, comprising administering to the subject an effective amount of RMC-5552, or a stereoisomer or tautomer thereof, wherein the effective amount is an amount effective to treat acquired resistance to Compound A, or a pharmaceutically acceptable salt thereof, in the subject.

Embodiment III-5. A method of treating acquired resistance to Compound B, or a pharmaceutically acceptable salt thereof, in a subject having a RAS G12C mutated NSCLC or colorectal cancer, comprising administering to the subject an effective amount of RMC-5552, or a stereoisomer or tautomer thereof, wherein the effective amount is an amount effective to treat acquired resistance to Compound B, or a pharmaceutically acceptable salt thereof, in the subject.

Embodiment IV-1. A method of treating a subject having a RAS G12C mutated NSCLC or colorectal cancer, comprising administering to the subject an effective amount of RMC-5552, or a stereoisomer or tautomer thereof, in combination with AMG 510 or MRTX849, or a pharmaceutically acceptable salt thereof.

Embodiment IV-1. A method of treating a subject having a RAS G12C mutated NSCLC or colorectal cancer, comprising administering to the subject an effective amount of RMC-5552, or a stereoisomer or tautomer thereof, in combination with a compound of Formula IVb of Appendix B-1, or a pharmaceutically acceptable salt thereof:

Formula VIb wherein A is a 3 to 6-membered heterocycloalkylene, a phenylene, or a hydroxy-substituted phenylene; B is -CH(C 1 -C 6 alkyl)-; L is a linker selected from the following:

W is a cross-linking group selected from the following:

Embodiment IV-3. A method of treating a subject having a RAS G12C mutated NSCLC or colorectal cancer, comprising administering to the subject an effective amount of RMC-5552, or a stereoisomer or tautomer thereof, in combination with a compound selected from compound A121, A131, A133, A145, A150, A173, A182, A191, A198, A199, A201, A244, A245, A246, A247, A248, A266, A290, A292, A310, A316, A317, A324, A325, A326,

A337, A339, A351, A365, A377, A391, A402, A412, A413, A414, A426, A476, A487, A499, A508, A509, A526, A528, A532, A533, A534, A551, A559, A560, A565, A566, A567, A568, A569, A584, A585, A591, A592, A599, A601, A613, A614, A615, A616, A617, A643, A644, A646, A647, A648, A657, A663, A672, A699, A708, A715, A717 and A733 of Appendix B-1, or a pharmaceutically acceptable salt thereof.

Embodiment IV-4. A method of treating a subject having a RAS G12C mutated NSCLC or colorectal cancer, comprising administering to the subject an effective amount of RMC-5552, or a stereoisomer or tautomer thereof, in combination with Compound A, or a pharmaceutically acceptable salt thereof.

Embodiment IV-5. A method of treating a subject having a RAS G12C mutated NSCLC or colorectal cancer, comprising administering to the subject an effective amount of RMC-5552, or a stereoisomer or tautomer thereof, in combination with Compound B, or a pharmaceutically acceptable salt thereof.

Embodiment V-1. A method of inducing apoptosis of a RAS G12C mutated NSCLC or colorectal tumor cell, comprising contacting the tumor cell with an effective amount of RMC- 5552, or a stereoisomer or tautomer thereof, in combination with AMG 510 or MRTX849, or a pharmaceutically acceptable salt thereof, wherein the effective amount is an amount effective to induce apoptosis of the tumor cell.

Embodiment V-2. A method of inducing apoptosis of a RAS G12C mutated NSCLC or colorectal tumor cell, comprising contacting the tumor cell with an effective amount of RMC-5552, or a stereoisomer or tautomer thereof, in combination with a compound of Formula IVb of Appendix B-1, or a pharmaceutically acceptable salt thereof: Formula VIb wherein A is a 3 to 6-membered heterocycloalkylene, a phenylene, or a hydroxy-substituted phenylene; B is -CH(C 1 -C 6 alkyl)-; L is a linker selected from the following: ; and

W is a cross-linking group selected from the following:

amount is an amount effective to induce apoptosis of the tumor cell.

Embodiment V-3. A method of inducing apoptosis of a RAS G12C mutated NSCLC or colorectal tumor cell, comprising contacting the tumor cell with an effective amount of RMC- 5552, or a stereoisomer or tautomer thereof, in combination with a compound selected from compound A121, A131, A133, A145, A150, A173, A182, A191, A198, A199, A201, A244, A245, A246, A247, A248, A266, A290, A292, A310, A316, A317, A324, A325, A326,

A337, A339, A351, A365, A377, A391, A402, A412, A413, A414, A426, A476, A487,

A499, A508, A509, A526, A528, A532, A533, A534, A551, A559, A560, A565, A566,

A567, A568, A569, A584, A585, A591, A592, A599, A601, A613, A614, A615, A616,

A617, A643, A644, A646, A647, A648, A657, A663, A672, A699, A708, A715, A717 and

A733 of Appendix B-1, or a pharmaceutically acceptable salt thereof, wherein the effective amount is an amount effective to induce apoptosis of the tumor cell.

Embodiment V-4. A method of inducing apoptosis of a RAS G12C mutated NSCLC or colorectal tumor cell, comprising contacting the tumor cell with an effective amount of RMC- 5552, or a stereoisomer or tautomer thereof, in combination with Compound A, or a pharmaceutically acceptable salt thereof, wherein the effective amount is an amount effective to induce apoptosis of the tumor cell. Embodiment V-5. A method of inducing apoptosis of a RAS G12C mutated NSCLC or colorectal tumor cell, comprising contacting the tumor cell with an effective amount of RMC- 5552, or a stereoisomer or tautomer thereof, in combination with Compound B, or a pharmaceutically acceptable salt thereof, wherein the effective amount is an amount effective to induce apoptosis of the tumor cell.

[00163] All of the U.S. patents, U.S. patent application publications, U.S. patent applications, PCT patent application, PCT patent application publications, foreign patents, foreign patent applications and non-patent publications referred to in this specification or listed in any Application Data Sheet are incorporated herein by reference in their entirety. From the foregoing it will be appreciated that, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention.

Examples

[00164] The disclosure is further illustrated by the following examples and synthesis examples, which are not to be construed as limiting this disclosure in scope or spirit to the specific procedures herein described. It is to be understood that the examples are provided to illustrate certain embodiments and that no limitation to the scope of the disclosure is intended thereby. It is to be further understood that resort may be had to various other embodiments, modifications, and equivalents thereof which may suggest themselves to those skilled in the art without departing from the spirit of the present disclosure and/or scope of the appended claims.

Example 1.

In Vitro Combinatorial Activity of RM-006 (also known as RMC-6272) and KRAS G12C (OFF) Inhibitor in NSCLC Cells with RAS & mTOR Signaling Co-activation

Objective:

[00165] The RAS and PI3K/mTOR signaling pathways are hyperactivated in many human cancers. In the PI3K/mTOR pathway, mTORC1 phosphorylates and inactivates the tumor suppressor 4EBP1, enabling cap-dependent translation, including translation of key oncogenes. We have developed a series of bi-steric mTORC1 -selective inhibitors that activate 4EBP1. As shown in Table 1, RM-006 (also known as RMC-6272), one representative example of these new bi-steric inhibitors, has potent and selective (>10 fold) inhibition of mTORC1 over mTORC2, and durably suppress phosphorylation of S6K and 4EBP1 in vitro and in vivo.

Table 1:

[00166] In this Example, we tested the in vitro combinatorial effect of the bi-steric mTOR inhibitor RM-006 (also known as RMC-6272) and the KRAS G12C (OFF) inhibitor AMG 510 on non-small cell lung cancer cell lines NCI-H2122 and NCI-H2030, which each have a KRAS G12C mutation and mTOR signaling co-activation.

Methods:

[00167] Cells were grown in culture as 3D spheroids. Briefly, 1000 cells/well (for NCI- 112122) and 1500 cells/well (for NCI-H2030) were seeded in round bottom ultra-low attachment 384-well plates in growth media supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin, and allowed to form spheroids for 24 hours at 37°C in 5% C02. Spheroid formation was confirmed visually, and spheroids were treated in duplicate with serial 3.16-fold dilutions of single-agent inhibitor or in combination (final DMSO concentration = 0.2%). Following drug exposure for five days, cell viability in spheroids was determined using the 3D-CellTiter-Glo® assay kit (Promega).

Results:

[00168] RM-006 (also known as RMC-6272) shows in vitro combinatorial anti-proliferative activity with AMG 510 in two NSCLC cell lines with co-occurring KRAS G12C and STK11 loss of function mutations. STK11 is a negative regulator of mTOR signaling. In FIG. 1A, we evaluated varying concentrations of AMG 510 in presence of constant RM-006 (also known as RMC-6272) (3 nM in H2122, left panel, and 10 nM in H2030, right panel), and showed combinatorial anti-proliferative activity at select concentrations of AMG 510. In FIG. 1B, we evaluated varying concentrations of RM-006 (also known as RMC-6272) in presence of constant AMG 510 (90 nM in H2122, left panel, or 10 nM in H2030, right panel), and showed combinatorial anti-proliferative activity at select concentrations of RM-006 (also known as RMC-6272). Thus, the combination of a bi-steric mTORC1 -selective inhibitor with a KRAS inhibitor drives tumor regression in a NSCLC model with co-activation of RAS & mTOR signaling.

Example 2.

In Vivo Combinatorial Activity of RM-006 (also known as RMC-6272) and KRAS G12C (OFF) Inhibitor in non-small cell lung cancer NCI-H358 KRAS G12C xenograft model

Objective:

[00169] Having demonstrated in Example 1 that the combined inhibition of the RAS and PI3K/mTOR signaling pathways provided for significant in vitro anti-tumor activity, we sought to extend our results to an in vivo tumor model. To that end, the combinatorial effects of RM- 006 (also known as RMC-6272) with AMG 510 on tumor cell growth in vivo were evaluated in the human non-small cell lung cancer NCI-H358 KRAS G12C xenograft model using female BALB/c nude mice (6-8 weeks old).

Methods:

[00170] Mice were implanted with NCI-H358 tumor cells in 50% Matrigel (5 x 106 cells/mouse) subcutaneously in the flank. Once tumors reached an average size of ~ 200 mm3, mice were randomized to treatment groups to start the administration of test articles or vehicle. RM-006 (also known as RMC-6272) was administered by intraperitoneal injection once weekly, and AMG 510 was administered by oral gavage daily. Body weight and tumor volume (using calipers) was measured twice weekly until study endpoints.

Results:

[00171] FIG. 2A shows a mean tumor volume plot and demonstrates that the combination of RM-006 (also known as RMC-6272) administered at 10 mg/kg IP weekly plus AMG 510 given at a submaximal dose of 5 mg/kg via PO daily led to tumor regression in NCI-H358 KRAS G12C xenograft model, which is a sensitive model to KRAS G12C inhibition alone. The end of study responses of each mouse represented as a waterfall plot is shown in FIG. 2B. The number of tumors with reduction in tumor volume greater than 10% of the baseline is indicated on the waterfall plot (FIG. 2B). [00172] In FIG. 2C, the combination of RM-006 (also known as RMC-6272) at 10 mg/kg IP weekly plus AMG 510 inhibitor at 30 mg/kg PO daily (a regression-driving dose but submaximal) resulted in a more durable response that delayed tumor regrowth after treatment cessation, relative to AMG 510 alone. Kaplan-Meier analysis shown in FIG. 2D demonstrates that when comparing to the single-agent AMG 510, combination with RM-006 (also known as RMC-6272) significantly delayed the regrowth of tumors back to 500 mm3 after treatment cessation, as assessed by Log-rank (Mantel-Cox) test with p = 0.0395. The combination treatment was well tolerated. These findings indicate that even in mutant KRAS tumor cells without known genomic aberrations conferring mTOR pathway activation, the concomitant targeting of both the RAS and mTOR signaling by an inhibitor of mutant KRAS and a bi-steric mTORC1-selective inhibitor may demonstrate benefit over mutant KRAS inhibitor alone.

Example 3.

In Vivo Combinatorial Activity of RM-006 (also known as RMC-6272) and KRAS G12C (OFF) Inhibitor in non-small cell lung cancer NCI-H2122 KRAS G12C ; STKlldel xenograft model

Objective:

[00173] To further explore the in vivo utility that the combined inhibition of the RAS and PI3K/mTOR signaling pathways provides, we investigated the combinatorial effects of RM- 006 (also known as RMC-6272) with AMG 510 on tumor cell growth in vivo in the human non-small cell lung cancer NCI-H2122 KRAS G12C ; STK11del xenograft model using female BALB/c nude mice (6-8 weeks old).

Methods:

[00174] Mice were implanted with NCI-H2122 tumor cells in 50% Matrigel (5 x 106 cells/mouse) subcutaneously in the flank. Once tumors reached an average size of ~ 166 mm3, mice were randomized to treatment groups to start the administration of test articles or vehicle. RM-006 (also known as RMC-6272) was administered by intraperitoneal injection once weekly, and AMG 510 was administered by oral gavage daily. Body weight and tumor volume (using calipers) was measured twice weekly until study endpoints.

Results:

[00175] As shown in the tumor volume plot in FIG. 3A, single-agent RM-006 (also known as RMC-6272) administered at 10 mg/kg IP weekly led to a tumor growth inhibition (“TGI”) of 27.4%, and single-agent AMG 510 administered at 100 mg/kg PO daily led to a TGI of 54.6% in the NCI-H2122 NSCLC CDX model with co-occurring KRAS G12C and STK11de. However, the combination drove tumor regressions in the NCI-H2122 model. The anti-tumor or activity by the combination treatment was statistically significant from the vehicle control group, with ***p<0.001, assessed by an ordinary One-way ANOVA of tumor volumes along with multiple comparisons via a post-hoc Tukey’s test in GraphPad Prism software. In FIG. 3B, waterfall plot shows individual tumor responses at the end of study, and 7/10 tumors from the combination group showed reductions in tumor volume greater than 10% of the baseline. The combination treatment was well tolerated. These data demonstrate that the Combination of RM-006 (also known as RMC-6272) and KRAS G12C (OFF) inhibition drives tumor regression in a NSCLC model with co-activation of RAS & mTOR signaling.

[00176] The NCI-H2122 model is an example of a NSCLC model that exhibited relatively lower anti-tumor or response to either KRAS G12C (OFF) inhibitor or mTORC1 inhibitor monotherapy, as evidenced by some tumor growth inhibition but no reductions in tumor volume in preclinical studies. In contrast, the combination of both inhibitors resulted in tumor regressions and exemplifies the use of this therapeutic regimen to overcome up-front or intrinsic resistance. NCI-H2122 tumor cells harbor activating mutations that drive oncogenic signaling via both the RAS and the mTOR signaling pathway. Thus, we hypothesize that neither single agent is able to sufficiently overcome the oncogenic flux driven by the co- activation of both pathways and combination therapy is required to induce apoptosis and tumor regressions.

Example 4.

In Vivo single-dose PKPD study of the combinatorial Activity of RM-006 (also known as RMC-6272) and KRAS G12C (OFF) Inhibitor in human non-small cell lung cancer NCI- H2122 KRAS G12C ; STK11 del xenograft model

Objective:

[00177] We investigated the pharmacokinetic and pharmacodynamic (PKPD) effects RM- 006 (also known as RMC-6272), AMG-510, and the combination of the two inhibitors had in human non-small cell lung cancer NCI-H2122 KRAS G12C ; STK11del xenograft model.

Methods:

[00178] RM-006 (also known as RMC-6272) was administered at 10 mg/kg IP, whereas

AMG 510 was administered at 100 mg/kg by oral gavage. The treatment groups with sample collections at various time points were summarized in Table 1 below. Plasma samples were collected for bioanalysis of the compounds, and tumor samples were collected to assess pathway modulation by quantitative image analyses of immunohistochemical (IHC) staining for phosphorylated proteins that are known biomarkers of mTOR and RAS pathway activity. Tumor sections were stained with monoclonal antibodies against pS6RP(Ser235/236), p4E- BP1(Thr37/46), and pERK (Thr202/Tyr204), and visualized with DAB chromogen and, counterstained with hematoxylin, and scanned to generate a digital image. Digital images were analyzed with Indica Lab’s HALO software using the area quantification module where colors and intensity were measured on a pixel by pixel basis. Whole tumor sections, excluding necrotic regions and murine tissue, were measured for intensity above background and the percent positivity calculated for the given area measured. Additionally, qPCR assay was used to measure the mRNA level of human DUSP6 as another marker for RAS/ERK signaling.

[00179] The treatment groups, doses, and time points for the single-dose PKPD study using NCI-H2122 tumors are shown in Table 2.

Table 2. Summary of treatment groups, doses, and time points for single-dose PKPD study using NCI-H2122 tumors.

Results:

[00180] As shown in FIG. 4A, the combination of RM-006 (also known as RMC-6272) at 10 mg/kg IP and AMG 510 100 mg/kg PO led to stronger inhibition of pS6RP (Ser235/236) across all time points, relative to each single agent. pS6RP (Ser235/236) is a key converging node that can be modulated by both the mTOR and RAS pathways. As shown in FIGS. 4B-4D, the effects on p4EBP1, pERK, and DUSP6 are consistent with the expected pathway modulation by RM-006 (also known as RMC-6272) and AMG 510 on mTOR and RAS signaling, respectively. Unbound plasma concentration of each single agent is shown as lines on the bar graphs of FIGS. 4A-4D. In combination, the PK profile of each agent is consistent with that of the single agent, indicative of no DDI, thus only single agent PK is shown. Representative IHC staining images for pS6RP (Ser235/236) and p4EBP1 (Thr37/46) at 4 and 48 hours after dosing are shown in FIGS. 4E and 4F, respectively.

[00181] Tumors from the single-dose study described in Figure 4 were stained for cleaved caspase3 (CC3) using a polyclonal antibody by IHC. HALO quantitative image analysis as described above was applied to assess the overall CC3 induction.

[00182] As shown in FIGS. 5A and 5B, in the human non-small cell lung cancer NCI-H2122 KRAS G12C ; STK11 del tumors, combination of RM-006 (also known as RMC-6272) and AMG 510 led to a significant induction of apoptosis as measured by cleaved caspase 3 IHC staining, relative to each single agent alone. Based on the time points we assessed, maximal induction of apoptosis occurred at 24 hours after a single dose, resulting in an induction of CC3 positivity~ 900% more than the control group. The % CC3 positivity of tumors from each treatment group at various time points is summarized as mean with SEM in Table 3 below. These values were used to generate the bar graph shown in FIG. 5A.

Table 3. Percent CC3 positivity of tumors from each treatment groups at indicated time points is summarized as mean and SEMwith N indicating the number of mice. The values from this table were used to generate the bar graph in FIG. 5A.

*also known as RM-006

[00183] Adult somatic cells almost all will die by apoptosis, a form of programmed cell death. Cancer cells, harboring alterations that result in impaired apoptotic signaling, often acquire the ability to evade death by inactivating cell death pathways (Long 2012). Hence, reduced apoptosis or its resistance plays a vital role in carcinogenesis (Hanahan 2000).

[00184] A successfully cancer therapy can promote cancer cell death while minimizing comparable damage to normal cells. Numerous in vitro and in vivo studies have indicated that tumor cell apoptosis induction is part of the mechanism of action of many approved drugs in cancer treatment in both preclinical and clinical settings (Gerl 2005). [00185] In this study, our results demonstrate that combination treatment of RM-006 (also known as RMC-6272) and KRAS G12C inhibitor can induce significant apoptosis in NCI-H2122 xenograft tumors in vivo. This is the first time to our knowledge that combination treatment with an mTOR inhibitor and KRAS G12C mutant selective inhibitor has shown to promote tumor apoptosis in vivo.

References:

[00186] Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000; 100:57-70.

[00187] Gerl R, Vaux DL. Apoptosis in the development and treatment of cancer, Carcinogenesis. 2005; 26(2):263-270

[00188] Long J, Ryan, K. New frontiers in promoting tumor cell death: targeting apoptosis, necroptosis and autophagy. Oncogene 2012; 31:5045-5060.

Example 5.

Combination of RM-006 (also known as RMC-6272) and a KRAS G12C (OFF) inhibitor significantly delays on-treatment resistance in a NSCLC model with RAS and mTOR signaling co-activation

Objective:

[00189] We investigated the in vivo combinatorial effects of RM-006 (also known as RMC- 6272) with AMG 510 on tumor cell growth in the human non-small cell lung cancer NCI- 112030 KRAS G12C ; STK11E317* xenograft model using female NOD SCID mice (4-5 weeks old).

Methods:

[00190] Mice were implanted with NCI-H2030 tumor cells in 50% Matrigel (1 x 107 cells/mouse) subcutaneously in the flank. Once tumors reached an average size of 150-200 mm3, mice were randomized to treatment groups to start the administration of test articles or vehicle. RM-006 (also known as RMC-6272) was administered by intraperitoneal injection once weekly, and AMG 510 was administered by oral gavage daily. Body weight and tumor volume (using calipers) was measured twice weekly until study endpoints.

Results:

[00191] In the human non-small cell lung cancer NCI-H2030 KRAS G12C ; STK11 E317* tumors, the combination of RM-006 (also known as RMC-6272) dosed at 3 mg/kg or 10 mg/kg IP weekly plus AMG 510 100 mg/kg PO daily resulted in durable tumor regression and delayed on-treatment resistance, relative to single agent AMG 510, as shown by the mean tumor volume plot presented in FIG. 6A. Kaplan-Meier analysis of tumors reaching baseline volume while on treatment showed the combination significantly prolonged the time for tumors to develop resistance, as assessed by Log-rank (Mantel-Cox) test (FIG. 6B). Table 4 below summarizes the comparisons and P values.

Table 4. Comparisons of treatment groups and summary of P values by Log-rank (Mantel- Cox) test, where RM-006 is also known as RMC-6272.

Example 6.

Combination of RM-006 (also known as RMC-6272) and a KRAS G12C (OFF) inhibitor attenuates AMG 510 on-treatment resistant tumor growth in the human non-small cell lung cancer NCI-H2030 KRAS G12C ; STK11 E317* xenograft model

Objective:

[00192] We evaluated whether combination treatment of RM-006 (also known as RMC- 6272) and AMG 510 could attenuate AMG 510 on-treatment resistant tumor growth in the human non-small cell lung cancer NCI-H2030 KRAS G12C ; STK11 E317* xenograft model after the development of resistance.

Methods:

At Day 59 post-implantation in the experiment described in Example 6, above, animals treated with AMG 510 administered by oral gavage daily at 100 mg/kg exhibited on-treatment resistance (see Figure. 7). At this time, RM-006 was administered to the same group of animals by intraperitoneal injection once weekly at 10 mg/kg, while AMG 510 treatment continued. Body weight and tumor volume (using calipers) was measured twice weekly until study endpoints.

Results: [00193] In the human non-small cell lung cancer NCI-H2030 KRAS G12C ; STK11 E317* tumors, AMG 510 100 mg/kg PO daily treatment group developed on-treatment resistance after 2-3 weeks of treatment (Figure 7). Adding RM-006 (also known as RMC-6272) (10 mg/kg IP weekly) in combination with AMG 510 (100 mg/kg PO daily) to the same group of animals attenuated resistant tumor growth, as shown by the individual tumor volume plot (Figure 7).

[00194] The NCI-H2030 model exemplifies a scenario wherein a KRAS G12C mutant tumor is initially sensitive to KRAS G12C (OFF) inhibitor monotherapy, as demonstrated by the initial tumor regressions observed following treatment in this model. However, upon longer-term treatment, xenograft tumors were able to regrow and exhibited on-treatment resistance. The combination of KRAS G12C (OFF) inhibitor and mTORC1 inhibitor significantly delayed the onset of this on-treatment resistance. Moreover, the addition of an mTORC1 inhibitor to KRAS G12C (OFF) inhibitor treatment at the onset of monotherapy resistance (to the latter) resulted in attenuation of tumor growth and in some cases, apparent regression following combination therapy.

[00195] In sum, these results support that mTOR activation limits therapeutic response to mutant KRAS G12C inhibition; and provide an initial demonstration that combinatorial inhibition of RAS and mTOR signaling is sufficient to forestall on-treatment resistance to KRAS G12C (OFF) inhibition.

Example 7.

Combination of RM-006 (also known as RMC-6272) and a KRAS G12C (OFF) inhibitor attenuates AMG 510 on tumor growth in the human colorectal cancer (CRC) patient derived xenograft (PDX) ST3235 (PIK3CA E545K ) model

Objective:

[00196] We evaluated whether combination treatment of RM-006 (also known as RMC- 6272) and AMG 510 could attenuate AMG 510 on tumor growth in the human colorectal cancer (CRC) patient derived xenograft (PDX) ST3235 (PIK3CA E545K ) model after the development of resistance.

Methods:

[00197] The combinatorial effects of RM-006 (also known as RMC-6272) with AMG 510 on tumor growth in vivo were evaluated in the human colorectal cancer (CRC) patient derived xenograft (PDX) model ST3235 KRAS G12C ; PIK3CA E545K using female athymic nude mice (6- 12 weeks old) (Figure 8). Tumor fragments of about 70 mg in weight from ST3235 CRC PDX model were implanted (s.c.) into the right flanks of athymic nude mice. When tumor sizes reached an average size of 150-200 mm 3 , mice were randomized to treatment groups to start the administration of test articles or vehicle. RM-006 (also known as RMC-6272) was administered by intraperitoneal injection once weekly, and AMG 510 was administered by oral gavage daily. Body weight and tumor volume (using calipers) was measured twice weekly until study endpoints.

Results:

[00198] Single-agent RM-006 (also known as RMC-6272) administered at 3 mg/kg IP weekly led to a TGI of 47.6%, and single-agent AMG 510 administered at 100 mg/kg PO daily led to a TGI of 71.5% in ST3235 human CRC PDX model with co-occurring KRAS G12C and PIK3CA E545K . However, combination of RM-006 (also known as RMC-6272) (3 mg/kg) and AMG 510 (100 mg/kg) displayed better tumor growth inhibition than either single agent group with TGI of 92.7%. The anti-tumor activity by the combination treatment was statistically significant compared with control group (***p<0.001, ordinary One-way ANOVA with multiple comparisons via a post-hoc Tukey’s test).

Example 8.

Combination of RM-006 (also known as RMC-6272) and Compound A, a KRAS G12C (ON) inhibitor, on tumor growth in the human lung cancer ST1989 KRAS G12C patient-derived xenograft model.

Objective:

We evaluated whether combination treatment of RM-006 (also known as RMC-6272) and Compound A, a KRAS G12C (ON)inhibitor as disclosed herein, could attenuate tumor cell growth in vivo in the human lung cancer ST 1989 KRAS G12C patient-derived xenograft model using female athymic nude mice. Compound A is a KRAS G12C (ON) inhibitor disclosed in Appendix B-1.

Methods:

[00199] The combinatorial effects of RM-006 (also known as RMC-6272) with Compound A on tumor cell growth in vivo were evaluated in the human lung cancer ST 1989 KRAS G12C patient-derived xenograft model using female athymic nude mice (6-12 weeks old). Mice were implanted with tumor fragment of approximately 70 mg in size subcutaneously in the flank region. Once tumors reached an average size in the range between 150-300 mm 3 , mice were randomized to treatment groups with three mice per group to start the administration of test articles or vehicle. RM-006 (also known as RMC-6272) was administered by intraperitoneal injection once weekly, and Compound A was administered by oral gavage daily. Body weight and tumor volume (using calipers) was measured twice weekly until study endpoints. End of study responses in individual tumors were plotted as a waterfall plot, and the numbers indicate number of tumor regression in each group. Tumor regression is defined as greater than 10% reduction of tumor volume at the end of study relative to initial volume.

Results:

[00200] Here in Figure 9, single-agent RM-006 (also known as RMC-6272) administered at 3 mg/kg IP weekly led to a tumor growth inhibition (TGI) of 31.6%, and single-agent Compound A administered at 100 mg/kg PO daily led to a TGI of 45.3% in ST1989 tumors. Importantly, the combination of RM-006 (also known as RMC-6272) 3 mg/kg plus Compound A 100 mg/kg led to a TGI of 96.5%. End of study responses were shown as a waterfall plot, which indicates 1 out 3 mice had tumor regression in the combination group, whereas no tumor regressions recorded in each single agent group. The combination treatment was tolerated.

Example 9.

Combinatorial effects of RMC-6272 (also known as RM-006) with Compound B, a KRAS G12C (ON) inhibitor, in a NSCLC CDX Model

Methods:

[00201] The combinatorial effects of bi-steric mTOR inhibitor RMC-6272 (also known as RM-006) with Compound B, a KRAS G12C (ON) inhibitor disclosed herein, on tumor cell growth in vivo were evaluated in the human NSCLC NCI-H2122 (KRAS G12C ; STK11 MUT ; REAP1 MUT ) cell line-derived xenograft model using female Balb/c nude mice (4-6 weeks old). Mice were implanted with NCI-H2122 cancer cells in 50% Matrigel (5 x 10 6 cells/mouse) subcutaneously in the flank. Once tumors reached an average size in the range between 150-200 mm 3 , mice were randomized to treatment groups with eight mice per group to start the administration of test articles or vehicle. RMC-6272 (also known as RM-006) was administered by intraperitoneal (ip) injection once weekly, and Compound B was administered by oral gavage (po) daily. Body weight and tumor volume (using calipers) was measured twice weekly until study endpoints. Compound B is a KRAS G12C (ON) inhibitor disclosed in Appendix B-1.

Results:

[00202] In Figure 10, single-agent RMC-6272 (also known as RM-006 administered at 8 mg/kg ip weekly led to a tumor growth inhibition (TGI) of 59.0%, and single-agent Compound B administered at 100 mg/kg po daily led to a TGI of 87.4% at Day 17 post-dosing started in NCI-H2122 xenografted tumors. Importantly, the combination of RMC-6272 (also known as RM-006) 8 mg/kg plus Compound B 100 mg/kg led to complete regression of all tumors in the group at Day 17 post-dosing started. And all tumors in the combination group still exhibited tumor regression at Day 31 post-dosing started. All treatment was tolerated during the study course.

Example 10.

Combinatorial effects of RMC-5552 with Compound B, a KRAS G12C (ON) inhibitor as in Example 9, in a NSCLC CDX Model

Methods:

[00203] The combinatorial effects of bi-steric mTOR inhibitor RMC-5552 with Compound B, a KRAS G12C (ON) inhibitor disclosed herein and as in Example 9, on tumor cell growth in vivo were evaluated in the human NSCLC NCI-H2122 (KRAS G12C ; STK11 MUT ; KEAP1 MUT ) cell line-derived xenograft model using female Balb/c nude mice (4-6 weeks old). Mice were implanted with NCI-H2122 cancer cells in 50% Matrigel (5 x 10 6 cells/mouse) subcutaneously in the flank. Once tumors reached an average size in the range between 150-200 mm 3 , mice were randomized to treatment groups with eight mice per group to start the administration of test articles or vehicle. RMC-5552 was administered by intraperitoneal (ip) injection once weekly, and Compound B was administered by oral gavage (po) daily. Body weight and tumor volume (using calipers) was measured twice weekly until study endpoints. Compound B is a KRAS G12C (ON) inhibitor disclosed in Appendix B-1.

Results:

[00204] In Figure 11, single-agent RMC-5552 administered at 10 mg/kg ip weekly led to a tumor growth inhibition (TGI) of 37.1%, and single-agent Compound B administered at 100 mg/kg po daily led to a TGI of 85.5% at Day 21 post-dosing started in NCI-H2122 xenografted tumors. Importantly, the combination of RMC-5552 10 mg/kg plus Compound B 100 mg/kg led to tumor growth inhibition (TGI) of 99.0% at Day 21 post-dosing started, with 3 out of 8 tumors exhibiting more than 10% tumor volume reduction from baseline. The anti -tumor activity of both Compound B (100 mg/kg po daily) and the combination treatment was statistically significant compared with control group (***p<0.001, ordinary One-way ANOVA with multiple comparisons via a post-hoc Tukey’s test). All treatment was tolerated during the study course.

Equivalents

[00205] While the present invention has been described in conjunction with the specific embodiments set forth above, many alternatives, modifications and other variations thereof will be apparent to those of ordinary skill in the art. All such alternatives, modifications and variations are intended to fall within the spirit and scope of the present invention. All of the U.S. patents, U.S. patent application publications, U.S. patent application, foreign patents, foreign patent application and non-patent publications referred to in this specification and/or listed in the Application Data Sheet are incorporated herein by reference, in their entirety. Aspects of the embodiments can be modified, if necessary to employ concepts of the various patents, application and publications to provide yet further embodiments. These and other changes can be made to the embodiments in light of the above-detailed description. In general, in the following claims, the terms used should not be construed to limit the claims to the specific embodiments disclosed in the specification and the claims, but should be construed to include all possible embodiments along with the full scope of equivalents to which such claims are entitled. Accordingly, the claims are not limited by the disclosure.

Appendix A-1

RAS INHIBITORS

Background

The vast majority of small molecule drugs act by binding a functionally Important pocket on a target protein, thereby modulating the activity of that protein. For example, cholesterol-lowering drugs known as statins bind the enzyme active site of HMG-CoA reductase, thus preventing the enzyme from engaging with its substrates. The fact that many such drug/target interacting pairs are known may have misled some into believing that a small molecule modulator could be discovered for most, if not ail, proteins provided a reasonable amount of time, effort, and resources. This is far from the case. Current estimates are that only about 10% of all human proteins are targetable by small molecules. Bojadzic and Buchwald, Curr Top Med Chem 18: 674-699 (2019). The other 90% are currently considered refractory or intractable toward above-mentioned small molecule drug discovery. Such targets are commonly referred to as “undruggable.” These undruggable targets include a vast and largely untapped reservoir of medically important human proteins. Thus, there exists a great deal of Interest in discovering new molecular modalities capable of modulating the function of such undruggable targets.

It has been well established in literature that Ras proteins (K-Ras, H-Ras and N-Ras) play an essential role in various human cancers and are therefore appropriate targets for anticancer therapy, indeed, mutations in Ras proteins account for approximately 30% of all human cancers in the United States, many of which are fatal. Dysregulation of Ras proteins by activating mutations, overexpression or upstream activation is common in human tumors, and activating mutations in Ras are frequently found in human cancer. For example, activating mutations at codon 12 in Ras proteins function by inhibiting both GTPase-activating protein (GAP)-dependent and intrinsic hydrolysis rates of GTP, significantly skewing the population of Ras mutant proteins to the “on” (GTP-bound) state (Ras(ON)), leading to oncogenic MARK signaling. Notably, Ras exhibits a picomolar affinity for GTP, enabling Ras to be activated even in the presence of low concentrations of this nucleotide. Mutations at codons 13 (e.g., G13D) and 61 (e.g., Q61K) of Ras are also responsible for oncogenic activity in some cancers.

Despite extensive drug discovery efforts against Ras during the last several decades, a drug directly targeting Ras is still not approved. Additional efforts are needed to uncover additional medicines for cancers driven by the various Ras mutations.

Summary

Provided herein are Ras inhibitors. The approach described herein entails formation of a high affinity three-component complex, or conjugate, between a synthetic ligand and two intracellular proteins which do not interact under normal physiological conditions: the target protein of interest (e.g., Ras), and a widely expressed cytosolic chaperone (presenter protein) in the cell (e.g., cyclophilin A). More specifically, in some embodiments, the inhibitors of Ras described herein induce a new binding pocket in Ras by driving formation of a high affinity tri- complex, or conjugate, between the Ras protein and the widely expressed cytosolic chaperone, cyclophilin A (CYRA). Without being bound by theory, the inventors believe that one way the inhibitory effect on Ras is effected by compounds of the invention and the complexes, or conjugates, they form is by steric occlusion of the interaction site between Ras and downstream effector molecules, such as RAF and PI3K, which are required for propagating the oncogenic signal.

As such, in some embodiments, the disclosure features a compound, or pharmaceutically acceptable salt thereof, of structural Formula I: wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 10- membered heteroarylene;

B is -CH(R 9 )- or >C=CR 9 R 9’ where the carbon is bound to the carbonyl carbon of -N(R 11 )C(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C 1 -C 4 alkylene, optionally substituted C 1 -C 4 alkenylene, optionally substituted C 1 -C 4 heteroalkylene, -C(O)O-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(O)NH-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C 1 -C 4 heteroalkylene, or 3 to 8-membered heteroarylene:

L is absent or a linker;

W is a cross-linking group comprising a carbodiimide, an oxazoline, a thiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziridine, a trifiuoromethyl ketone, a boronlc acid, a boronic ester, anN-ethoxycarbonyl-2-ethoxy-1 ,2-dihydroquinoline (EEDQ), an iso-EEDQ or other EEDG derivative, an epoxide, an oxazoiium, or a glycal ;

X 1 is optionally substituted C 1 -C 2 alkylene, NR, O, or S(G) n ;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2:

R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R’, C(O)OR’, C(G)N(R’) 2 , S(O)R', S(O) 2 R’, or S(O) 2 N(R') 2 ; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl; Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 is CH, CH 2 , or N;

Y 6 is C(O), CH, CH 2 , or N;

R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl, or

R 1 and R 2 combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl ;

R 2 is absent, hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C 1 -C 3 alkyl), C=O, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7a and R 8a are, independently, hydrogen, halo, optionally substituted C 1 -C 3 alkyl, or combine with the carbon to which they are attached to form a carbonyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is hydrogen, F, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl, or R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 9' is hydrogen or optionally substituted C 1 -C 6 alkyl;

R 10 is hydrogen, halo, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl;

R 10a is hydrogen or halo;

R 11 is hydrogen or C 1 -C 3 alkyl; and

R 34 is hydrogen or C 1 -C 3 alkyl (e.g., methyl).

Also provided are pharmaceutical compositions comprising a compound of Formula I, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.

Further provided is a conjugate, or salt thereof, comprising the structure of Formula IV:

M-L-P Formula IV wherein L is a linker;

P is a monovalent organic moiety; and M has the structure of Formula V:

Formula V wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene;

B is -CH(R 9 )- or >C=CR 9 R 9' where the carbon is bound to the carbonyl carbon of -N(R 11 )C(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C 1 -C 4 alkylene, optionally substituted C 1 -C 4 alkenylene, optionally substituted C 1 -C 4 heteroalkylene, -C(O)O-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(O)NH-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C 1 -C 4 heteroalkylene, or 3 to 8-membered heteroarylene;

X 1 is optionally substituted C 1 - C 2 alkylene, NR, O, or S(O) n ; X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2; R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R', C(O)OR', C(O)N(R') 2 , S(O)R', S(O) 2 R', or S(O) 2 N(R') 2 ; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 is CH, CH 2 , or N;

Y 6 is C(O), CH, CH 2 , or N;

R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl, or

R 1 and R 2 combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 2 is absent, hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C 1 -C 3 alkyl), C=O, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7a and R 8a are, independently, hydrogen, halo, optionally substituted C 1 -C 3 alkyl, or combine with the carbon to which they are attached to form a carbonyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl; R 9 is hydrogen, F, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl, or

R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl ;

R 9' is hydrogen or optionally substituted C 1 -C 6 alkyl;

R 10 is hydrogen, halo, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl;

R 10a is hydrogen or halo;

R 11 is hydrogen or C 1 -C 3 alkyl; and R 34 is hydrogen or C 1 -C 3 alkyl (e.g., methyl).

Also provided is a method of treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof.

In some embodiments, a method is provided of treating a Ras protein-related disorder in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof.

Further provided is a method of inhibiting a Ras protein in a cell, the method comprising contacting the cell with an effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof.

It is specifically contemplated that any limitation discussed with respect to one embodiment of the invention may apply to any other embodiment of the invention. Furthermore, any compound or composition of the invention may be used in any method of the invention, and any method of the invention may be used to produce or to utilize any compound or composition of the invention.

Brief Description of the Figures

FIG. 1A: A compound of the present invention, Compound A, deeply and durably inhibits oncogenic signals in a pancreatic CDX model (HPAC CDX model, PDAC, KRAS G12D/WT). Single dose experiment, n = 3/time point, all dose levels well tolerated.

FIG. 1B: Treatment of KRAS G12D tumors in vivo with a compound of the present invention, Compound A, drives tumor regressions in a pancreatic CDX model (HPAC CDX model, PDAC, KRAS G12D/WT). n = 10/group, * **p<0.001 . All dose levels well tolerated.

Definitions and Chemical Terms

In this application, unless otherwise clear from context, (i) the term "a” means “one or more”; (ii) the term "or" is used to mean "and/or" unless explicitly indicated to refer to alternatives only or the alternative are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and "and/or"; (iii) the terms “comprising" and “including” are understood to encompass itemized components or steps whether presented by themselves or together with one or more additional components or steps; and (iv) where ranges are provided, endpoints are included.

As used herein, the term “about” is used to indicate that a value includes the standard deviation of error for the device or method being employed to determine the value. In certain embodiments, the term “about" refers to a range of values that fall within 25%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less in either direction (greater than or less than) of a stated value, unless otherwise stated or otherwise evident from the context (e.g., where such number would exceed 100% of a possible value).

As used herein, the term “adjacent” in the context of describing adjacent atoms refers to bivalent atoms that are directly connected by a covalent bond.

A "compound of the present invention” and similar terms as used herein, whether explicitly noted or not, refers to Ras inhibitors described herein, including compounds of Formula I and subformula thereof, and compounds of Table 1 and Table 2, as well as salts (e.g., pharmaceutically acceptable salts), solvates, hydrates, stereoisomers (including atropisomers), and tautomers thereof.

The term “wild-type” refers to an entity having a structure or activity as found in nature in a “normal" (as contrasted with mutant, diseased, altered, etc) state or context. Those of ordinary skill in the art will appreciate that wild-type genes and polypeptides often exist in multiple different forms (e.g., alleles).

Those skilled in the art will appreciate that certain compounds described herein can exist in one or more different isomeric (e.g., stereoisomers, geometric isomers, atropisomers, tautomers) or isotopic (e.g., in which one or more atoms has been substituted with a different isotope of the atom, such as hydrogen substituted for deuterium) forms. Unless otherwise indicated or clear from context, a depicted structure can be understood to represent any such isomeric or isotopic form, individually or in combination.

Compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated. Compounds of the present disclosure that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically active starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of olefins, C=N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present disclosure. Cis and trans geometric isomers of the compounds of the present disclosure are described and may be isolated as a mixture of isomers or as separated isomeric forms.

In some embodiments, one or more compounds depicted herein may exist in different tautomeric forms. As will be clear from context, unless explicitly excluded, references to such compounds encompass all such tautomeric forms. In some embodiments, tautomeric forms result from the swapping of a single bond with an adjacent double bond and the concomitant migration of a proton. In certain embodiments, a tautomeric form may be a prototropic tautomer, which is an isomeric protonation states having the same empirical formula and total charge as a reference form. Examples of moieties with prototropic tautomeric forms are ketone - enol pairs, amide - imidic acid pairs, lactam - lactim pairs, amide - imidic acid pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, such as, 1H- and 3H-imidazole, 1H-, 2H- and 4H-1 ,2,4-triazole, 1H- and 2H- isoindole, and 1H- and 2H-pyrazole. In some embodiments, tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution. In certain embodiments, tautomeric forms result from acetal interconversion.

Unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. Exemplary isotopes that can be incorporated into compounds of the present invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, and iodine, such as 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 32 P, 33P, 35 S, 18 F, 36 Cl, 123 | and 125 l. Isotopically-labeled compounds (e.g., those labeled with 3 H and 14 C) can be useful in compound or substrate tissue distribution assays. Tritiated (i.e., 3 H) and carbon-14 (i.e., 14 C) isotopes can be useful for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements). In some embodiments, one or more hydrogen atoms are replaced by 2 H or 3 H, or one or more carbon atoms are replaced by 13 C- or 14 C-enriched carbon. Positron emitting isotopes such as 15 O, 13 N, 11 C, and 18 F are useful for positron emission tomography (PET) studies to examine substrate receptor occupancy. Preparations of isotopically labelled compounds are known to those of skill in the art. For example, isotopically labeled compounds can generally be prepared by following procedures analogous to those disclosed for compounds of the present invention described herein, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.

As is known in the art, many chemical entities can adopt a variety of different solid forms such as, for example, amorphous forms or crystalline forms (e.g., polymorphs, hydrates, solvate). In some embodiments, compounds of the present invention may be utilized in any such form, including in any solid form. In some embodiments, compounds described or depicted herein may be provided or utilized in hydrate or solvate form.

At various places in the present specification, substituents of compounds of the present disclosure are disclosed in groups or in ranges. It is specifically intended that the present disclosure include each and every individual subcombination of the members of such groups and ranges. For example, the term "C 1 -C 6 alkyl" is specifically intended to individually disclose methyl, ethyl, C 3 alkyl, C 4 alkyl, C 5 alkyl, and C 6 alkyl. Furthermore, where a compound includes a plurality of positions at which substituents are disclosed in groups or in ranges, unless otherwise indicated, the present disclosure is intended to cover individual compounds and groups of compounds (e.g., genera and subgenera) containing each and every individual subcombination of members at each position.

The term "optionally substituted X" (e.g., “optionally substituted alkyl") is intended to be equivalent to “X, wherein X is optionally substituted" (e.g., “alkyl, wherein said alkyl is optionally substituted"). It is not intended to mean that the feature “X" (e.g., alkyl) per se is optional. As described herein, certain compounds of interest may contain one or more “optionally substituted" moieties. In general, the term "substituted", whether preceded by the term "optionally" or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent, e.g., any of the substituents or groups described herein. Unless otherwise indicated, an “optionally substituted" group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. For example, in the term “optionally substituted C 1 -C 6 alkyl-C 2 -C 9 heteroaryl,” the alkyl portion, the heteroaryl portion, or both, may be optionally substituted. Combinations of substituents envisioned by the present disclosure are preferably those that result in the formation of stable or chemically feasible compounds. The term “stable”, as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein.

Suitable monovalent substituents on a substitutable carbon atom of an "optionally substituted" group may be, independently, deuterium; halogen; -(CH 2 ) 0 - 4 R°; -(CH 2 ) 0 - 4 OR°; -O(CH 2 ) 0 - 4 R°; -O-(CH 2 ) 0 - 4 C(O)OR°; -(CH 2 ) 0 - 4 CH(OR°) 2 ; -(CH 2 ) 0 - 4 SR°; -(CH 2 ) 0 - 4 Ph, which may be substituted with

R°; -(CH 2 ) 0 - 4 O(CH 2 ) 0 - 1 Ph which may be substituted with R°; -CH=CHPh, which may be substituted with R°; -(CH 2 ) 0 - 4 O(CH 2 )o-i-pyridyl which may be substituted with R°; 4 to 8-membered saturated or unsaturated heterocycloalkyl (e.g., pyridyl); 3 to 8-membered saturated or unsaturated cycloalkyl (e.g., cyclopropyl, cyclobutyl, or cyclopentyl); -NO 2 ; -CN; -Na; -(CH 2 ) 0 - 4 N(R°) 2 ; -(CH 2 ) 0 - 4 N(R°)C(O)R°; -N(R°)C(S)R°; -(CH 2 ) 0 - 4 N(R°)C(O)NR°2; -N(R°)C(S)NR°2; -(CH 2 ) 0 - 4 N(R°)C(O)OR°; -N(R°)N(R°)C(O)R°;

-N(R°)N(R°)C(O)NR°2; -N(R°)N(R°)C(O)OR° ; -(CH 2 ) 0 - 4 C(O)R°; -C(S)R°; -(CH 2 ) 0 - 4 C(O)OR°; -(CH 2 ) 0 - 4 -C(O)-N(R°) 2 ; -(CH 2 ) 0 - 4 -C(O)-N(R°)-S(O) 2 -R°; -C(NCN)NR° 2 ; -(CH 2 ) 0 - 4 C(O)SR°; -(CH 2 ) 0 - 4 C(O)OSi R° 3 ; -(CH 2 ) 0 - 4 OC(O)R°; -OC(O)(CH 2 ) 0 - 4 SR°; -SC(S)SR°; -(CH 2 ) 0 - 4 SC(O)R°; -(CH 2 ) 0 - 4 C(O)NR°2;

-C(S)NR° 2 ; -C(S)SR°; -(CH 2 ) 0 - 4 OC(O)NR°2; -C(O)N(OR°)R°; -C(O)C(O)R°; -C(O)CH 2 C(O)R°; -C(NOR°)R°; -(CH 2 ) 0 - 4 SSR°; -(CH 2 ) 0 - 4 S(O) 2 R°; -(CH 2 ) 0 - 4 S(O) 2 OR°; -(CH 2 ) 0 - 4 OS(O) 2 R°; -S(O) 2 NR° 2 ;

-(CH 2 ) 0 - 4 S(O)R°; -N(R°)S(O) 2 NR° 2 ; -N(R°)S(O) 2 R°; -N(OR°)R°; -C(NOR°)NR° 2 ; -C(NH)NR° 2 ; -P(O) 2 R°; -P(O)R°2; -P(O)(OR°) 2 ; -OP(O)R° 2 ; -OP(O)(OR°) 2 ; -OP(O)(OR°)R°, -SiR° 3 ; -(C 1 -C 4 straight or branched alkylene)O-N(R°) 2 ; or -(C 1 -C 4 straight or branched alkylene)C(O)O-N(R°) 2 , wherein each R° may be substituted as defined below and is independently hydrogen, -C 1 -C 6 aliphatic, -CH 2 Ph, -O(CH 2 ) 0 - 1 Ph, -CH 2 -(5 to 6 membered heteroaryl ring), or a 3 to 6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R°, taken together with their intervening atom(s), form a 3 to 12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, which may be substituted as defined below.

Suitable monovalent substituents on R° (or the ring formed by taking two independent occurrences of R° together with their intervening atoms), may be, independently, halogen, straight or branched alkylene wherein each is unsubstituted or where preceded by “halo" is substituted only with one or more halogens, and is independently selected from C 1 -C 4 aliphatic, -CH 2 Ph, -O(CH 2 ) 0 - 1 Ph, or a 5 to 6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents on a saturated carbon atom of R° include =O and =S.

Suitable divalent substituents on a saturated carbon atom of an “optionally substituted” group include the following: wherein each independent occurrence of is selected from hydrogen, C 1 -C 6 aliphatic which may be substituted as defined below, or an unsubstituted 5 to 6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents that are bound to vicinal substitutable carbons of an “optionally substituted" group include: , wherein each independent occurrence of is selected from hydrogen, C 1 -C 6 aliphatic which may be substituted as defined below, or an unsubstituted 5 to 6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.

Suitable substituents on the aliphatic group of include halogen, , wherein each is unsubstituted or where preceded by "halo” is substituted only with one or more halogens, and is independently C 1 -C 4 aliphatic, -CH 2 Ph, -O(CH 2 ) 0 - 1 Ph, or a 5 to 6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.

Suitable substituents on a substitutable nitrogen of an “optionally substituted” group include -R , -NR 2 , -C(O)R , -C(O)OR , -C(O)C(O)R , -C(O)CH 2 C(O) R , -S(O) 2 R , -S(O) 2 NR 2 , -C(S)NR 2 ,

-C(NH)NR 2 , or -N(R )S(O) 2 R ; wherein each R is independently hydrogen, C 1 -C 6 aliphatic which may be substituted as defined below, unsubstituted -OPh, or an unsubstituted 3 to 6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R , taken together with their intervening atom(s) form an unsubstituted 3 to 12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.

Suitable substituents on an aliphatic group of R are independently halogen, or -NO 2 , wherein each is unsubstituted or where preceded by “halo" is substituted only with one or more halogens, and is independently C 1 -C 4 aliphatic, -CH 2 Ph, -O(CH 2 ) 0 - 1 Ph, or a 5 to 6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents on a saturated carbon atom of R include =O and =S.

The term "acetyl," as used herein, refers to the group -C(O)CH 3 .

The term "alkoxy,” as used herein, refers to a -O-C 1 -C 20 alkyl group, wherein the alkoxy group is attached to the remainder of the compound through an oxygen atom.

The term "alkyl,” as used herein, refers to a saturated, straight or branched monovalent hydrocarbon group containing from 1 to 20 (e.g., from 1 to 10 or from 1 to 6) carbons. In some embodiments, an alkyl group is unbranched (i.e., is linear); in some embodiments, an alkyl group is branched. Alkyl groups are exemplified by, but not limited to, methyl, ethyl, n- and /so-propyl, n-, sec-, iso- and tert-butyl, and neopentyl.

The term "alkylene," as used herein, represents a saturated divalent hydrocarbon group derived from a straight or branched chain saturated hydrocarbon by the removal of two hydrogen atoms, and is exemplified by methylene, ethylene, isopropylene, and the like. The term “C x -C y alkylene” represents alkylene groups having between x and y carbons. Exemplary values for x are 1 , 2, 3, 4, 5, and 6, and exemplary values for y are 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, or 20 (e.g., C 1 -C 6 , C 1 -C 10 , C 2 -C 20 , C 2 -C 6 , C 2 -C 10 , or C 2 -C 20 alkylene). In some embodiments, the alkylene can be further substituted with 1 , 2, 3, or 4 substituent groups as defined herein.

The term “alkenyl,” as used herein, represents monovalent straight or branched chain groups of, unless otherwise specified, from 2 to 20 carbons (e.g., from 2 to 6 or from 2 to 10 carbons) containing one or more carbon-carbon double bonds and is exemplified by ethenyl, 1 -propenyl, 2-propenyl,

2-methyl-1-propenyl, 1-butenyl, and 2-butenyl. Alkenyls include both cis and trans isomers. The term “alkenylene," as used herein, represents a divalent straight or branched chain groups of, unless otherwise specified, from 2 to 20 carbons (e.g., from 2 to 6 or from 2 to 10 carbons) containing one or more carbon-carbon double bonds.

The term “alkynyl,” as used herein, represents monovalent straight or branched chain groups from 2 to 20 carbon atoms (e.g., from 2 to 4, from 2 to 6, or from 2 to 10 carbons) containing a carbon-carbon triple bond and is exemplified by ethynyl, and 1 -propynyl.

The term “alkynyl sulfone,” as used herein, represents a group comprising the structure wherein R is any chemically feasible substituent described herein.

The term “amino," as used herein, represents -N(R ) 2 , e.g., -NH 2 and -N(CH 3 ) 2 .

The term “aminoalkyl,” as used herein, represents an alkyl moiety substituted on one or more carbon atoms with one or more amino moieties.

The term “amino acid," as described herein, refers to a molecule having a side chain, an amino group, and an acid group (e.g., -CO 2 H or -SO 3 H), wherein the amino acid is attached to the parent molecular group by the side chain, amino group, or acid group (e.g., the side chain). As used herein, the term “amino acid” in its broadest sense, refers to any compound or substance that can be incorporated into a polypeptide chain, e.g., through formation of one or more peptide bonds. In some embodiments, an amino acid has the general structure H 2 N-C(H)(R)-COOH. In some embodiments, an amino acid is a naturally-occurring amino acid. In some embodiments, an amino acid is a synthetic amino acid; in some embodiments, an amino acid is a D-amino acid; in some embodiments, an amino acid is an L-amino acid. "Standard amino acid" refers to any of the twenty standard L-amino acids commonly found in naturally occurring peptides. Exemplary amino acids include alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, histidine, optionally substituted hydroxylnorvaline, isoleucine, leucine, lysine, methionine, norvaline, ornithine, phenylalanine, proline, pyrrolysine, selenocysteine, serine, taurine, threonine, tryptophan, tyrosine, and valine.

The term “aryl,” as used herein, represents a monovalent monocyclic, bicyclic, or multicyclic ring system formed by carbon atoms, wherein the ring attached to the pendant group is aromatic. Examples of aryl groups are phenyl, naphthyl, phenanthrenyl, and anthracenyl. An aryl ring can be attached to its pendant group at any heteroatom or carbon ring atom that results in a stable structure and any of the ring atoms can be optionally substituted unless otherwise specified.

The term “C 0 ,” as used herein, represents a bond. For example, part of the term -N(C(O)-(C 0 -C 5 alkylene-H)- includes -N(C(O)-(C 0 alkylene-H)-, which is also represented by -N(C(O)-H)-.

The terms “carbocyclic" and “carbocyclyl,” as used herein, refer to a monovalent, optionally substituted 3 to 12-membered monocyclic, bicyclic, or tricyclic ring structure, which may be bridged, fused or spirocyclic, in which all the rings are formed by carbon atoms and at least one ring is non-aromatic. Carbocyclic structures include cycloalkyl, cycloalkenyl, and cycloalkynyl groups. Examples of carbocyclyl groups are cyclohexyl, cyclohexenyl, cyclooctynyl, 1 ,2-dihydronaphthyl, 1 ,2,3,4-tetrahydronaphthyl, fluorenyl, indenyl, indanyl, decalinyl, and the like. A carbocyclic ring can be attached to its pendant group at any ring atom that results in a stable structure and any of the ring atoms can be optionally substituted unless otherwise specified.

The term “carbonyl," as used herein, represents a C(O) group, which can also be represented as C=O. The term “carboxyl," as used herein, means -CO 2 H, (C=O)(OH), COOH, or C(O)OH or the unprotonated counterparts.

The term “cyano,” as used herein, represents a -CN group.

The term “cycloalkyl," as used herein, represents a monovalent saturated cyclic hydrocarbon group, which may be bridged, fused, or spirocyclic having from three to eight ring carbons, unless otherwise specified, and is exemplified by cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cycloheptyl.

The term “cycloalkenyl,” as used herein, represents a monovalent, non-aromatic, saturated cyclic hydrocarbon group, which may be bridged, fused, or spirocyclic having from three to eight ring carbons, unless otherwise specified, and containing one or more carbon-carbon double bonds.

The term “diastereomer," as used herein, means stereoisomers that are not mirror images of one another and are non-superimposable on one another.

The term “enantiomer," as used herein, means each individual optically active form of a compound of the invention, having an optical purity or enantiomeric excess (as determined by methods standard in the art) of at least 80% (i.e., at least 90% of one enantiomer and at most 10% of the other enantiomer), preferably at least 90% and more preferably at least 98%.

The term “guanidinyl," refers to a group having the structure: , wherein each R is, independently, any any chemically feasible substituent described herein.

The term “guanidinoalkyl alkyl,” as used herein, represents an alkyl moiety substituted on one or more carbon atoms with one or more guanidinyl moieties.

The term “haloacetyl," as used herein, refers to an acetyl group wherein at least one of the hydrogens has been replaced by a halogen.

The term “haloalkyl," as used herein, represents an alkyl moiety substituted on one or more carbon atoms with one or more of the same of different halogen moieties.

The term “halogen," as used herein, represents a halogen selected from bromine, chlorine, iodine, or fluorine.

The term "heteroalkyl," as used herein, refers to an "alkyl" group, as defined herein, in which at least one carbon atom has been replaced with a heteroatom (e.g., an O, N, or S atom). The heteroatom may appear in the middle or at the end of the radical.

The term “heteroaryl," as used herein, represents a monovalent, monocyclic or polycyclic ring structure that contains at least one fully aromatic ring: i.e., they contain 4n+2 pi electrons within the monocyclic or polycyclic ring system and contains at least one ring heteroatom selected from N, O, or S in that aromatic ring. Exemplary unsubstituted heteroaryl groups are of 1 to 12 (e.g., 1 to 11 , 1 to 10, 1 to 9, 2 to 12, 2 to 11 , 2 to 10, or 2 to 9) carbons. The term “heteroaryl” includes bicyclic, tricyclic, and tetracyclic groups in which any of the above heteroaromatic rings is fused to one or more, aryl or carbocyclic rings, e.g., a phenyl ring, or a cyclohexane ring. Examples of heteroaryl groups include, but are not limited to, pyridyl, pyrazolyl, benzooxazolyl, benzoimidazolyl, benzothiazolyl, imidazolyl, thiazolyl, quinolinyl, tetrahydroquinolinyl, and 4-azaindolyl. A heteroaryl ring can be attached to its pendant group at any ring atom that results in a stable structure and any of the ring atoms can be optionally substituted unless otherwise specified. In some embodiment, the heteroaryl is substituted with 1 , 2, 3, or 4 substituents groups.

The term “heterocycloalkyl,” as used herein, represents a monovalent, monocyclic, bicyclic or polycyclic ring system, which may be bridged, fused, or spirocyclic, wherein at least one ring is non- aromatic and wherein the non-aromatic ring contains one, two, three, or four heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur. The 5-membered ring has zero to two double bonds, and the 6- and 7-membered rings have zero to three double bonds. Exemplary unsubstituted heterocycloalkyl groups are of 1 to 12 (e.g., 1 to 11 , 1 to 10, 1 to 9, 2 to 12, 2 to 11 , 2 to 10, or 2 to 9) carbons. The term "heterocycloalkyl” also represents a heterocyclic compound having a bridged multicyclic structure in which one or more carbons or heteroatoms bridges two non-adjacent members of a monocyclic ring, e.g., a quinuclidinyl group. The term “heterocycloalkyl" includes bicyclic, tricyclic, and tetracyclic groups in which any of the above heterocyclic rings is fused to one or more aromatic, carbocyclic, heteroaromatic, or heterocyclic rings, e.g., an aryl ring, a cyclohexane ring, a cyclohexene ring, a cyclopentane ring, a cyclopentene ring, a pyridine ring, or a pyrrolidine ring. Examples of heterocycloalkyl groups are pyrrolidinyl, piperidinyl, 1 ,2,3,4-tetrahydroquinolinyl, decahydroquinolinyl, dihydropyrrolopyridine, and decahydronapthyridinyl. A heterocycloalkyl ring can be attached to its pendant group at any ring atom that results in a stable structure and any of the ring atoms can be optionally substituted unless otherwise specified.

The term “hydroxy," as used herein, represents a -OH group.

The term “hydroxyalkyl," as used herein, represents an alkyl moiety substituted on one or more carbon atoms with one or more -OH moieties.

The term "isomer," as used herein, means any tautomer, stereoisomer, atropiosmer, enantiomer, or diastereomer of any compound of the invention. It is recognized that the compounds of the invention can have one or more chiral centers or double bonds and, therefore, exist as stereoisomers, such as double-bond isomers (i.e., geometric E/Z isomers) or diastereomers (e.g., enantiomers (i.e., (+) or (-)) or cis/trans isomers). According to the invention, the chemical structures depicted herein, and therefore the compounds of the invention, encompass all the corresponding stereoisomers, that is, both the stereomerically pure form (e.g., geometrically pure, enantiomerically pure, or diastereomerically pure) and enantiomeric and stereoisomeric mixtures, e.g., racemates. Enantiomeric and stereoisomeric mixtures of compounds of the invention can typically be resolved into their component enantiomers or stereoisomers by well-known methods, such as chiral-phase gas chromatography, chiral-phase high performance liquid chromatography, crystallizing the compound as a chiral salt complex, or crystallizing the compound in a chiral solvent. Enantiomers and stereoisomers can also be obtained from stereomerically or enantiomerically pure intermediates, reagents, and catalysts by well-known asymmetric synthetic methods.

As used herein, the term “linker" refers to a divalent organic moiety connecting moiety B to moiety W in a compound of Formula I, such that the resulting compound is capable of achieving an IC50 of 2 uM or less in the Ras-RAF disruption assay protocol provided in the Examples below, and provided here:

The purpose of this biochemical assay is to measure the ability of test compounds to facilitate ternary complex formation between a nucleotide-loaded Ras isoform and cyclophilin A; the resulting ternary complex disrupts binding to a BRAF RBD construct, inhibiting Ras signaling through a RAF effector.

In assay buffer containing 25 mM HEPES pH 7.3, 0.002% Tween20, 0.1% BSA, 100 mM NaCl and 5 mM MgCl 2 , tagless Cyciophilin A, His6-K-Ras-GMPPNP (or other Ras variant), and GST-BRAF RBD are combined in a 384-well assay plate at final concentrations of 25 μM, 12.5 nM and 50 nM, respectively. Compound is present in plate wells as a 10-point 3-fold dilution series starting at a final concentration of 30 μM. After incubation at 25°C for 3 hours, a mixture of Anti- His Eu-W1024 and anti-GST allophycocyanin is then added to assay sample wells at final concentrations of 10 nM and 50 nM, respectively, and the reaction incubated for an additional 1 .5 hours. TR-FRET signal is read on a microplate reader (Ex 320 nm, Em 665/615 nm). Compounds that facilitate disruption of a Ras:RAF complex are identified as those eliciting a decrease in the TR-FRET ratio relative to DMSO control wells.

In some embodiments, the linker comprises 20 or fewer linear atoms. In some embodiments, the linker comprises 15 or fewer linear atoms. In some embodiments, the linker comprises 10 or fewer linear atoms. In some embodiments, the linker has a molecular weight of under 500 g/mol. in some embodiments, the linker has a molecular weight of under 400 g/mol. In some embodiments, the linker has a molecular weight of under 300 g/mol. in some embodiments, the linker has a molecular weight of under 200 g/mol. In some embodiments, the linker has a molecular weight of under 100 g/mol. in some embodiments, the linker has a molecular weight of under 50 g/mol.

As used herein, a “monovalent organic moiety” is less than 500 kDa. in some embodiments, a "monovalent organic moiety” is less than 400 kDa. In some embodiments, a “monovalent organic moiety” is less than 300 kDa. In some embodiments, a “monovalent organic moiety” is less than 200 kDa. in some embodiments, a “monovalent organic moiety” is less than 100 kDa. in some embodiments, a "monovalent organic moiety” is less than 50 kDa. In some embodiments, a “monovalent organic moiety” is less than 25 kDa. In some embodiments, a “monovalent organic moiety” is less than 20 kDa. In some embodiments, a “monovalent organic moiety” is less than 15 kDa. in some embodiments, a “monovalent organic moiety” is less than 10 kDa. In some embodiments, a “monovalent organic moiety” is less than 1 kDa. In some embodiments, a "monovalent organic moiety” is less than 500 g/mol. In some embodiments, a “monovalent organic moiety” ranges between 500 g/mol and 500 kDa.

The form “stereoisomer,” as used herein, refers to all possible different isomeric as well as conformational forms which a compound may possess (e.g., a compound of any formula described herein), in particular ail possible stereochemically and conformationally isomeric forms, ail diastereomers, enantiomers or conformers of the basic molecular structure, including atropisomers. Some compounds of the present invention may exist in different tautomeric forms, ail of the latter being included within the scope of the present invention.

The term “sulfonyl,” as used herein, represents an -S(O) 2 - group.

The term “thiocarbonyl,” as used herein, refers to a -C(S)- group.

The term “vinyl ketone,” as used herein, refers to a group comprising a carbonyl group directly connected to a carbon-carbon double bond.

The term “vinyl sulfone,” as used herein, refers to a group comprising a sulfonyl group directed connected to a carbon-carbon double bond. The term “ynone,” as used herein, refers to a group comprising the structure wherein R is any any chemically feasible substituent described herein.

Those of ordinary skill In the art, reading the present disclosure, will appreciate that certain compounds described herein may be provided or utilized in any of a variety of forms such as, for example, salt forms, protected forms, pro-drug forms, ester forms, isomeric forms (e.g., optical or structural isomers), isotopic forms, etc. in some embodiments, reference to a particular compound may relate to a specific form of that compound. In some embodiments, reference to a particular compound may relate to that compound in any form. In some embodiments, for example, a preparation of a single stereoisomer of a compound may be considered to be a different form of the compound than a racemic mixture of the compound; a particular salt of a compound may be considered to be a different form from another salt form of the compound; a preparation containing one conformational isomer ((Z) or (E)) of a double bond may be considered to be a different form from one containing the other conformational isomer ((E) or (Z)) of the double bond; a preparation in which one or more atoms is a different isotope than is present in a reference preparation may be considered to be a different form.

Detailed Description

Compounds

Provided herein are Ras inhibitors. The approach described herein entaiis formation of a high affinity three-component complex, or conjugate, between a synthetic ligand and two intracellular proteins which do not interact under normal physiological conditions: the target protein of interest (e,g., Ras), and a widely expressed cytosolic chaperone (presenter protein) in the cell (e.g., cyclcphilin A). More specifically, in some embodiments, the inhibitors of Ras described herein induce a new binding pocket in Ras by driving formation of a high affinity tri-complex, or conjugate, between the Ras protein and the widely expressed cytosolic chaperone, cyclophilin A (CYRA), Without being bound by theory, the inventors believe that one way the inhibitory effect on Ras is effected by compounds of the invention and the complexes, or conjugates, they form is by steric occlusion of the interaction site between Ras and downstream effector molecules, such as RAF, which are required for propagating the oncogenic Signal.

Without being bound by theory, the inventors postulate that both covalent and non-covaient interactions of a compound of the present invention with Ras and the chaperone protein (e.g., cyclophilin A) may contribute to the inhibition of Ras activity. In some embodiments, a compound of the present invention forms a covalent adduct with a side chain of a Ras protein (e.g., the -CH 2 -COOH or -CH 2 -COO- side chain of the aspartic acid at position 12 or 13 of a mutant Ras protein). Covalent adducts may also be formed with other side chains of Ras. In addition or alternatively, non-covalent interactions may be at play: for example, van der Waals, hydrophobic, hydrophilic, and hydrogen bond interactions, and combinations thereof, may contribute to the ability of the compounds of the present invention to form complexes and act as Ras inhibitors. Accordingly, a variety of Ras proteins may be inhibited by compounds of the present invention (e.g., K-Ras, N-Ras, H-Ras, and mutants thereof at positions 12, 13 and 61 , such as G12C, G12D, G12V, G12S, G13C, G13D, and Q61L, and others described herein). Methods of determining covalent adduct formation are known in the art. One method of determining covalent adduct formation is to perform a “cross-linking” assay, such as described in the Examples, and below:

Note - the following protocol describes a procedure for monitoring cross-linking of K-Ras G12C (GMP-PNP) to a compound of the invention. This protocol may also be executed substituting other Ras proteins or nucleotides, such as K-Ras G12D.

The purpose of this biochemical assay is to measure the ability of test compounds to covalently label nucleotide-loaded K-Ras isoforms. In assay buffer containing 12,5 mM HEPES pH 7.4, 75 mM NaCl, 1 mM MgCl 2 , 1 mM BME, 5 μM Cyclophilin A and 2 μM test compound, a 5 μM stock of GMP-PNP-loaded K-Ras (1-169) G12C is diluted 10-fold to yield a final concentration of 0,5 μM; with final sample volume being 100 μL.

The sample is incubated at 25°C for a time period of up to 24 hours prior to quenching by the addition of 10 μL of 5% Formic Acid. Quenched samples are centrifuged at 15000 rpm for 15 minutes in a benchtop centrifuge before injecting a 10 μL aliquot onto a reverse phase G4 column and eluting into the mass spectrometer with an Increasing acetonitrile gradient in the mobile phase. Analysis of raw data may be carried out using Waters MassLynx MS software, with % bound calculated from the deconvoluted protein peaks for labeled and unlabeled K-Ras. Accordingly, provided herein is a compound, or pharmaceutically acceptable salt thereof, having the structure of Formula I:

Formula I wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 10- membered heteroarylene; B is -CH(R 9 )- or >C=CR 9 R 9’ where the carbon is bound to the carbonyl carbon of -N(R 11 )C(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C 1 -C 4 alkylene, optionally substituted C 1 -C 4 alkenylene, optionally substituted C 1 -C 4 heteroalkylene, -C(O)O-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(O)NH-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C 1 -C 4 heteroalkylene, or 3 to 8-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a carbodiimide, an oxazoline, a thiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziridine, a trifiuoromethyl ketone, a boronic acid, a boronic ester, an N-ethoxycarbonyl-2-ethoxy-1 ,2-dihydroquinoline (EEDQ), an iso-EEDQ or other EEDQ derivative, an epoxide, an oxazolium, or a glycal;

X 1 is optionally substituted C 1 - C 2 alkylene, NR, O, or S(O) n ;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R’, C(O)OR’, C(O)N(R’) 2 , S(O)R’, S(O) 2 R’, or S(O)sN(R’) 2 ; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 is CH, CH 2 , or N;

Y 6 is C(O), CH, CH 2 , or N;

R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl, or

R 1 and R 2 combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 2 is absent, hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered eycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cyclcalkyl or optionally substituted 3 to 7-membered heterocycloalkyl; R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C 1 -C 3 alkyl), C=O, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; R 7a and R 8a are, Independently, hydrogen, halo, optionally substituted C 1 -C 3 alkyl, or combinewith the carbon to which they are attached to form a carbonyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C8 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is hydrogen, F, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl, or

R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 9 is hydrogen or optionally substituted C 1 -C 6 alkyl;

R 10 is hydrogen, halo, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl ;

R 10a is hydrogen or halo; and

R 11 is hydrogen or C 1 -C 3 alkyl; and

R 34 is hydrogen or C 1 -C 3 alkyl (e.g., methyl).

In some embodiments, R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl.

In some embodiments, R 34 is hydrogen.

In some embodiments of compounds of the present invention, G is optionally substituted C 1 -C 4 heteroalkylene.

In some embodiments, a compound of fhe present invention has the structure of Formula la, or a pharmaceutically acceptable salt thereof:

wherein the dotted lines represent zero, one, two, three, or four non-adiacent double bonds;

A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -N(R 11 )C(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a carbodiimide, an oxazoline, a thiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziridine, a trifluoromethyl ketone, a boronlc acid, a boronic ester, an N-ethoxycarbonyl-2-ethoxy-1 ,2-dihydroquinaline (EEDQ), an iso-EEDQ or other EEDQ derivative, an epoxide, an oxazolium, or a glyeal;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 . alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R’, C(O)OR', C(O)N(R’) 2 , S(O)R’, S(O) 2 R’, or S(O) 2 N(R') 2 ; each R' is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 and Y 6 are, independently, CH or N;

R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl;

R 2 is hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O- C 1 -C 3 alkyl), C=O, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cyeioalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 10 is hydrogen, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl; and

R 11 is hydrogen or C 1 -C 3 alkyl.

In some embodiments of compounds of the present Invention, X 2 is NH. In some embodiments, X 3 is CH.

In some embodiments of compounds of the present Invention, R 11 is hydrogen. In some embodiments, R 11 is C 1 -C 3 alkyl, such as methyl.

In some embodiments, a compound of the present invention has the structure of Formula lb, or a pharmaceutically acceptable salt thereof:

Formula lb wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R1 0 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a carbodiimide, an oxazoline, a thiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziridine, a trifluoromethyl ketone, a boronic acid, a boronic ester, an N-ethoxyearbonyl-2-ethoxy-1 ,2-dihydroquinoline (EEDQ), an iso-EEDQ or other EEDQ derivative, an epoxide, an oxazolium, or a glycal; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R’, C(O)OR’, C(O)N(R') 2 ,S(O)R’, S(O) 2 R’, or S(O) 2 N(R’) 2 ; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 and Y 6 are, independently, CH or N;

R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl;

R 2 is hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl; R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C 1 -C 3 alkyl), C=O, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; and

R 10 is hydrogen, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl.

In some embodiments of compounds of the present Invention, X 1 is optionally substituted C 1 -C 2 alkylene. In some embodiments, X 1 is methylene.

In some embodiments of compounds of the present Invention, R 4 is hydrogen.

In some embodiments of compounds of the present Invention, R 5 is hydrogen. In some embodiments, R 5 is C 1 -C 4 alkyl optionally substituted with halogen. In some embodiments, R 5 is methyl.

In some embodiments of compounds of fhe present Invention, Y 4 is C. in some embodiments, R 4 is hydrogen. In some embodiments, Y 5 is CH. in some embodiments, Y 6 is CH. In some embodiments, Y 1 is C. in some embodiments, Y 2 is C. In some embodiments, Y 3 is N. in some embodiments, R 3 is absent. In some embodiments, Y 7 is C.

In some embodiments, a compound of the present invention has the structure of Formula ic, or a pharmaceutically acceptable salt thereof:

Formula lc wherein A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a carbodiimide, an oxazoline, a thiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziridine, a trifluoromethyl ketone, a boronic acid, a boronic ester, an N-ethoxycarbonyl-2-ethoxy-1 ,2-dihydroquinoline (EEDQ), an iso-EEDQ or other EEDQ derivative, an epoxide, an oxazolium, or a glycal;

R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl;

R 2 is hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl:

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C 1 -C 3 alkyl), C=O, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl; R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; and

R 10 is hydrogen, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl.

In some embodiments of compounds of the present invention, R 6 is hydrogen.

In some embodiments of compounds of the present invention, R 2 is hydrogen, cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 6- membered heterocycloalkyl. in some embodiments, R 2 is optionally substituted C 1 -C 6 alkyl, such as ethyl.

In some embodiments of compounds of the present Invention, R 7 is optionally substituted C 1 -C 3 alkyl. In some embodiments, R 7 is C 1 -C 3 alkyl.

In some embodiments of compounds of the present Invention, R 8 is optionally substituted C 1 -C 3 alkyl. In some embodiments, R 8 is C 1 -C 3 alkyl.

In some embodiments, a compound of the present invention has the structure of Formula id, or a pharmaceutically acceptable salt thereof:

Formula Id wherein A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene; B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a carbodiimide, an oxazoline, a thiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziridine, a trifluoromethyl ketone, a boronic acid, a boronic ester, an N-ethoxyearbonyl-2-ethoxy-1 ,2-dihydroquinoline (EEDQ), an iso-EEDQ or other EEDG derivative, an epoxide, an oxazolium, or a glycal; R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl; R 2 is C 1 -C 6 alkyl or 3 to 6-membered cycloalkyl;

R 7 is C 1 -C 3 alkyl;

R 8 is C 1 -C 3 alkyl; and

R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl.

In some embodiments of compounds of the present Invention, R 1 is 5 to 10-membered heteroaryl. in some embodiments, R 1 is optionally substituted 6-membered aryl or optionally substituted 6-membered heteroaryl.

In some embodiments, a compound of the present invention has the structure of Formula le, or a pharmaceutically acceptable salt thereof:

Formula le wherein A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker; W is a cross-linking group comprising a carbodiimide, an oxazoline, a thiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziridine, a trifiuoromethyl ketone, a boronic acid, a boronic ester, an N-ethoxycarbonyl-2-ethoxy-1 ,2-dihydroquinoline (EEDQ), an iso-EEDQ or other EEDQ derivative, an epoxide, an oxazollum, or a glycal; R 2 is C 1 -C 6 alkyl or 3 to 6-membered cycloalkyl;

R 7 is C 1 -C 3 alkyl;

R 8 is C 1 -C 3 alkyl; and R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl

X e is N or CH; and R 12 is optionally substituted C 1 -C 6 alkyl or optionally substituted C 1 -C 6 heteroalkyl.

In some embodiments of compounds of the present Invention, X e is N. In some embodiments, X e is CH. n some embodiments of compounds of the present invention, R 12 is optionally substituted C 1 -C 6 heteroalkyl. in some embodiments, R 12 is . In some embodiments,

In some embodiments, a compound of the present invention has the structure of Formula If, or a pharmaceutically acceptable salt thereof:

Formula If wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene; B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -N(R 11 )C(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene. or 5 to 6-membered heteroarylene;

G is optionally substituted C 1 -C 4 alkylene, optionally substituted C 1 -C 4 alkenylene, optionally substituted C 1 -C 4 heteroalkylene, -C(O)O-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(O)NH-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C 1 -C 4 heteroalkylene, or 3 to 8-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a carbodiimide, an oxazoline, a thiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziridine, a trifluoromethyl ketone, a boronic acid, a boronic ester, an N-ethoxycarbonyl-2-ethoxy-1 ,2-dihydroquinoline (EEDQ), an iso-EEDQ or other EEDQ derivative, an epoxide, an oxazolium, or a glycal:

X 1 is optionally substituted C 1 -C 2 alkylene, NR, O, or S(O) n ;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R’, C(O)OR’, C(O)N(R’) 2 , S(O)R’, S(O) 2 R’, or S(O)sN(R’) 2 ; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, Independently, C or N;

Y 5 and Y 6 are, independently, CH or N;

R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl;

R 2 is hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C 1 -C 3 alkyl), C=O, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 10 is hydrogen, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl; and R 1 ' is hydrogen or C 1 -C 3 alkyl.

In some embodiments, a compound of the present invention has the structure of Formula VI, or a pharmaceutically acceptable salt thereof:

Formula VI wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene (e.g., phenyl or phenol), or optionally substituted 5 to 10-membered heteroarylene;

B is -CH(R 9 )- or >C=CR 9 R 9 where the carbon is bound to the carbonyl carbon of -N(R ,1 )C(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C 1 -C 4 alkylene, optionally substituted C 1 -C* alkenylene, optionally substituted C 1 -C 4 heteroalkylene, -C(O)O-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(O)NH-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C 1 -C 4 heteroalkylene, or 3 to 8-membered heteroarylene;

L is absent or a linker; W is a cross-linking group comprising a carbodiimide, an oxazoline, a thiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziridine, a trifiuoromethyl ketone, a boronic acid, a boronic ester, an N-ethoxycarbonyl-2-ethoxy-1 ,2-dihydroquinoline (EEDQ), an iso-EEDQ or other EEDQ derivative, an epoxide, an oxazolium, or a glycal ;

X 1 is optionally substituted C 1 -C 2 alkylene, NR, O, or S(O) n ;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2; R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R’, C(O)OR’, C(O)N(R’) 2 , S(O)R’, S(O) 2 R * , or S(O) 2 N(R’) 2 ; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 is CH, CH 2 , or N:

Y 6 is C(O), CH, CH 2 , or N;

R 2 is absent, hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C 1 -C 3 alkyl), C=O, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7a and R 8a are, independently, hydrogen, halo, optionally substituted C 1 -C 3 alkyl, or combine with the carbon to which they are attached to form a carbonyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl; R 9 is hydrogen, F, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl, or R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 9' is hydrogen or optionally substituted C 1 -C 6 alkyl; R 10 is hydrogen, halo, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl; R 10a is hydrogen or halo; R 11 is hydrogen or C 1 -C 3 alkyl; R 34 is hydrogen or C 1 -C 3 alkyl; and

X e and X f are, independently, N or CH.

In some embodiments, a compound of the present invention has the structure of Formula Via, or a pharmaceutically acceptable salt thereof:

Formula VIa wherein A is optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene (e.g., phenyl or phenol), or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon ot -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a carbodiimide, an oxazoline, a thiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziridine, a trifluoromethyl ketone, a boronic acid, a boronic ester, an N-ethoxycarbonyl-2-ethoxy-1 ,2-dihydroquinoline (EEDQ), an iso-EEDQ or other EEDQ derivative, an epoxide, an oxazoiium, or a glycal ;

X 1 is optionally substituted C 1 -C 2 alkylene, NR, O, or S(O) n ;

X 2 is O or NH; n is 0, 1 , or 2; R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R', C(O)OR’, C(O)N(R’) 2 , S(O)R’, S(O) 2 R’, or S(O) 2 N(R') 2 ; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl; R 2 is C 1 -C 6 alkyl or 3 to 6-membered cycloalkyl; R 7 is C 1 -C 3 alkyl; R 8 is C 1 -C 3 alkyl; and R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

X e and X f are, independently, N or CH; R 11 is hydrogen or C 1 -C 3 alkyl; and R 21 is hydrogen or C 1 -C 3 alkyl.

In some embodiments of a compound of the present invention, X e is N and X f is CH. In some embodiments, X e is CH and X f is N.

In some embodiments, a compound of the present invention has the structure of Formula Vlb, or a pharmaceutically acceptable salt thereof:

Formula VIb wherein A optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene (e.g., phenyl or phenol), or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a carbodiimide, an oxazoline, a thiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziridine, a trifluoromethyl ketone, a boronic acid, a boronic ester, an N-ethoxycarbonyl-2-ethoxy-1 ,2-dihydroquinoline (EEDQ), an iso-EEDQ or other EEDQ derivative, an epoxide, an oxazolium, or a glycal;

R 9 is optionally substituted Ch-C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; and

X e and X f are, independently, N or CH.

In some embodiments of a compound of the present invention, X e is N and X f is CH. In some embodiments, X e is CH and X f is N. In some embodiments, a compound of the present invention has the structure of Formula VII, or a pharmaceutically acceptable salt thereof: wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 10- membered heteroarylene;

R is -CH(R 9 )- or >C=CR 9 R 9' where the carbon is bound to the carbonyl carbon of -N(R 11 )C(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heteroeyeloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C 1 -C 4 alkylene, optionally substituted Gi-O. alkenylene, optionally substituted C 1 -C 4 heteroalkylene, -C(O)O-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(O)NH-CH(R 8 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C 1 -C 4 heteroalkylene, or 3 to 8-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a carbodiimide, an oxazoline, a thiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziridine, a trifiuoromethyl ketone, a boronic acid, a boronic ester, an N-ethoxycarbonyl-2- ethoxy-1 ,2-dihydroquinoline (EEDQ), an iso -EEDQ or other EEDQ derivative, an epoxide, an oxazolium, or a glycal;

X 1 is optionally substituted C 1 -C 2 alkylene, NR, O, or S(O) n ;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2;

R is hydrogen, cyano. optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R', C(O)OR’, C(O)N(R') 2 , S(O)R’, S(O) 2 R’, or S(O) 2 N{R’i2; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, Independently, C or N;

Y 5 is CH, CH 2 , or N; Y 6 is C(O), CH, CH 2 , or N; R 1 is

R 2 is absent, hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to S-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form G=CR 7' R 8' ; C=N(OH), C=N(O- C 1 -C 3 alkyl), C=O, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7a and R 8a are, Independently, hydrogen, halo, optionally substituted C 1 -C 3 alkyl, or combine with the carbon to which they are attached to form a carbonyl; R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is hydrogen, F, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl, or

R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 9' is hydrogen or optionally substituted C 1 -C 6 alkyl; R 10 is hydrogen, halo, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl; R 10a is hydrogen or halo; R 11 is hydrogen or C 1 -C 3 alkyl; and R 34 is hydrogen or C 1 -C 3 alkyl (e.g., methyl).

In some embodiments of compounds of the present invention, A is optionally substituted 6- membered arylene. in some embodiments, A has the structure: wherein R 13 is hydrogen, hydroxy, amino, optionally substituted C 1 -C 6 alkyl, or optionally substituted C 1 -C 6 heteroalkyl. In some embodiments, R 13 is hydrogen. In some embodiments, R 13 is hydroxy.

In some embodiments of compounds of the present invention, B is -CHR 9 -. In some embodiments, R 9 is optionally substituted C 1 -C 6 alkyl or optionally substituted 3 to 6-membered cycloalkyl. In some embodiments, R 9 is: , , In some embodiments, R 9 is: . in some embodiments, R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl.

In some embodiments, B is optionally substituted 6-membered arylene. In some embodiments, B is 6-membered arylene. in some embodiments, B is:

In some embodiments of compounds of the present invention, R 7 is methyl.

In some embodiments of compounds of the present invention, R 8 is methyl.

In some embodiments, R 34 is hydrogen.

In some embodiments of compounds of the present invention, the linker is the structure of Formula II:

A 1 -(B 1 ) f -(C 1 ) g -(B 2 ) h -( D 1 )-(B 3 ) i -(C 2 ) j -(B 4 ) k -A 2 Formula II where A 1 is a bond between the linker and B; A 2 is a bond between W and the linker; B 1 , B 2 , B 3 , and B 4 each, independently, is selected from optionally substituted C 1 -C 2 alkylene, optionally substituted C 1 -C 3 heteroalkylene, O, S, and NR N ; R N is hydrogen, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, optionally substituted 3 to 14-membered heteroeycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted C 1 -C 7 heteroalkyl; C and C 2 are each, independently, selected from carbonyl, thiocarbonyl, sulphonyl, or phosphoryl; f, g, h, i, j, and k are each, independently, 0 or 1 ; and D 1 is optionally substituted C 1 -C 10 alkylene, optionally substituted C 2 -C 10 alkenylene, optionally substituted C 2 -C 10 alkynylene, optionally substituted 3 to 14- membered heterocycloalkylene, optionally substituted 5 to 10-membered heteroarylene, optionally substituted 3 to 8-membered cycloalkylene, optionally substituted 6 to 10-membered arylene, optionally substituted C 2 -C 10 polyethylene glycolene, or optionally substituted C 1 -C 10 heteroalkylene, or a chemical bond linking A 1 -(B 1 ) f -(C 1 ) g -(B 2 ) h - to -(B 3 ) i -(C 2 ) j -(B 4 ) k -A 2 . In some embodiments, the Iinker is acyclic. In some embodiments, the linker has the structure of Formula IIa:

Formula IIa wherein X a is absent or N;

R 14 is absent, hydrogen or optionally substituted C 1 -C 6 alkyl; and L 2 is absent, -SO 2 -, optionally substituted C 1 -C 4 alkylene or optionally substituted C 1 -C 4 heteroalkylene, wherein at least one of X a , R 14 , or L 2 is present. In some embodiments, the Iinker has the structure: some embodiments, the Iinker is or a comprises a cyclic group. In some embodiments, the Iinker has the structure of Formula lIb:

Formula lIb wherein 0 is 0 or 1 ;

R 15 is hydrogen or optionally substituted C 1 -C 6 alkyl;

Cy is optionally substituted 3 to S-membered cycloalkylene, optionally substituted 3 to 8- membered heterocycloalkylene, optionally substituted 6-10 membered arylene, or optionally substituted 5 to 10-membered heteroarylene; and

L 3 is absent, -SO 2 -, optionally substituted C 1 -C 4 alkylene or optionally substituted C 1 -C 4 heteroalkylene. In some embodiments, the Iinker has the structure:

168

. In some embodiments, a linker of Formula II is selected from the group consisting of

In some embodiments of compounds of the present invention, W comprises a carbodiimide. In some embodiments, W has the structure of Formula IlIa:

Formula IlIa wherein R 14 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 6 to 10-membered aryl, optionally substituted 3 to 14-membered heterocycloalkyl, or optionally substituted 5 to 10-membered heteroaryl. In some embodiments, W has the structure:

In some embodiments, W comprises an oxazoline or thiazoline. in some embodiments, W has the structure of Formula IIIb: Formula lIb wherein X 1 is O or S;

X 2 is absent or NR 19 ;

R 15 , R 16 , R 17 , and R 18 are, independently, hydrogen or optionally substituted C 1 -C 6 alkyl; and R 19 is hydrogen, C(O)(optionally substituted C 1 -C 6 alkyl), optionally substituted C 1 -C 6 alkyl, optionally substituted 6 to 10-membered aryl, optionally substituted 3 to 14-membered heterocycloalkyl, or optionally substituted 5 to 10-membered heteroaryl. In some embodiments, W is

In some embodiments, W comprises a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, or a chloroethyl thiocarbamate. In some embodiments, W has the structure of Formula lllc:

Formula lllc wherein X 3 is O or S;

X 4 is O, S, NR 26 ;

R 21 , R 22 , R 23 , R 24 , and R 26 are, independently, hydrogen or optionally substituted C 1 -C 6 alkyl; and R 26 is hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted 6 to 10-membered aryl, optionally substituted 3 to 14-membered heterocycloalkyl, or optionally substituted 5 to 10-membered heteroaryl. In some embodiments, W is

In some embodiments, W comprises an aziridine. In some embodiments, W has the structure of Formula IIId1 , Formula IIId2, Formula Illd3, or Formula IIId4: wherein X 5 is absent or NR 30 ;

Y is absent or C(O), C(S), S(O), SO 2 , or optionally substituted C 1 -C 3 alkylene; R 27 is hydrogen, -C(O)R 32 , -C(O)OR 32 , -SOR 33 , -SO 2 R 33 , optionally substituted C 1 -C 6 alkyl, optionally substituted 6 to 10-membered aryl, optionally substituted 3 to 14-membered heterocycloalkyl, or optionally substituted 5 to 10-membered heteroaryl; R 28 and R 29 are, independently, hydrogen, CN, C(O)R 31 , CO 2 R 31 , C(O)R 31 R 31 optionally substituted C 1 -C 6 alkyl, optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 6 to 10- membered aryl, optionally substituted 3 to 14-membered heterocycloalkyl, or optionally substituted 5 to 10-membered heteroaryl; each R 31 is, independently, hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted 6 to 10-membered aryl, optionally substituted 3 to 14-membered heterocycloalkyl, or optionally substituted 5 to 10-membered heteroaryl;

R 30 is hydrogen or optionally substituted C 1 -C 6 alkyl; and

R 32 and R 33 are, independently, hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted 6 to 10-membered aryl, optionally substituted 3 to 14-membered heterocycloalkyl, or optionally substituted 5 to 10-membered heteroaryl. In some embodiments, W is: In some embodiments, W comprises an epoxide. In some embodiments, W is

In some embodiments, W is a cross-linking group bound to an organic moiety that is a Ras binding moiety, i.e., RBM-W, wherein upon contact of an RBM-W compound with a Ras protein, the RBM- W binds to the Ras protein to form a conjugate. For example, the W moiety of an RBM-W compound may bind, e.g., cross-link, with an amino acid of the Ras protein to form the conjugate. In some embodiments, the Ras binding moiety is a K-Ras binding moiety. In some embodiments, the K-Ras binding moiety binds to a residue of a K-Ras Switch-Il binding pocket of the K-Ras protein. In some embodiments, the Ras binding moiety is an H-Ras binding moiety that binds to a residue of an H-Ras Switch-Il binding pocket of an H-Ras protein. In some embodiments, the Ras binding moiety is an N-Ras binding moiety that binds to a residue of an N-Ras Switch-Il binding pocket of an N-Ras protein. The W of an RBM-W compound may comprise any W described herein. The Ras binding moiety typically has a molecular weight of under 1200 Da. See, e.g., see, e.g., Johnson et al., 292:12981-12993 (2017) for a description of Ras protein domains, incorporated herein by reference.

In some embodiments, a compound of the present invention is selected from Table 1 , or a pharmaceutically acceptable salt or stereoisomer thereof. In some embodiments, a compound of the present invention is selected from Table 1 , or a pharmaceutically acceptable salt or atropisomer thereof.

Table 1 : Certain Compounds of the Present invention

* Stereochemistry of the aziridine carbon is assumed.

Note that some compounds are shown with bonds as fiat or wedged. In some instances, the relative stereochemistry of stereoisomers has been determined; in some instances, the absolute stereochemistry has been determined. In some Instances, a single Example number corresponds to a mixture of stereoisomers. All stereoisomers of the compounds of the foregoing table are contemplated by the present invention. In particular embodiments, an atropisomer of a compound of the foregoing table is contemplated. In some embodiments, a compound of Table 2 is provided, or a pharmaceutically acceptable salt thereof. In some embodiments, a compound of the present invention is selected from Table 2 or a pharmaceutically acceptable salt or atropisomer thereof. Table 2: Certain Compounds of the Present invention

Note that some compounds are shown with bonds as fiat or wedged. In some instances, the relative stereochemistry of stereoisomers has been determined; in some instances, the absolute stereochemistry has been determined. All stereoisomers of the compounds of the foregoing table are contemplated by the present invention. In particular embodiments, an atropisomer of a compound of the foregoing table is contemplated . in some embodiments, a compound of the present invention is or acts as a prodrug, such as with respect to administration to a cell or to a subject in need thereof.

Also provided are pharmaceutical compositions comprising a compound of the present invention, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.

Further provided is a conjugate, or salt thereof, comprising the structure of Formula IV:

M-L-P Formula IV wherein L is a linker; P is a monovalent organic moiety; and

M has the structure of Formula Va: wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds; A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene; B is -CH(R 9 )- or >C=CR 9 R 9 where the carbon is bound to the carbonyl carbon of -N(R 11 )C(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C 1 -C 4 alkylene. optionally substituted C 1 -C 4 alkenylene, optionally substituted C 1 -C 4 heteroalkylene, -C(O)O-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(O)NH-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C 1 -C 4 heteroalkylene, or 3 to 8-membered heteroarylene;

X 1 is optionally substituted C 1 -C 2 alkylene, NR, G, or S(O) n ;

X 2 is C or NH;

X 3 is N or CH; n is 0, 1 , or 2; R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R’, C(O)OR’, C(O)N(R’) 2 , S(O)R’, S(O) 2 R’, or S(O) 2 N(R’) 2 ; each R' is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, Independently, C or N;

Y 5 is CH, CH 2 , or N;

Y 6 is C(O), CH, CH 2 , or N;

R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl, or

R 1 and R 2 combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 2 is absent, hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C 1 -C 3 alkyl), C= O, C= S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7a and R 8a are, independently, hydrogen, halo, optionally substituted C 1 -C 3 alkyl, or combine with the carbon to which they are attached to form a carbonyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl, or

R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 9 is hydrogen or optionally substituted C 1 -C 6 alkyl;

R 10 is hydrogen, halo, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl;

R 10a is hydrogen or halo; and

R 1 ' is hydrogen or C 1 -C 3 alkyl. in some embodiments, the conjugate has the structure of Formula IV:

M-L-P Formula IV wherein L is a linker;

P is a monovalent organic moiety; and

M has the structure of Formula Vb: wherein the dotted lines represent zero, one, two, three, or four non-adlacent double bonds: A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -N(R 11 )C(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C 1 -C 4 alkylene. optionally substituted C 1 -C 4 alkenylene, optionally substituted C 1 -C 4 heteroalkylene, -C(O)O-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(O)NH-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C 1 -C 4 heteroalkylene, or 3 to 8-membered heteroarylene;

X 1 is optionally substituted C 1 -C 2 alkylene, NR, O, or S(O) n ;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R’, C(O)OR’, C(O)N(R’) 2 , S(O)R’, S(O) 2 R’, or S(O)sN(R’) s ; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, Independently, C or N;

Y 5 and Y 6 are, independently, CH or N;

R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl;

R 2 is hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C 1 -C 3 alkyl), C= O, C= S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl; R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; R 10 is hydrogen, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl; and

R 11 is hydrogen or C 1 -C 3 alkyl.

In some embodiments, the conjugate has the structure of Formula IV:

M-L-P Formula IV wherein L is a linker;

P is a monovalent organic moiety; and

M has the structure of Formula Vc:

Formula Vc wherein A is optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene (e.g., phenyl or phenol), or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heteroeycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

X 1 is optionally substituted C 1 -C 2 alkylene, NR, O, or S(O) n ;

X 2 is O or NH; n is 0, 1 , or 2; R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R', C(O)OR’, C(O)N(R’) 2 , S(O)R’, S(O) 2 R’, or S(O) 2 N(R’) 2 ; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl;

X e and X f are, independently, N or CH; R 2 is C 1 -C 6 alkyl or 3 to 6-membered cycloalkyl;

R 7 is C 1 -C 3 alkyl; R 8 is C 1 -C 3 alkyl; and R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; R 11 is hydrogen or C 1 -C 3 alkyl; and R 34 is hydrogen or C 1 -C 3 alkyl.

In some embodiments of a compound of the present invention, X e is N and X f is CH. in some embodiments, X e is CH and X f is N.

In some embodiments, the conjugate has the structure of Formula IV:

M-L-P

Formula IV wherein L is a linker;

P is a monovalent organic moiety; and M has the structure of Formula Vd:

Formula Vd wherein A optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene (e.g., phenyl or phenol), or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a carbodiimide, an oxazoline, a thiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziridine, a trlfiuoromethyl ketone, a boronic acid, a boronic ester, an N-ethoxycarbonyl-2- ethoxy-1 ,2-dihydroquinoline (EEDQ), an iso-EEDQ or other EEDQ derivative, an epoxide, an oxazolium, or a glycal; R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; and

X e and X f are, independently, N or CH.

In some embodiments of a compound of the present invention, X e is N and X f is CH. In some embodiments, X e is CH and X f is N.

In some embodiments of conjugates of the present invention, the linker has the structure of Formula II:

A 1 -(B 1 ) f -(C 1 ) g -(B 2 ) h -(D 1 )-(B 3 ) i -(C 2 ) j -(B 4 ) k -A 2 Formula II where A 1 is a bond between the linker and B; A 2 is a bond between P and the linker; B 1 , B 2 , B 3 , and B 4 each, independently, is selected from optionally substituted C 1 -C 2 alkylene, optionally substituted C 1 -C 3 heteroalkylene, O, S, and NR N ; R N is hydrogen, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, optionally substituted 3 to 14-membered heterocycloalkyl , optionally substituted 6 to 10-membered aryl, or optionally substituted C 1 -C 7 heteroalkyl;C 1 and C 2 are each, independently, selected from carbonyl, thiocarbonyl, sulphonyl, or phosphoryl; f, g, h, i, j, and k are each, independently, 0 or 1 ; and D 1 is optionally substituted C 1 -C 10 alkylene, optionally substituted C 2 -C 10 alkenylene, optionally substituted C 2 -C 10 alkynylene, optionally substituted 3 to 14- membered heterocycloalkylene, optionally substituted 5 to 10-membered heteroarylene, optionally substituted 3 to 8-membered cycloalkylene, optionally substituted 6 to 10-membered arylene, optionally substituted C 2 -C 10 polyethylene glycolene, or optionally substituted C 1 -C 10 heteroalkylene, or a chemical bond linking A 1 -(B 1 ) f -(C 1 ) g -(B 2 ) h -to-(B 3 ) i -(C 2 ) j -(B 4 ) k -A 2 . In some embodiments of conjugates of the present invention, the linker is bound to the monovalent organic moiety through a bond to a carboxyl group of an amino acid residue of the monovalent organic moiety.

In some embodiments of conjugates of the present invention, the monovalent organic moiety is a protein. In some embodiments, the protein is a Ras protein. In some embodiments, the Ras protein is K- Ras G12D or K-Ras G13D.

Further provided is a method of treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof. The cancer may, for example, be pancreatic cancer, colorectal cancer, non-small cell lung cancer, acute myeloid leukemia, multiple myeloma, thyroid gland adenocarcinoma, a myelodysplastic syndrome, or squamous cell lung carcinoma. In some embodiments, the cancer comprises a Ras mutation, such as K-Ras G12D or K-Ras G13D. Other Ras mutations are described herein.

Further provided is a method of treating a Ras protein -related disorder in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof.

Further provided is a method of inhibiting a Ras protein in a cell, the method comprising contacting the cell with an effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof. For example, the Ras protein is K-Ras G12D or K-Ras G13D. Other Ras proteins are described herein. The cell may be a cancer cell, such as a pancreatic cancer cell, a colorectal cancer cell, a non-small cell lung cancer cell, an acute myeloid leukemia cell, a multiple myeloma cell, a thyroid gland adenocarcinoma cell, a myelodysplastic syndrome cell, or a squamous cell lung carcinoma cell. Other cancer types are described herein. The cell may be in vivo or in vitro.

With respect to compounds of the present invention, one stereoisomer may exhibit better inhibition than another stereoisomer. For example, one atropisomer may exhibit inhibition, whereas the other atropisomer may exhibit little or no inhibition,

Methods of Synthesis

The compounds described herein may be made from commercially available starting materials or synthesized using known organic, inorganic, or enzymatic processes. The compounds of the present invention can be prepared in a number of ways well known to those skilled in the art of organic synthesis. By way of example, compounds of the present invention can be synthesized using the methods described in the Schemes below, together with synthetic methods known in the art of synthetic organic chemistry, or variations thereon as appreciated by those skilled in the art. These methods include but are not limited to those methods described In the Schemes below. Compounds of Table 1 herein were prepared using methods disclosed herein or were prepared using methods disclosed herein combined with the knowledge of one of skill in the art. Compounds of Table 2 may be prepared using methods disclosed herein or may be prepared using methods disclosed herein combined with the knowledge of one of skill in the art. Scheme 1. General synthesis of macrocyclic esters

A general synthesis of macrocyclic esters is outlined in Scheme 1 . An appropriately substituted aryl-3-(5-bromo-1-ethyl-1H-indol-3-yl)-2,2-dimethylpropan-1- ol (1) can be prepared in three steps starting from protected 3-(5-bromo-2-iodo-1H-indol-3-yl)-2,2-dimethylpropan-1-ol and appropriately substituted boronic acid, including palladium mediated coupling, alkylation, and de-protection reactions.

Methyl-amino-hexahydropyridazine-3-carboxylate-boronic ester (2) can be prepared in three steps, Including protection, iridium catalyst mediated borylation, and coupling with methyl (S)- hexahydropyridazine-3-carboxylate.

The final macrocyclic esters can be made by coupling of methyl-amino-hexahydropyridazine-3- carboxylate-boronic ester (2) and aryl-3-(5-bromo-1-ethyl-1H-indol-3-yl)-2,2-dimethylpropan-1- ol (1) in the presence of Pd catalyst followed by hydrolysis and macroiactonization steps to result in an appropriately protected macrocyclic Intermediate (4). Additional deprotection or functionalization steps are required to produce a final compound. For example, a person of skill in the art would be able to install into a macrocyclic ester a desired -B-L-W group of a compound of Formula (I), where B, L and W are defined herein, including by using methods exemplified in certain Schemes below and in the Example section herein.

Scheme 2, Alternative general synthesis of macrocyclic esters

Alternatively, macrocyclic esters can be prepared as described in Scheme 2. An appropriately protected bromo-indolyl (5) can be coupled in the presence of Pd catalyst with boronic ester (3), followed by iodination, deprotection, and ester hydrolysis. Subsequent coupling with methyl (S)- hexahydropyridazine-3-carboxylate, followed by hydrolysis and macroiactonization can result in iodo intermediate (6). Coupling in the presence of Pd catalyst with an appropriately substituted boronic ester can yield fully a protected macrocycle (4). Additional deprotection or functionalization steps are required to produce a final compound. For example, a person of skill in the art would be able to install into a macrocyclic ester a desired -B-L-W group of a compound of Formula (I), where B, L and W are defined herein, including by using methods exemplified in certain Schemes below and in the Example section herein. Scheme 3. General synthesis of aziridine containing macrocycles

As shown In Scheme 3, compounds of this type may be prepared by the reaction of an appropriate amine (1) with an aziridine containing carboxylic acid (2) in the presence of standard amide coupling reagents, followed by deprofection of the aziridine, if R 1 is a protecting group, and deprotection of the phenol, if required, to produce the final compound (4).

Scheme 4. General synthesis of carbodiimide containing macrocycles

As shown In Scheme 4, compounds of this type may be prepared by the reaction of an appropriate amine (1) with a thiourea containing carboxylic acid (2) in the presence of standard amide coupling reagents, followed by conversion of the thiourea (3) to a carbodiimide (4) in the presence of 2- chloro-1-methylpyridin-1-ium iodide.

Scheme 5, General synthesis of chloroethyl urea containing macrocycles

As shown In Scheme 5, compounds of this type may be prepared by the reaction of an appropriate amine (1) with an isocyanate (2) under basic conditions, followed by deprotection of the phenol, if required, to produce the final compound (4).

Scheme 6. General synthesis of amino oxazoline containing macrocycles

As shown in Scheme 6, compounds of this type may be prepared by cyclization of an appropriate chloroethyl urea (1) under elevated temperatures to produce the final compound (2).

Scheme 7, General synthesis of epoxide containing macrocycles

As shown in Scheme 7, compounds of this type may be prepared by the reaction of an appropriate amine (1) with an epoxide containing carboxylic acid (2) in the presence of standard amide coupling reagents to produce the final compound (3).

In addition, compounds of the disclosure can be synthesized using the methods described in the Examples below, together with synthetic methods known in the art of synthetic organic chemistry, or variations thereon as appreciated by those skilled in the art. These methods include but are not limited to those methods described in the Examples below. For example, a person of skill in the art would be able to install into a macrocyclic ester a desired -B-L-W group of a compound of Formula (I), where B, L and W are defined herein, including by using methods exemplified in certain Schemes above and in the Example section herein.

Pharmaceutical Compositions and Methods of Use Pharmaceutical Compositions and Methods of Administration

The compounds with which the invention is concerned are Ras inhibitors, and are useful in the treatment of cancer. Accordingly, one embodiment of the present invention provides pharmaceutical compositions containing a compound of the invention or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, as well as methods of using the compounds of the invention to prepare such compositions.

As used herein, the term “pharmaceutical composition” refers to a compound, such as a compound of the present invention, or a pharmaceutically acceptable salt thereof, formulated together with a pharmaceutically acceptable excipient.

In some embodiments, a compound is present in a pharmaceutical composition In unit dose amount appropriate for administration in a therapeutic regimen that shows a statistically significant probability of achieving a predetermined therapeutic effect when administered to a relevant population, in some embodiments, pharmaceutical compositions may be specially formulated for administration in solid or liquid form, including those adapted for the following: oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), tablets, e.g,, those targeted for buccal, sublingual, and systemic absorption, boluses, powders, granules, pastes for application to the tongue; parenteral administration, for example, by subcutaneous, intramuscular, intravenous or epidural Injection as, for example, a sterile solution or suspension, or sustained-release formulation; topical application, for example, as a cream, ointment, or a controlled-release patch or spray applied to the skin, lungs, or oral cavity; intravaginally or intrarectally, for example, as a pessary, cream, or foam; sublingually; ocularly; transdermally; or nasally, pulmonary, and to other mucosal surfaces.

A ‘pharmaceutically acceptable excipient,” as used herein, refers any inactive ingredient (for example, a vehicle capable of suspending or dissolving the active compound) having the properties of being nontoxic and non-inflammatory in a subject. Typical excipients include, for example: antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspensing or dispersing agents, sweeteners, or waters of hydration. Excipients include, but are not limited to: butylated optionally substituted hydroxyltoiuene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmellose, crosslinked polyvinyl pyrrolidone, citric acid, crospovidone, cysteine, ethylcellulose, geiatin, optionally substituted hydroxylpropyl cellulose, optionally substituted hydroxylpropyl methylcellulose, lactose, magnesium stearate, maititol, mannitol, methionine, methylcellulose, methyl paraben, microcrystalline cellulose, polyethylene glycol, polyvinyl pyrrolidone, povidone, pregelatinized starch, propyl paraben, retinyl palmitate, shellac, silicon dioxide, sodium carboxymethyl cellulose, sodium citrate, sodium starch glycolate, sorbitol, starch (corn), stearic acid, stearic acid, sucrose, talc, titanium dioxide, vitamin A, vitamin E, vitamin G, and xylitol. Those of ordinary skill in the art are familiar with a variety of agents and materials useful as excipients. See, e.g., e.g., Ansel, et al., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems. Philadelphia: Lippincott, Williams & Wilkins, 2004; Gennaro, et al., Remington: The Science and Practice of Pharmacy. Philadelphia: Lippincott, Williams & Wilkins, 2000; and Rowe, Handbook of Pharmaceutical Excipients. Chicago, Pharmaceutical Press, 2005. In some embodiments, a composition includes at least two different pharmaceutically acceptable excipients.

Compounds described herein, whether expressly stated or not, may be provided or utilized in salt form, e.g., a pharmaceutically acceptable salt form, unless expressly stated to the contrary. The term "pharmaceutically acceptable salt,” as use herein, refers to those salts of the compounds described herein that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and other animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, pharmaceutically acceptable salts are described in: Berge et al., J. Pharmaceutical Sciences 66:1-19, 1977 and In Pharmaceutical Saits: Properties, Seiection, and Use, (Eds. P.H. Stahl and C.G. Wermuth), Wiley-VCH, 2008. The salts can be prepared in situ during the final isolation and purification of the compounds described herein or separately by reacting the free base group with a suitable organic acid.

The compounds of the invention may have ionizable groups so as to be capable of preparation as pharmaceutically acceptable salts. These salts may be acid addition salts involving inorganic or organic acids or the salts may, in the case of acidic forms of the compounds of the invention, be prepared from inorganic or organic bases. In some embodiments, the compounds are prepared or used as pharmaceutically acceptable salts prepared as addition products of pharmaceutically acceptable acids or bases. Suitable pharmaceutically acceptable acids and bases are well-known in the art, such as hydrochloric, sulfuric, hydrobromic, acetic, lactic, citric, or tartaric acids for forming acid addition salts, and potassium hydroxide, sodium hydroxide, ammonium hydroxide, caffeine, various amines, and the like for forming basic salts. Methods for preparation of the appropriate salts are well-established in the art.

Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-optionally substituted hydroxyl-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, paimitate. pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivaiate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, toluenesulfonate, undecanoate, valerate salts, and the like. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like.

As used herein, the term "subject” refers to any member of the animal kingdom. In some embodiments, "subject” refers to humans, at any stage of development. In some embodiments, “subject" refers to a human patient. In some embodiments, “subject" refers to non-human animals. In some embodiments, the non-human animal is a mammal (e.g., a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a sheep, cattle, a primate, or a pig). In some embodiments, subjects include, but are not limited to, mammals, birds, reptiles, amphibians, fish, or worms. In some embodiments, a subject may be a transgenic animal, genetically-engineered animal, or a clone.

As used herein, the term "dosage form” refers to a physically discrete unit of a compound (e.g., a compound of the present invention) for administration to a subject. Each unit contains a predetermined quantify of compound. In some embodiments, such quantity is a unit dosage amount (or a whole fraction thereof) appropriate for administration in accordance with a dosing regimen that has been determined to correlate with a desired or beneficial outcome when administered to a relevant population (l.e., with a therapeutic dosing regimen). Those of ordinary skill in the art appreciate that the total amount of a therapeutic composition or compound administered to a particular subject is determined by one or more attending physicians and may involve administration of multiple dosage forms.

As used herein, the term “dosing regimen” refers to a set of unit doses (typically more than one) that are administered individually to a subject, typically separated by periods of time. In some embodiments, a given therapeutic compound (e.g,, a compound of the present invention) has a recommended dosing regimen, which may involve one or more doses. In some embodiments, a dosing regimen comprises a plurality of doses each of which are separated from one another by a time period of the same length; in some embodiments, a dosing regimen comprises a plurality of doses and at least two different time periods separating individual doses. In some embodiments, all doses within a dosing regimen are of the same unit dose amount. In some embodiments, different doses within a dosing regimen are of different amounts. In some embodiments, a dosing regimen comprises a first dose In a first dose amount, followed by one or more additional doses in a second dose amount different from the first dose amount. In some embodiments, a dosing regimen comprises a first dose in a first dose amount, followed by one or more additional doses in a second dose amount same as the first dose amount. In some embodiments, a dosing regimen is correlated with a desired or beneficial outcome when administered across a relevant population (i.e., is a therapeutic dosing regimen).

A “therapeutic regimen" refers to a dosing regimen whose administration across a relevant population is correlated with a desired or beneficial therapeutic outcome.

The term “treatment” (also “treat” or “treating"), in its broadest sense, refers to any administration of a substance (e.g., a compound of the present invention) that partially or completely alleviates, ameliorates, relieves, inhibits, delays onset of, reduces severity of, or reduces incidence of one or more symptoms, features, or causes of a particular disease, disorder, or condition. In some embodiments, such treatment may be administered to a subject who does not exhibit signs of the relevant disease, disorder or condition, or of a subject who exhibits only early signs of the disease, disorder, or condition. Alternatively, or additionally, In some embodiments, treatment may be administered to a subject who exhibits one or more established signs of the relevant disease, disorder, or condition. In some embodiments, treatment may be of a subject who has been diagnosed as suffering from the relevant disease, disorder, or condition. In some embodiments, treatment may be of a subject known to have one or more susceptibility factors that are statistically correlated with increased risk of development of the relevant disease, disorder, or condition.

The term “therapeutically effective amount” means an amount that is sufficient, when administered to a population suffering from or susceptible to a disease, disorder, or condition In accordance with a therapeutic dosing regimen, to treat the disease, disorder, or condition. In some embodiments, a therapeutically effective amount is one that reduces the incidence or severity of, or delays onset of, one or more symptoms of the disease, disorder, or condition. Those of ordinary skill in the art will appreciate that the form “therapeutically effective amount” does not in fact require successful treatment be achieved in a particular individual. Rather, a therapeutically effective amount may be that amount that provides a particular desired pharmacological response in a significant number of subjects when administered to patients in need of such treatment. It is specifically understood that particular subjects may, in tact, be “refractory” to a “therapeutically effective amount.” In some embodiments, reference to a therapeutically effective amount may be a reference to an amount as measured in one or more specific tissues (e.g., a tissue affected by the disease, disorder or condition) or fluids (e.g., blood, saliva, serum, sweat, tears, urine). Those of ordinary skill in the art will appreciate that, in some embodiments, a therapeutically effective amount may be formulated or administered in a single dose. In some embodiments, a therapeutically effective amount may be formulated or administered in a plurality of doses, for example, as part of a dosing regimen.

For use as treatment of subjects, the compounds of the invention, or a pharmaceutically acceptable salt thereof, can be formulated as pharmaceutical or veterinary compositions. Depending on the subject to be treated, the mode of administration, and the type of treatment desired, e.g., prevention, prophylaxis, or therapy, the compounds, or a pharmaceutically acceptable salt thereof, are formulated in ways consonant with these parameters. A summary of such techniques may be found in Remington: The Science and Practice of Pharmacy, 21 st Edition, Lippincott Williams & Wilkins, (2005); and Encyclopedia of Pharmaceutical Technofogy, eds. J. Swarbrick and J. C. Boylan, 1988-1999, Marcel Dekker, New York, each of which is incorporated herein by reference.

Compositions can be prepared according to conventional mixing, granulating, or coating methods, respectively, and the present pharmaceutical compositions can contain from about 0.1% to about 99%, from about 5% to about 90%, or from about 1% to about 20% of a compound of the present invention, or pharmaceutically acceptable salt thereof, by weight or volume. In some embodiments, compounds, or a pharmaceutically acceptable salt thereof, described herein may be present in amounts totaling 1-95% by weight of the total weight of a composition, such as a pharmaceutical composition.

The composition may be provided In a dosage form that is suitable for intraarticular, oral, parenteral (e.g., intravenous, intramuscular), rectal, cutaneous, subcutaneous, topical, transdermai, sublingual, nasal, vaginal, intravesicular, intraurethrai, intrathecal, epidural, aural, or ocular administration, or by Injection, inhalation, or direct contact with the nasal, genitourinary, reproductive, or oral mucosa. Thus, the pharmaceutical composition may be in the form of, e.g., tablets, capsules, pills, powders, granulates, suspensions, emulsions, solutions, gels including hydrogels, pastes, ointments, creams, plasters, drenches, osmotic delivery devices, suppositories, enemas, injectabies, implants, sprays, preparations suitable for iontophoretic delivery, or aerosols. The compositions may bo formulated according to conventional pharmaceutical practice.

As used herein, the term "administration” refers to the administration of a composition (e.g., a compound, or a preparation that includes a compound as described herein) to a subject or system. Administration to an animal subject (e.g., to a human) may be by any appropriate route. For example, in some embodiments, administration may be bronchial (including by bronchial instillation), buccal, enteral, interdermai, intra-arterial, intradermal, intragastrlc, intramedullary, intramuscular, intranasal, intraperitoneai, intrathecal, Intravenous, intraventricular, mucosal, nasal, oral, rectal, subcutaneous, sublingual, topical, tracheal (including by intratracheal instillation), transdermai, vaginal, or vitreal.

Formulations may be prepared in a manner suitable tor systemic administration or topical or local administration. Systemic formulations include those designed for injection (e.g., intramuscular, intravenous or subcutaneous injection) or may be prepared for transdermai, transmucosai, or oral administration. A formulation will generally include a diluent as well as, in some cases, adjuvants, buffers, preservatives and the like. Compounds, or a pharmaceutically acceptable salt thereof, can be administered also in liposomal compositions or as microemulsions.

For injection, formulations can be prepared in conventional forms as liquid solutions or suspensions or as solid forms suitable for solution or suspension in liquid prior to injection or as emulsions. Suitable excipients include, for example, water, saline, dextrose, glycerol, and the like. Such compositions may also contain amounts of nontoxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents, and the like, such as, for example, sodium acetate, sorbitan monolaurate, and so forth.

Various sustained release systems for drugs have also been devised. See, for example, U.S. Patent No. 5,624,677.

Systemic administration may also include relatively noninvasive methods such as the use of suppositories, transdermai patches, transmucosai delivery, and intranasal administration. Oral administration is also suitable for compounds of the Invention, or pharmaceutically acceptable salts thereof. Suitable forms Include syrups, capsules, and tablets, as is understood in the art.

Each compound, or a pharmaceutically acceptable salt thereof, as described herein, may be formulated in a variety of ways that are known in the art. For example, the first and second agents of the combination therapy may be formulated together or separately. Other modalities of combination therapy arc described herein.

The individually or separately formulated agents can be packaged together as a kit. Non-limiting examples include, but are not limited to, kits that contain, e.g., two pills, a pill and a powder, a suppository and a liquid in a vial, two topical creams, etc. The kit can include optional components that aid in the administration of the unit dose to subjects, such as vials for reconstituting powder forms, syringes for injection, customized IV delivery systems, inhalers, etc. Additionally, the unit dose kit can contain instructions for preparation and administration of the compositions. The kit may be manufactured as a single use unit dose for one subject, multiple uses for a particular subject (at a constant dose or in which the individual compounds, or a pharmaceutically acceptable salt thereof, may vary in potency as therapy progresses); or the kit may contain multiple doses suitable for administration to multiple subjects (“bulk packaging"). The kit components may be assembled in cartons, blister packs, bottles, tubes, and the like.

Formulations for oral use include tablets containing the active ingredient(s) in a mixture with non-toxic pharmaceutically acceptable excipients. These excipients may be, for example, inert diluents or fillers (e.g., sucrose, sorbitol, sugar, mannitol, microcrystalline cellulose, starches including potato starch, calcium carbonate, sodium chloride, lactose, calcium phosphate, calcium sulfate, or sodium phosphate); granulating and disintegrating agents (e.g., cellulose derivatives including microcrystalline cellulose, starches including potato starch, croscarmellose sodium, alginates, or alginic acid); binding agents (e.g., sucrose, glucose, sorbitol, acacia, alginic acid, sodium alginate, gelatin, starch, pregelatinized starch, microcrystalline cellulose, magnesium aluminum silicate, carboxymethylcellulose sodium, methylcellulose, optionally substituted hydroxylpropyl methyleellulose, ethylcellulose, polyvinylpyrrolidone, or polyethylene glycol); and lubricating agents, glidants, and antiadhesives (e.g., magnesium stearate, zinc stearate, stearic acid, silicas, hydrogenated vegetable oils, or talc). Other pharmaceutically acceptable excipients can be colorants, flavoring agents, plasticizers, humectants, buffering agents, and the like.

Two or more compounds may be mixed together in a tablet, capsule, or other vehicle, or may be partitioned, !n one example, the first compound is contained on the inside of the tablet, and the second compound is on the outside, such that a substantial portion of the second compound is released prior to the release of the first compound.

Formulations for oral use may also be provided as chewable tablets, or as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent (e.g., potato starch, lactose, microcrystailine cellulose, calcium carbonate, calcium phosphate or kaolin), or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin, or olive oil. Powders, granulates, and pellets may be prepared using the ingredients mentioned above under tablets and capsules in a conventional manner using, e.g., a mixer, a fluid bed apparatus or a spray drying equipment.

Dissolution or diffusion-controlled release can be achieved by appropriate coating of a tablet, capsule, pellet, or granulate formulation of compounds, or by incorporating the compound, or a pharmaceutically acceptable salt thereof, Into an appropriate matrix. A controlled release coating may include one or more of the coating substances mentioned above or, e.g., shellac, beeswax, glycowax, castor wax, carnauba wax, stearyl alcohol, glyceryl monostearate, glyceryl distearate, glycerol palmitostearate, ethylcellulose, acrylic resins, dl-polylactic acid, cellulose acetate butyrate, polyvinyl chloride, polyvinyl acetate, vinyl pyrroiidone, polyethylene, polymethacrylate, methylmethacrylate, 2-optionally substituted hydroxylmethacrylate, methacrylate hydrogels, 1 ,3 butylene glycol, ethylene glycol methacrylate, or polyethylene glycols. In a controlled release matrix formulation, the matrix material may also Include, e.g., hydrated methylcellulose, carnauba wax and stearyl alcohol, carbopol

934, silicone, glyceryl tristearate, methyl acrylate-methyl methacrylate, polyvinyl chloride, polyethylene, or halogenated fluorocarbon.

The liquid forms In which the compounds, or a pharmaceutically acceptable salt thereof, and compositions of the present invention can be incorporated for administration orally Include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil. or peanut oil, as well as elixirs and similar pharmaceutical vehicles.

Generally, when administered to a human, the oral dosage of any of the compounds of the invention, or a pharmaceutically acceptable sait thereof, will depend on the nature of the compound, and can readlly be determined by one skilled in the art. A dosage may be, for example, about 0.001 mg to about 2000 mg per day, about 1 mg to about 1000 mg per day, about 5 mg to about 500 mg per day, about 100 mg to about 1500 mg per day, about 500 mg to about 1500 mg per day, about 500 mg to about 2000 mg per day, or any range derivable therein.

In some embodiments, the pharmaceutical composition may further comprise an additional compound having antiproliferative activity. Depending on the mode of administration, compounds, or a pharmaceutically acceptable salt thereof, will be formulated into suitable compositions to permit facile delivery. Each compound, or a pharmaceutically acceptable salt thereof, of a combination therapy may be formulated in a variety of ways that are known in the art. For example, the first and second agents of the combination therapy may be formulated together or separately. Desirably, the first and second agents are formulated together for the simultaneous or near simultaneous administration of the agents.

It will be appreciated that the compounds and pharmaceutical compositions of the present invention can be formulated and employed in combination therapies, that is, the compounds and pharmaceutical compositions can be formulated with or administered concurrently with, prior to, or subsequent to, one or more other desired therapeutics or medical procedures. The particular combination of therapies (therapeutics or procedures) to employ in a combination regimen will take Into account compatibility of the desired therapeutics or procedures and the desired therapeutic effect to be achieved. It will also be appreciated that the therapies employed may achieve a desired effect for the same disorder, or they may achieve different effects (e.g., control of any adverse effects).

Administration of each drug in a combination therapy, as described herein, can, independently, be one to four times dally for one day to one year, and may even be for the life of the subject. Chronic, long-term administration may be indicated.

Numbered Embodiments

[1] A compound, or pharmaceutically acceptable salt thereof, having the structure of Formula I: Formula I wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 10- membered heteroarylene;

R is -CH(R 9 )- or >C=CR 9 R 9' where the carbon is bound to the carbonyl carbon of -N(R 11 )C(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C 1 -C 4 alkylene, optionally substituted Gi-O. alkenylene, optionally substituted C 1 -C 4 heteroalkylene, -C(O)O-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(O)NH-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C 1 -C 4 heteroalkylene, or 3 to 8-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a carbodiimide, an oxazoline, a thiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziridine, a trlfluoromethyl ketone, a boronic acid, a boronic ester, an N -ethoxycarbonyl-2- ethoxy-1 ,2-dihydroquinoline (EEDQ), an iso-EEDQ or other EEDQ derivative, an epoxide, an oxazolium, or a glycal;

X' is optionally substituted C 1 -C 2 alkylene, NR, O, or S(G) r< ;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R', C(O)OR’, C(O)N(R') 2 , S(O)R’, S(O) 2 R’, or S(G) 2 N(R’)2; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, Independently, C or N;

Y 5 is CH, CH 2 or N;

Y 6 is C(O), CH, CH 2 , or N:

R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl, or

R 1 and R 2 combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 2 is absent, hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl; R 4 is absent, hydrogen, halogen, eyano, or methyl optionally substituted with 1 to 3 halogens; R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl; R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl; R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(0G:-C 3 alkyl), C=O, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7a and R 8a are, independently, hydrogen, halo, optionally substituted C 1 -C 3 alkyl, or combine with the carbon to which they are attached to form a carbonyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, eyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8’ combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is hydrogen, F, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl , or

R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 9’ is hydrogen or optionally substituted C 1 -C 6 alkyl;

R 10 is hydrogen, halo, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl;

R 10a is hydrogen or halo;

R 11 is hydrogen or C 1 -C 3 alkyl; and

R 34 is hydrogen or C 1 -C 3 alkyl.

[2] The compound, or pharmaceutically acceptable salt thereof, of paragraph [1], wherein G is optionally substituted C 1 -C 4 heteroalkylene.

[3] The compound, or pharmaceutically acceptable salt thereof, of paragraph [1] or [2], wherein the compound has the structure of Formula la:

Formula la wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-memberod heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -N(R 11 )C(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a carbodiimide, an oxazoline, a thiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziridine, a trifluoromethyl ketone, a boronic acid, a boronic ester, an N-ethoxycarbonyl-2-ethoxy-1 ,2-dihydroquinoline (EEDQ), an iso-EEDQ or other EEDQ derivative, an epoxide, an oxazolium, or a glycal;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C -C alkynyl, C(O)R’, C(O)OR’, C(O)N(R’) 2 , S(O)R’, S(O) 2 R’, or S(O)sN(R’) s ; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, Independently, C or N;

Y 5 and Y 6 are, independently, CH or N;

R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl;

R 2 is hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl; R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C 1 -C 3 alkyl), C=O, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8’ combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 10 is hydrogen, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl; and R 11 is hydrogen or C 1 -C 3 alkyl.

[4] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[3], wherein X 2 is NH.

[5] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[4], wherein X 3 is CH.

[6] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[5], wherein R 11 is hydrogen.

[7] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [5], wherein R 11 is C 1 -C 3 alkyl.

[8] The compound, or pharmaceutically acceptable salt thereof, of paragraph [7], wherein R 11 is methyl. [9] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [6], wherein the compound has the structure of Formula lb:

Formula lb wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a carbodiimide, an oxazoline, a thiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziribine, a trifluoromethyl ketone, a boronic acid, a boronic ester, an N -ethoxycarbonyl-2-ethoxy-1 ,2-dihydroquinoline (EEDQ), an iso-EEDQ or other EEDQ derivative, an epoxide, an oxazolium, or a glycal; n is 0, 1 , or 2: R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R', C(O)OR’, C(O)N(R’) 2 , S(O)R', S(O) 2 R’, or S(O) 2 N(R') 2 ; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CM, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 and Y 6 are, independently, CH or N; R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl; R 2 is hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl; R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C 1 -C 3 alkyl), C=O, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cyeioalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; and

R 10 is hydrogen, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl.

[10] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [9] wherein X 1 is optionally substituted C 1 -C 2 alkylene.

[11] The compound, or pharmaceutically acceptable salt thereof, of paragraph [10], wherein X 1 is methylene.

[12] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [11], wherein R 5 is hydrogen.

[13] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [11], wherein R 5 is C 1 -C 4 alkyl optionally substituted with halogen,

[14] The compound, or pharmaceutically acceptable salt thereof, of paragraph [13], wherein R 5 is methyl.

[15] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[14], wherein Y 4 is C.

[16] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[15], wherein R 4 is hydrogen. [17] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[16], wherein Y 5 is CH.

[18] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[17], wherein Y s is CH.

[19] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[18], wherein Y 1 is C.

[20] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[19], wherein Y 2 is C.

[21] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[20], wherein Y 3 is N.

[22] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[21], wherein R 3 is absent.

[23] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[22], wherein Y 7 is C.

[24] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [6] or [9] to [23], wherein the compound has the structure of Formula Ic:

Formula lc wherein A is -N(H or CH 3 )C(O)-(CH 2 )· where the amino nitrogen is bound to the carbon atom of - CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heteroeyeloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a carbodiimide, an oxazoline, a thiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziridine, a trifluoromethyl ketone, a boronic acid, a boronic ester, an N-ethoxycarbonyl-2-ethoxy-1 ,2-dihydroquinoline (EEDQ), an iso-EEDQ or other EEDQ derivative, an epoxide, an oxazoiium, or a glycal ;

R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl or optionally substituted 5 to 10-membered heteroaryl; R 2 is hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membored heterocycloalkyl optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl; R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl; R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl; R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C 1 -C 3 alkyl), C=O, C=S, C=NH, optionally substituted 3 to 6-membered eycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; and

R 10 is hydrogen, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl.

[25] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[24], wherein R 6 is hydrogen.

[26] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[25], wherein R 2 is hydrogen, cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted 3 to 6- mombered cycloalkyl, or optionally substituted 3 to 6-membered heterocycloalkyl.

[27] The compound, or pharmaceutically acceptable salt thereof, of paragraph [26], wherein R 2 is optionally substituted C 1 -C 6 alkyl.

[28] The compound, or pharmaceutically acceptable salt thereof, of paragraph [27], wherein R 2 is ethyl.

[29] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs 11] to [28], wherein R 7 is optionally substituted C 1 -C 3 alkyl. [30] The compound, or pharmaceutically acceptable salt thereof, of paragraph [29], wherein R 7 is C 1 -C 3 alkyl,

[31] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [30], wherein R 8 is optionally substituted C 1 -C 3 alkyl.

[32] The compound, or pharmaceutically acceptable salt thereof, of paragraph [31], wherein R 8 is C 1 -C 3 alkyl.

[33] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [32], wherein the compound has the structure of Formula Id:

Formula Id wherein A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a carbodiimide, an oxazoline, a ihiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziridine, a trifluoromethyl ketone, a boronic acid, a boronic ester, an N-ethoxycarbonyl-2-ethoxy-1 ,2-dihydroquinoline (EEDQ), an iso-EEDQ or other EEDQ derivative, an epoxide, an oxazoiium, or a glycal;

R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl;

R 2 is C 1 -C 6 alkyl or 3 to 6-membered cycloalkyl;

R 7 is C 1 -C 3 alkyl;

R 8 is C 1 -C 3 alkyl; and

R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl,

[34] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [33], wherein R 1 is 5 to 10-membered heteroaryl. [35] The compound, or pharmaceutically acceptable salt thereof, of paragraph [34], wherein R' is optionally substituted 6-membered aryl or optionally substituted 6-membered heteroaryl.

[36] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [35], wherein the compound has the structure of Formula ie:

Formula le wherein A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionaliy substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a carbodlimide, an oxazoline, a thiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziridine, a trifluoromethyl ketone, a boronlc acid, a boronic ester, an N-ethoxycarbonyl-2-ethoxy-1 ,2-dihydroquinoline (EEDQ), an iso-EEDQ or other EEDQ derivative, an epoxide, an oxazoiium, or a glycal;

R 2 is C 1 -C 6 alkyl or 3 to 6-membered cycloalkyl;

R 7 is C 1 -C 3 alkyl;

R 8 is C 1 -C 3 alkyl; and

R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl

X e and X f are, independently, N or CH; and

R 12 is optionally substituted C 1 -C 6 alkyl or optionally substituted C 1 -C 6 heteroalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl,

[37] The compound, or pharmaceutically acceptable salt thereof, of paragraph [36], wherein X e is N and X f is CH.

[38] The compound, or pharmaceutically acceptable salt thereof, of paragraph [36], wherein X e is CH and X f is N. [39] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [36] to [38], wherein R 12 is optionally substituted C 1 -C 6 heteroalkyl.

[40] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [36] to [39], wherein R 12 is

[41] The compound, or pharmaceutically acceptable salt thereof, of paragraph [1] or [2], wherein the compound has the structure of Formula VI:

Formula VI wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 10- membered heteroarylene;

B is -CH(R 9 )- or >C=OR S R 9 where the carbon is bound to the carbonyl carbon of -N(R 11 )C(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C 1 -C 4 alkylene, optionally substituted C 1 -C 4 alkenylene, optionally substituted C 1 -C 4 heteroalkylene, -C(O)O-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(O)NH-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C 1 -C 4 heteroalkylene, or 3 to 8-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a carbodiimide, an oxazoline, a thiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziridine, a trifluoromethyl ketone, a boronic acid, a boronic ester, an N-ethoxycarbonyl-2-ethoxy-1 .2-dihydroquinoline (EEDQ), an iso-EEDQ or other EEDQ derivative, an epoxide, an oxazoiium, or a glycal;

X 1 is optionally substituted C 1 -C 2 alkylene, NR, O, or S(O) n ;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2; R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R', C(O)OR’, C(O)N(R’) 2 , S(O)R’, S(O) 2 R’, or S(O) 2 N(R') 2 ; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CM, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 is CH, CH 2 , or N;

Y 6 is C(O), CH, CH 2 , or N; R 2 is absent, hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2~ C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or eyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R B is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cyeioalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C ^ CR 7' R 8' ; CYN(OH), ON(O-C 1 -C 3 alkyl), C=O, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7a and R 83 are, independently, hydrogen, halo, optionally substituted C 1 -C 3 alkyl, or combine with the carbon to which they are attached to form a carbonyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is hydrogen, F, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl, or

R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 9’ is hydrogen or optionally substituted C 1 -C 6 alkyl;

R 10 is hydrogen, halo, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl; R 10a is hydrogen or halo: R 11 is hydrogen or C 1 -C 3 alkyl; R 34 is hydrogen or C 1 -C 3 alkyl; and

X e and X f are, independently, N or CH.

[42] The compound, or pharmaceutically acceptable salt thereof, of paragraph [41 ], wherein the compound has the structure of Formula VIa:

Formula VIa wherein A is optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene. optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a carbodiimide, an oxazoline, a thiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziridine, a trifluoromethyl ketone, a boronic acid, a boronic ester, an N-ethoxycarbonyl-2-ethoxy-1 ,2-dihydroquinoline (EEDQ), an iso-EEDQ or other EEDQ derivative, an epoxide, an oxazolium, or a glycal;

X 1 is optionally substituted C 1 -C 2 alkylene, NR, O, or S(O) n ;

X 2 is O or NH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R’, C(O)OR', C(O)N(R’) 2 , S(O)R’, S(O) 2 R’, or S(O) 2 N(R’) 2 ; each R' is, independently, H or optionally substituted C 1 -C 4 alkyl;

R 2 is C 1 -C 6 alkyl or 3 to 6-membered cycloalkyl;

R 7 is C 1 -Cs alkyl;

R 8' is C;-C 3 alkyl; and

R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

X 8 and X f are, independently, N or CH;

R 11 is hydrogen or C 1 -C 3 alkyl; and R 21 is hydrogen or C 1 -C 3 alkyl.

[43] The compound, or pharmaceutically acceptable salt thereof, of paragraph [41] or [42], wherein the compound has the structure of Formula VIb:

Formula VIb wherein A optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a carbodiimide, an oxazoline, a thiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziridine, a trifiuoromethyl ketone, a boronic acid, a boronic ester, an N-ethoxycarbonyl-2-ethoxy-1 ,2-dihydroquinoline (EEDQ), an iso-EEDQ or other EEDQ derivative, an epoxide, an oxazoiium, or a glycal;

R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; and

X e and X are, independently, N or CH.

[44] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [43], wherein A is optionally substituted 6-membered arylene.

[45] The compound, or pharmaceutically acceptable salt thereof, of paragraph [44], wherein A has the structure: wherein R 13 is hydrogen, hydroxy, amino, optionally substituted C 1 -C 6 alkyl, or optionally substituted C 1 -C 6 heteroalkyl.

[46] The compound, or pharmaceutically acceptable salt thereof, of paragraph [45], wherein R 13 is hydrogen. [47] The compound, or pharmaceutically acceptable salt thereof, of paragraph [45], wherein R 13 is hydroxy.

[48] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [47], wherein B is -CHR 9 -.

[49] The compound, or pharmaceutically acceptable salt thereof, of paragraph [48], wherein R 9 is optionally substituted C 1 -C 6 alkyl or optionally substituted 3 to 6-membered cycloalkyl.

[50] The compound, or pharmaceutically acceptable salt thereof, of paragraph [49], wherein R 9

[51] The compound, or pharmaceutically acceptable salt thereof, of paragraph [50], wherein R 9

[52] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[47], wherein B is optionally substituted 6-membered arylene.

[53] The compound, or pharmaceutically acceptable salt thereof, of paragraph [52], wherein B is 6-membered arylene.

[54] The compound, or pharmaceutically acceptable salt thereof, of paragraph [53], wherein B is:

[55] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[54], wherein R 7 is methyl.

[56] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[55], wherein R 8 is methyl.

[57] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[56], wherein the linker is the structure of Formula P:

A 1 -(B 1 ) f -(C 1 ) g -(B 2 ) h -(D 1 )-(B 3 ) i -(C 2 ) j -(B 4 ) k -A 2

Formula II where A 1 is a bond between the linker and B; A 2 is a bond between W and the linker; B 1 , B 2 , B 3 , and B 4 each, independently, is selected from optionally substituted C 1 -C 2 alkylene, optionally substituted C 1 -C 3 heteroalkylene, O, S, and NR N ; R N is hydrogen, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 8 to 10-membered aryl, or optionally substituted C 1 -C 7 heteroalkyl; C 1 and C 2 are each, independently, selected from carbonyl, thiocarbonyl, sulphonyl, or phosphoryl; f, g, h, i, j, and k are each, independently, 0 or 1 ; and D 1 is optionally substituted C 1 -C 10 alkylene, optionally substituted C 2 -C 10 alkenylene, optionally substituted C 2 -C 10 alkynylene, optionally substituted 3 to 14- membered heterocycloalkylene, optionally substituted 5 to 10-membered heteroarylene, optionally substituted 3 to 8-membered cycloalkylene, optionally substituted 6 to 10-membered arylene, optionally substituted C 2 -C 10 polyethylene glycolene, or optionally substituted C 1 -C 10 heteroalkylene, or a chemical bond linking A 1 -(B 1 ) f -(C 1 ) g -(B 2 ) h - to -(B 3 ) i -(C 2 ) j -(B 4 ) k -A 2 . [58] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [57], wherein the linker is acyclic.

[59] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [58], wherein the linker has the structure of Formula iia:

Formula IIa wherein X 3 is absent or N;

R 14 is absent, hydrogen or optionally substituted C 1 -C 6 alkyl; and L 2 is absent, -SO 2 -, optionally substituted C 1 -C 4 alkylene or optionally substituted C 1 -C 4 heteroalkylene, wherein at least one of X a , R 14 , or L 2 is present.

[60] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [59], wherein the linker has the structure:

[81] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [57], wherein the linker is or a comprises a cyclic group.

[62] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [57] or [61], wherein the linker has the structure of Formula lIb:

Formula lIb wherein 0 is 0 or 1 ;

R 15 is hydrogen or optionally substituted C 1 -C 6 alkyl;

Cy is optionally substituted 3 to 8-membered cycloalkylene, optionally substituted 3 to 8- membered heterocycloalkylene, optionally substituted 6-10 membered arylene, or optionally substituted 5 to 10-membered heteroarylene; and L 3 is absent, -80s-, optionally substituted C 1 -C 4 alkylene or optionally substituted C 1 -C 4 heteroalkylene.

[63] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [62], wherein the linker has the structure:

[64] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [63], wherein W comprises a carbodiimide.

[65] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [64], wherein W has the structure of Formula IIIa: Formula IlIa wherein R 14 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 6 to 10-membered aryl, optionally substituted 3 to 14-membered heterocycloalkyl, or optionally substituted 5 to 10-membered heteroaryl.

[66] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [65], wherein W has the structure:

[67] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [63], wherein W comprises an oxazoline or thiazoline.

[68] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [67], wherein W has the structure of Formula IIIb: wherein X 1 is O or S;

X 2 is absent or NR 19 ;

R 15 , R 16 , R 17 , and R 1S are, independently, hydrogen or optionally substituted C 1 -C 6 alkyl; and R 19 is hydrogen, C(O)(optionally substituted C 1 -C 6 alkyl), optionally substituted C 1 -C 6 alkyl, optionally substituted 6 to 10-membered aryl, optionally substituted 3 to 14-membered heterocycloalkyl, or optionally substituted 5 to 10-membered heteroaryl.

[69] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [68], wherein W

[70] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [63], wherein W comprises a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, or a chloroethyl thiocarbamate.

[71] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [70], wherein W has the structure of Formula IIIc: wherein X 3 is O or S;

X 4 is O, S, NR 26 ;

R 21 , R 22 , R 23 , R 24 , and R 26 are, independently, hydrogen or optionally substituted C 1 -C 6 alkyl; and R 25 is hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted 6 to 10-membered aryl, optionally substituted 3 to 14-membered heterocycloalkyl, or optionally substituted 5 to 10-membered heteroaryl.

[72] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [71], wherein W

[73] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [63], wherein W comprises an aziridine. [74] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [73], wherein W has the structure of Formula IIId1 , Formula Mld2, Formula U!d3, or Formula II!d4:

Formula IIId1 Formula IIId2 Formula IIId3 Formula IIId4 wherein X s is absent or NR 30 ;

Y is absent or C(O), C{S), S(O), SO?, or optionally substituted C 1 -C 3 alkylene;

R 27 is hydrogen, -C(O)R 32 , -C(O)OR 32 , -SO 2 R 33 , -SOP. 33 , optionally substituted C 1 -C 6 alkyl, optionally substituted 6 to 10-membered aryl, optionally substituted 3 to 14-membered heterocycloalkyl or optionally substituted 5 to 10-membered heteroaryl;

R 28 and R 29 are, independently, hydrogen, CN, C(O)R 31 , CO 2 R 31 , C(O)R 3, R 31 optionally substituted C 1 -C 6 alkyl, optionally substituted 3 to 10-membered cycloalkyl, optionally substituted 6 to 10- membered aryl, optionally substituted 3 to 14-membered heterocycloalkyl, or optionally substituted 5 to 10-membered heteroaryl; each R 31 is, independently, hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted 6 to 10-membered aryl, optionally substituted 3 to 14-membered heterocycloalkyl, or optionally substituted 5 to 10-membered heteroaryl;

R 30 is hydrogen or optionally substituted C 1 -C 6 alkyl; and

R 32 and R 33 are, independently, hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted 6 to 10-membered aryl, optionally substituted 3 to 14-membered heterocycloalkyl, or optionally substituted 5 to 10-membered heteroaryl,

[75] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [73] or [74], wherein W is:

[76] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [63], wherein W comprises an epoxide.

[77] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [76], wherein W

[78] A compound, or a pharmaceutically acceptable salt thereof, of Table 1 or Table 2.

[79] A pharmaceutical composition comprising a compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [78] and a pharmaceutically acceptable excipient.

[80] A conjugate, or salt thereof, comprising the structure of Formula IV:

M-L-P Formula IV wherein L is a linker;

P is a monovalent organic moiety; and M has the structure of Formula V: wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene;

B is -CH(R 9 )- or >C=CR 9 R 9' where the carbon is bound to the carbonyl carbon of -N(R 11 )C(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C 1 -C 4 alkylene, optionally substituted C 1 -C 4 alkenylene, optionally substituted C 1 -C 4 heteroalkylene, -C(O)O-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(O)NH-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C 1 -C 4 heteroalkylene, or 3 to 8-membered heteroarylene;

X 1 is optionally substituted C 1 -C 2 alkylene, NR, G, or S(O) n ;

X 2 is O or NH:

X 3 is N or CH; n is 0, 1 , or 2; R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R', C(O)OR’, C(O)N(R’) 2 , S(O)R’, S(O) 2 R’, or S(O) 2 N(R') 2 ; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, Independently, C or N ;

Y 5 is CH, CH 2 , or N;

Y 6 is C(O), CH, CH 2 , or N; R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl, or

R 1 and R 2 combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;; R 2 is absent, hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl; R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens; R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl; R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl; R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C 1 -C 3 alkyl), C=O, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7a and R 8a are, independently, hydrogen, halo, optionally substituted C 1 -C 3 alkyl, or combine with the carbon to which they are attached to form a carbonyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxyl, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cyeioalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is hydrogen, F, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl, or

R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 9’ is hydrogen or optionally substituted C 1 -C 6 alkyl;

R 10a is hydrogen or halo;

R 11 is hydrogen or C 1 -C 3 alkyl; and

R 34 is hydrogen or C 1 -C 3 alkyl.

[81] A conjugate, or salt thereof, of paragraph [80], wherein M has the structure of Formula Vc:

Formula Vc wherein A is optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted S-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )·- where the carbon is bound to the carbonyl carbon of -NI ~ IC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6--membered arylene, or 5 to 8--membered heteroarylene;

X' is optionally substituted C 1 -C 2 alkylene, NR, O, or S(O)n;

X 2 is O or NH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R’, C(O)OR’, C(O)N(R’) 2 , S(O)R’, S(O) 2 R’, or S(O) 2 N{R’i2; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl;

X e and X f are, independently, N or CH;

R 2 is C 1 -C 6 alkyl or 3 to 6-membered cycloalkyl;

R 7 is C 1 -Ca alkyl;

R 8 is C 1 -C 3 alkyl; and

R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-merrsbered heterocycloalkyl;

R 11 is hydrogen or C 1 -C 3 alkyl; and

R 34 is hydrogen or C 1 -C 3 alkyl.

In some embodiments of a compound of the present invention, X e is N and X f is CH. In some embodiments, X e is CH and X f is N. [82] The conjugate, or salt thereof, of paragraph [80] or [81], wherein M has the structure of

Formula Vd:

Formula Vd wherein A optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a carbodiimide, an oxazoline, a thiazoline, a chloroethyl urea, a chloroethyl thiourea, a chloroethyl carbamate, a chloroethyl thiocarbamate, an aziridine, a trifiuoromethyl ketone, a boronic acid, a boronic ester, an N-ethoxycarbonyl-2-ethoxy-1 ,2-dihydroquinoline (EEDQ), an iso-EEDQ or other EEDQ derivative, an epoxide, an oxazolium, or a glycal;

R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; and

X e and X* are, independently, N or CH.

[83] The conjugate, or salt thereof, of any one of paragraphs [80] to [82], wherein the linker has the structure of Formula II:

A 1 -(B 1 ) f -(C 1 ) g -(B 2 ) h -(D 1 )-(B 3 ) i -(C 2 ) j -(B 4 ) k -A 2

Formula M where A 1 is a bond between the linker and B; A 2 is a bond between P and the linker; B 1 , B 2 , B 3 , and B 4 each, independently, is selected from optionally substituted C 1 -C 2 alkylene, optionally substituted C 1 -C 3 heteroalkylene, O, S, and NR N ; R N is hydrogen, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted C 1 -C 7 heteroalkyl; C 1 and C 2 are each, independently, selected from carbonyl, thiocarbonyl, sulphonyl, or phosphoryl; f, g, h, i, j, and k are each, independently, 0 or 1 ; and D 1 is optionally substituted C 1 -C 10 alkylene, optionally substituted C 2 -C 10 alkenylene, optionally substituted C 2 -C 10 alkynylene, optionally substituted 3 to 14- membered heterocycloalkylene, optionally substituted 5 to 10-membered heteroarylene, optionally substituted 3 to 8-membered cycloalkylene, optionally substituted 6 to 10-membered arylene, optionally substituted C 2 -C 10 polyethylene glycolene, or optionally substituted C 1 -C 10 heteroalkylene, or a chemical bond linking A 1 -(B 1 ) f -(C 1 ) g -(B 2 ) h - to -(B 3 ) i -(C 2 ) j -(B 4 ) k -A 2 .

[84] The conjugate, or salt thereof, of any one of paragraphs [80] to [83], wherein the monovalent organic moiety is a protein.

[85] The conjugate, or salt thereof, of paragraph [84], wherein the protein is a Ras protein.

[86] The conjugate, or salt thereof, of paragraph [85], wherein the Ras protein is K-Ras G12D or K-Ras G13D.

[87] The conjugate, or salt thereof, of any one of paragraphs [80] to [86], wherein the linker is bound to the monovalent organic moiety through a bond to a carboxyl group of an amino acid residue of the monovalent organic moiety.

[88] A method of treating cancer In a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [78] or a pharmaceutical composition of paragraph [79].

[89] The method of paragraph [88], wherein the cancer is pancreatic cancer, non-small cell lung cancer, colorectal cancer or endometrial cancer.

[90] The method of paragraph [88] or [89], wherein the cancer comprises a Ras mutation.

[91] The method of paragraph [90], wherein the Ras mutation is K-Ras G12D or K-Ras G13D.

[92] A method of treating a Ras protein-related disorder in a subject In need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [78] or a pharmaceutical composition of paragraph [79],

[93] A method of Inhibiting a Ras protein in a cell, the method comprising contacting the cell with an effective amount of a compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [78] or a pharmaceutical composition of paragraph [79],

[94] The method of paragraph [92] or [93], wherein the Ras protein is K-Ras G12D or K-Ras

G13D.

[95] The method of paragraph [93] or [94], wherein the celi is a cancer cell.

[96] The method of paragraph [95], wherein the cancer cell is a pancreatic cancer cell, a non- small cell lung cancer cell, a colorectal cancer celi, or an endometrial celi.

Examples

The disclosure is further Illustrated by the following examples and synthesis examples, which are not to be construed as limiting this disclosure in scope or spirit to the specific procedures herein described. It is to be understood that the examples are provided to Illustrate certain embodiments and that no limitation to the scope of the disclosure is intended thereby. It is to be further understood that resort may be had to various other embodiments, modifications, and equivalents thereof which may suggest themselves to those skilled in the art without departing from the spirit of the present disclosure or scope of the appended claims.

Chemical Syntheses Definitions used in the following examples and elsewhere herein are: instrumentation

Mass spectrometry data collection took place with a Shimadzu LCMS-2020 or Waters Acquity UPLC with either a GDa detector or SG Detector 2. Samples were injected in their liquid phase onto a G- 18 reverse phase column to remove assay buffer and prepare the samples for the mass spectrometer. The compounds were eluted from the column using an acetonitrile gradient and fed into the mass analyzer. Initial data analysis took place with either Shimadzu LabSolutions or Waters MassLynx. NMR data was collected with either a Broker AVANCE III HD 400MHz or a Bruker Ascend 500MHz instrument and the raw data was analyzed with either TopSpin or Mestreiab Mnova,

Synthesis of Intermediates

Intermediate 1. Synthesis of 3-{5-bromo-1-ethyl-2-[2-[(1S)"1-methoxyethyl]pyr!din-3- yl]irsdol-3-yl)-2,2-dimethyipropars-1-ol

Step 1: Synthesis of 1-(5-bromo-1H-indol-3-yl)-3-((tert-butyldiphenylsllyl)oxy)-2 ,2-dimethylpropan- 1-one

To a mixture of 3-((tert-butyldiphenylsllyl)oxy)-2.2-dimethylpropanoyi chloride (65 g, 137 mmol, crude) in DCM (120 mL) at 0 °C under an atmosphere of N 2 was added 1 M SnCl 4 in DCM (137 mL,

137 mmol) slowly. The mixture was stirred at 0 °C for 30 min, then a solution of 5-hromo-1 fflndole (26.8 g, 137 mmol) in DCM (40 mL) was added dropwise. The mixture was stirred at 0 °C for 45 min, then diluted with EtOAc (300 mL), washed with brine (4 x 100 mL), dried over NaaSCh and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 1-(5-bromo-1H-indol-3-yl)-3-((tert-butyldiphenylsllyi)oxy)-2 ,2-dimethylpropan-1- one (55 g, 75% yield). LCMS (ESI) m/z: [M + Na] calcd for C 29 H 32 BrNO 2 SiNa 556.1 ; found 556.3.

Step 2: Synthesis of 1-(5-bromo-1H-indol-3-yl)-3-((tert-butyldiphenylsllyl)oxy)-2 ,2-dimethylpropan- 1-one

To a mixture of 1-(5-bromo-1H-indol-3-yl)-3-((tert-butyldiphenylsilyi)oxy)-2 ,2-dimethylpropan-1- one (50 g, 93.6 mmol) in THF (100 mL) at 0 °C under an atmosphere of Na was added LiBX (6.1 g,

281 mmol). The mixture was heated to 60 °C and stirred for 20 h, then MeOH (10 mL) and EtOAc (100 mL) were added and the mixture washed with brine (50 ml.), dried over NaaSCU, filtered and the filtrate concentrated under reduced pressure. The residue was diluted with DCM (50 mL), cooled to 10 °C and diludine (9.5 g, 37.4 mrnol) and (890 mg, 4.7 mmol) were added. The mixture was stirred at 10 °C for 2 h, filtered, the filtrate concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 1-(5-bromo-1H-indol-3-yl)-3-((ieri-butyldiphenylsllyi)oxy)-2 ,2- dimethylpropan-1-one (41 g, 84% yield). LCMS (ESI) m/z: [M + H] calcd for C 29 H 34 BrNOSi: 519.2; found 520,1 ; 1 H NMR (400 MHz, GDCl 3 ) δ 7.96 (s, 1H), 7,75 - 7.68 (m, 5H), 7.46 - 7,35 (m, 6H), 7,23 - 7.19 (m, 2H), 6.87 (d, J = 2.1 Hz, 1H), 3.40 (s, 2H), 2.72 (s, 2H), 1.14 (s, 9H), 0.89 (s, 6H).

Step 3: Synthesis of 5-bromo-3-(3-((tert-butyldiphenylsllyi)oxy)-2,2-dimethylprop yl)-2-iodo-1H- indole

To a mixture of 1-(5-bromo-1H-indol-3-yl)-3-((tert-butyldiphenylsllyl)oxy)-2 .2-dimethylpropan-1- one (1 ,5 g, 2.9 mmol) and l 2 (731 mg, 2.9 rnmol) in IMF (15 ml) at room temperature was added AgOTf (888 mg, 3.5 mmol). The mixture was stirred at room temperature for 2 h, then diluted with EtOAc (200 ml.) and washed with saturated Na 2 S 2 O 3 (100 ml.), dried over anhydrous NA 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 5-bromo-3-(3-((tert-butyldiphenylsllyl)oxy)-2,2-dimethylprop yl)-2-iodo-1H-indole (900 mg, 72% yield) as a solid. 1 H NMR (400 MHz, DMSO-d 6 ) d 11 .70 (s, 1H), 7.68 (d, J = 1 .3 Hz, 1H), 7.64 - 7.62 (m, 4H), 7.46 - 7.43 (m, 6H), 7.24 - 7.22 (d, 1H), 7.14 - 7.12 (dd, J = 8.6, 1.6 Hz, 1H), 3.48 (s, 2H), 2.63 (s, 2H), 1 .08 (s, 9H), 0.88 (s, 6H).

Step 4: Synthesis of (1 S)-1-(3-bromopyridin-2-yl)ethanol

To a stirred mixture of HCOOH (66.3 g, 1 .44 mol) in Et 3 N (1002 mL, 7.2 mol) at 0 c C under an atmosphere of Ar was added (4S,5S)-2-chloro-2-methyl-1-(4-methylbenzenesulfonyl)-4,5-di phenyl-1 ,3- diaza-2-ruthenacyclopentane cymene (3.9 g, 6.0 mmol) portion-wise. The mixture was heated to 40 °C and stirred for 15 min, then cooled to room temperature and 1-(3-bromopyridin-2-yl)ethanone (120 g,

600 mmol) added in portions. The mixture was heated to 40 °C and stirred for an additional 2 h, then the solvent was concentrated under reduced pressure. Brine (2 L) was added to the residue, the mixture was extracted with EtOAc (4 x 700 mL), dried over anhydrous NA 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give (1 S)-1-(3-bromopyridin-2-yl)ethanol (100 g, 74% yield) a an oil. LCMS (ESI) m/z: [M + H] calcd for CyHsBrNO: 201 .98; found 201 ,9.

Step 5: Synthesis of 3-bromo-2-[(1 S)-1-methoxyethyl]pyridine

To a stirred mixture of (1 S)-1-(3-bromopyridin-2-yl)ethanoi (100 g, 495 mmol) in DMF (1 L) at 0 ,3 C was added NaH, 60% dispersion in oil (14,25 g, 594 mmol) in portions. The mixture was stirred at 0 °C for 1 h. Mel (140.5 g, 990 mmol) was added dropwise at 0 °G and the mixture was allowed to warm to room temperature and stirred for 2 h. The mixture was cooled to 0 °C and saturated NH 4 CI (5 L) was added. The mixture was extracted with EtOAc (3 x 1 ,5 L), dried over anhydrous NA 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 3-bromo-2-[(1 S)-1-methoxyethyl]pyridine (90 g, 75% yield) as an oil. LCMS (ESI) m/z: [M + H] calcd for C 8 H 10 BrNO: 215.99; found 215.9,

Step 6: Synthesis of 2-[(1 S)-1-methoxyethyl]-3-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yi)pyi1dine

To a stirred mixture of 3-bromo-2-[(1 S)-1-methoxyethyl]pyridine (90 g, 417 mmol) in toluene (900 mL) at room temperature under an atmosphere of Ar was added bis(pinacolato)diboron (127 g,

500 mmol) and KOAc (81 .8 g, 833 mmol) and Pd(dppf)Cl2 (30.5 g, 41 .7 mmol). The mixture was heated to

100 °C and stirred for 3 h. The filtrate was concentrated under reduced pressure and the residue was purified by AI 2 O 3 column chromatography to give 2-[(1 S)-1-methoxyethyl]-3-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)pyridine (100 g, 63% yield) as a semi-solid. LCMS (ESI) m/z: [M + H] calcd for C 14 H 22 BNO 3 ; 264.17; found 264.1 .

Step 7: Synthesis of 5-bromo-3-[3-[(tert-butyldiphenylsilyl)oxy]-2,2-dimethylprop yl]-2-[2-[(1 S)- 1- methoxyethyl]pyridin-3-yl]-1H-indole

To a stirred mixture of 5-bromo-3-[3-[(tert-butyldiphenylsllyl)oxy]-2,2-dimethylprop yl]-2-iodo-1 H- indole (140 g, 217 mmol) and 2-[(1 S)-1-methoxyethyl]-3-(4,4,5,5-tetramethyl-1 ,3,2-dioxaboroian-2- yl)pyridine (100 g, 380 mmol) in 1 ,4-dioxane (1 .4 L) at room temperature under an atmosphere of Ar was added K 2 CO 3 (74.8 g, 541 mmol), Pd(dppf)Cl 2 (15.9 g, 21 .7 mmol) and H 2 O (280 ml.) in portions. The mixture was heated to 85 °C and stirred for 4 h, then cooled, H 2 O (5 L) added and the mixture extracted with EtOAc (3 x 2 L). The combined organic layers were washed with brine (2 x 1 L), dried over anhydrous NA 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 5-bromo-3-[3-[(ierf-butyldiphenylsllyl)oxy]-2,2- dimethylpropyl3-2-[2-[(1 S)-1- methoxyethyl]pyridin- 3-yl ]- 1H-indole (71 g, 45% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for C 37 H 43 BrN 2 O 2 Si: 655.23; found 655.1 .

Step 8: Synthesis of 5-bromo-3-[3-[(tert-butyldiphenylsllyl)oxy]-2,2-dimethylprop yl]-1 -ethyl-2-[2- [(18)-1-methoxyethyl]pyridin-3-yl]indole

To a stirred mixture of 5-bromo-3-[3-[(tert-butyldiphenylsllyl)oxy]-2,2-dimethylprop yl]-2-[2-[(1 S)-1- methoxyethyl]pyridin-3-yl]-1H-indole (71 g, 108 mmol) in DMF (0.8 L) at 0 °C under an atmosphere of M2 was added CS2CO 3 (70.6 g, 217 mmol) and Etl (33.8 g, 217 mmol) in portions. The mixture was warmed to room temperature and stirred for 16 h then H 2 O (4 L) added and the mixture extracted with EtOAc (3 x 1 .5 L). The combined organic layers were washed with brine (2 x 1 L), dried over anhydrous NA 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 5-bromo-3-[3-[(tert-butyldiphenylsllyi)oxy]-2,2-dimethylprop yl]-1-ethyl-2- [2-[(1 S)-1-methoxyethyl]pyridin-3-yl]indole (66 g, 80% yield) as an oll. LCMS (ESI) m/z: [M + H] calcd for C 39 H 47 BrN 2 O 2 Si: 683.26; found 683.3.

Step 9: Synthesis of 3-(5-bromo-1-ethyl-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]indol-3-yl)-2,2- dimethylpropan-1-ol

To a stirred mixture of TBAF (172.6 g, 660 mmol) in THF (660 mL) at room temperature under an atmosphere of N 2 was added 5-bromo-3-[3-[(tert-butyldiphenylsllyl)oxy]-2,2-dimethylprop yl]-1-ethyl-2-[2- [(1 S)-1-methoxyethyl]pyridin-3-yl]indole (66 g, 97 mmol) in portions. The mixture was heated to 50 °C and stirred for 16 h, cooled, diluted with H 2 O (5 L) and extracted with EtOAc (3 x 1 .5 L), The combined organic layers were washed with brine (2 x 1 L), dried over anhydrous NA 2 SO 4 and filtered. After filtration, the filtrate was concentrated under reduced pressure. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 3-(5-bromo-1-ethyl-2- [2-[(1 S)-1-methoxyethyl]pyridin-3-yl]indol-3-yl)-2,2-dimethylpropa n-1-ol (30 g, 62% yield) as a solid.

LCMS (ESI) m/z: [M + H] calcd for C 23 H 29 BrN 2 O 2 : 445.14; found 445.1 . intermediate 1. Alternative Synthesis through Fisher indoie Route.

Step 1: Synthesis of 5-[2-[(1 S)-1 -methoxyethyl]pyridin-3-yl]-2,2-dimethyl-5-oxopentanoic acid

To a mixture of /-PrMgCl (2M in in THF, 0.5 L) at -10 °C under an atmosphere of NK was added n- BuLi, 2.5 M in hexane (333 mL, 833 mmol) dropwise over 15 min. The mixture was stirred for 30 min at - 10 °C then 3-bromo-2-[(1 S)-1-methoxyethyl]pyridine (180 g, 833 mmol) in THF (0.5 L) added dropwise over 30 min at -10 °C. The resulting mixture was warmed to -5 “C and stirred for 1 h, then 3,3- dimethyloxane-2,6-dione (118 g, 833 mmol) in THF (1 .2 L) was added dropwise over 30 min at -5 °C. The mixture was warmed to 0 ,3 C and stirred for 1 .5 h, then quenched with the addition of pre-cooied 4M HCi in 1 ,4-dioxane (0.6 L) at 0 °C to adjust pH ~5, The mixture was diluted with H 2 O (3 L) at 0 c C and extracted with EtOAc (3 x 2.5 L). The combined organic layers were dried over anhydrous NasSCU, filtered, the filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 5-[2-((1 S)-1-methoxyethyl]pyrldln-3-yl]-2,2-dimethyl-5-oxopentanoic acid (87 g, 34% yield) as a solid. LGMS (ESI) m/z: [M + H] calcd for C 15 H 21 NO 4 : 280.15; found 280.1 .

Step 2: Synthesis of 3-(5-bromo-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]-1H-indol-3-yl)-2,2- dimethylpropanoic acid and ethyl (S)-3-(5-bromo-2-(2-(1-methoxyethyl)pyridin-3-yl)-1H-indol-3 -yl)-2,2- dimethylpropanoate

To a mixture of 5-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]-2,2-dimethyl-5-oxopentanoic acid (78 g, 279 mmol) in EtOH (0.78 L) at room temperature under an atmosphere of N 2 was added (4- bromophenyl)hydrazine HCI salt (68.7 g, 307 mmol) in portions. The mixture was heated to 85 °C and stirred for 2 h, cooled to room temperature, then 4M HCI In 1 ,4-dioxane (69.8 mL, 279 mmol) added dropwise. The mixture was heated to 85 °C and stirred for an additional 3 h, then concentrated under reduced pressure and the residue was dissolved in TFA (0.78 L). The mixture was heated to 60 °C and stirred for 1 .5 h, concentrated under reduced pressure and the residue adjusted to pH ~5 with saturated NaHCO 3 , then extracted with EtOAc (3 x 1 .5 L). The combined organic layers were dried over anhydrous NA 2 SO 4 , filtered, the filtrate concentrated under reduced pressure and the residue was purified by silica gei column chromatography to give 3-(5-bromo-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]-1H-indol-3-yl)-2,2- dimethylpropanoic acid and ethyl (S)-3-(5-bromo-2-(2-{1-methoxyethyl)pyridin-3-yl)-1H-indol-3 -yl)-2,2- dimethylpropanoate (78 g, crude), LCMS (ESI) m/z : [M + H] calcd for C 21 H 23 BrN 2 O 3 : 430.1 and C 23 H 27 BrN 2 O 3 : 459.12; found 431 .1 (carboxylic acid) and 459.1 .

Step 3: Synthesis of ethyl 3-(5-bromo-1-ethyl-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]indol-3-yl)-

2.2-dimethylpropanoate

To a mixture of 3-(5-bromo-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]-1H-indol-3-yl)-2,2- dimethylpropanolc acid and ethyl (S)-3-(5-bromo-2-(2-(1-methoxyethyl)pyridin-3-yl)-1H-indol-3 -yl)-2,2- dimethylpropanoate (198 g, 459 mmol) in DMF (1 .8 L) at 0 °C under an atmosphere of N 2 was added Cs 2 CO 3 (449 g, 1 .38 mol) in portions. Etl (215 g, 1 .38 mmol) in DMF (200 ml.) was then added dropwise at 0 °C. The mixture was warmed to room temperature and stirred for 4 h then diluted with brine (5 L) and extracted with EtOAc (3 x 2.5 L). The combined organic layers were washed with brine (2 x 1 .5 L), dried over anhydrous Na 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give ethyl 3-(5-bromo-1-ethyl-2-[2-[(1 S)-1- methoxyethyl]pyridin-3-yl]jindol-3-yl)-2,2-dimethylpropanoat e (160 g, 57% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for C 25 H 31 BrN 2 O 3 : 487.17; found 487.2.

Step 4: Synthesis of 3-(5-bromo-1-ethyl-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1 H-indol-3-yl)-2,2- dimethylpropan-1-ol

To a mixture of ethyl 3-(5-bromo-1-ethyl-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]indol-3-yl)-2,2- dimethylpropanoate (160 g, 328 mmol) in THF (1 .6 L) at 0 °C under an atmosphere of N 2 was added LiBH 4 (28.6 g, 1 .3 mol). The mixture was heated to 60 °C for 16 h, cooled, and quenched with pre-cooled (0 °C) aqueous NH 4 Cl (5 L). The mixture was extracted with EtOAc (3 x 2 L) and the combined organic layers were washed with brine (2 x 1 L), dried over anhydrous Na 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give to two atropisomers of 3-(5-bromo-1-ethyl-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1H -indol-3-yl)-

2.2-dlmethylpropan-1-oi (as single atropisomers) (60 g, 38% yield) and (40 g, 26% yield) both as solids. LCMS (ESI) m/z: [M + H] calcd for C 23 H 29 BrN 2 O 2 : 445.14; found 445.2.

Intermediate 2 and Intermediate 4. Synthesis of (S)-1-((S)-2-((tert-butoxycarbonyl)amlno)-3- (3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-5-

((triisopropylsilyl)oxy)phenyl)propanoyl)hexahydropyridaz ine-3-carboxylate

Step 1: Synthesis of (S)-methyl 2-(tert-butoxycarbonylamino)-3-(3-(triisopropylsilyloxy)phen yl)- propanoate

To a mixture of (S)-methyl 2-(tert-butoxycarbonylamino)-3-(3-hydroxyphenyl)propanoate (10.0 g, 33.9 mmol) in DCM (100 mL) was added imidazole (4.6 g, 67.8 mmol) and TIPSCI (7.8 g, 40.7 mmol).

The mixture was stirred at room temperature overnight then diluted with DCM (200 mL) and washed with H 2 O (3 x 150 mL). The organic layer was dried over anhydrous Na 2 SO 4 . filtered, concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give (S)-methyl 2- (tert-butoxycarbonylamino)-3-(3-(triisopropylsilyloxy)phenyl )-propanoate (15.0 g, 98% yield) as an oil. LCMS (ESI) m/r. [M + Na] calcd for C 24 H 41 NO 5 SiNa : 474.22; found 474.2.

Step 2. Synthesis of (S)-methyl 2-(tert-butoxycarbonylamino)-3-(3-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)-5-(triisopropylsilyloxy)phenyl)-propanoat e

A mixture of (S)-methyl 2-(tert-butoxycarbonylamino)-3-(3-(triisopropylsilyloxy)phen yl)-propanoate (7.5 g, 16.6 mmol), PinB 2 (6.3 g, 24.9 mmol), [lr(OMe)(COD)] 2 (1.1 g, 1.7 mmol) and 4-tert-butyl-2-(4-tert· butyl-2-pyridyl)pyridine (1.3 g, 5.0 mmol) was purged with Ar, then THF (75 mL) was added and the mixture placed under an atmosphere of Ar and sealed. The mixture was heated to 80 °C and stirred for 16 h, concentrated under reduced pressure, and the residue was purified by silica gel column chromatography to give (S)-methyl 2-(tert-butoxycarbonylamino)-3-(3-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)-5-(triisopropylsilyloxy)phenyl)-propanoat e (7.5 g, 78% yield) as a solid. LCMS (ESI) m/z. [M + Na] calcd for C 30 H 52 BN O 7 SiNa: 600.35; found 600.4; 1 H NMR (300 MHz, CD 3 OD) δ 7.18 (s,

1 H), 7.11 (s, 1 H), 6.85 (s, 1 H), 4.34 (m, 1 H), 3.68 (s, 3H), 3.08 (m, 1 H), 2.86 (m, 1 H), 1.41 - 1.20 (m, 26H), 1.20 - 1.01 (m, 22H), 0.98 - 0.79 (m, 4H).

Step 3: Synthesis of (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(4,4,5,5-tetramethyl -1 ,3,2- dioxaborolan-2-yl)-5-((triisopropylsilyl)oxy)phenyl)propanoi c acid

To a mixture of triisopropylsilyl (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(4,4,5,5-tetramethyl -1 ,3,2- dioxaborolan-2-yl)-5-((triisopropylsilyl)oxy)phenyl)propanoa te (4.95 g, 6.9 mmol) in MeOH (53 mL) at 0 °C was added LiOH (840 mg, 34.4 mmol) in H2O (35 mL). The mixture was stirred at 0 °C for 2 h, then acidified to pH ~5 with 1 M HCI and extracted with EtOAc (2 x 250 mL). The combined organic layers were washed with brine (3 x 100 mL), dried over anhydrous Na 2 SO 4 , filtered and the filtrate concentrated under reduced pressure to give (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(4,4,5,5-tetramethyl -1 ,3,2-dioxaborolan- 2-yl)-5-((triisopropylsilyl)oxy)phenyl)propanoic acid (3.7 g, 95% yield), which was used directly in the next step without further purification. LCMS (ESI) m/z. [M + NH*] calcd for C 29 H 50 BNO 7 SiNH 4 : 581.38; found 581.4.

Step 4: Synthesis of methyl (S)-1-((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(4,4,5,5-tetr amethyl- 1 ,3,2-dioxaborolan-2-yl)-5-((triisopropylsilyl)oxy)phenyl)pro panoyl)hexahydropyridazine-3-carboxylate To a mixture of methyl (S)-hexahydropyridazine-3-carboxylate (6.48 g, 45.0 mmol) in DCM (200 mL) at 0 °C was added NMM (41.0 g, 405 mmol), (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(4, 4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)-5-((triisopropylsilyl)oxy)phenyl)pro panoic acid (24 g, 42.6 mmol) in DCM (50 mL) then HOBt (1.21 g, 9.0 mmol) and EDCI HCI salt (12.9 g, 67.6 mmol). The mixture was warmed to room temperature and stirred for 16 h, then diluted with DCM (200 mL) and washed with H 2 O (3 x 150 mL). The organic layer was dried over anhydrous Na 2 SO 4 , filtered, the filtrate concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give methyl (S)-1- ((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(4,4,5,5-tetramethy l-1 ,3,2-dioxaborolan-2-yl)-5- ((triisopropylsilyl)oxy)phenyl)propanoyl)hexahydropyridazine -3-carboxylate (22 g, 71% yield) as an oil. LCMS (ESI) m/z. [M + H] calcd for C 35 H 60 BN 3 O 8 Si : 690.42; found 690.5.

Intermediate 3. Synthesis of (S)tert-butyl 3-methyl-2-((S)-N-methylpyrrolidine-3- carboxamido)butanoate

Step 1: Synthesis of (S)-tert-butyl 3-(((S)-1 -(tert-butoxy)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamoyl)pyrrolidine-1-carboxylate

To a mixture of (S)-1-(tert-butoxycarbonyl)pyrrolidine-3-carboxylic acid (2.2 g, 10.2 mmol) in DMF (10 mL) at room temperature was added HATU (7.8 g, 20.4 mmol) and DIPEA (5 mL). After stirring at room temperature for 10 min, tert-butyl methyl-L-valinate (3.8g, 20.4 mmol) in DMF (10 mL) was added. The mixture was stirred at room temperature for 3 h, then diluted with DCM (40 mL) and H 2 O (30 mL). The aqueous and organic layers were separated, and the organic layer was washed with H 2 O (3 x 30 mL), brine (30 mL), dried over anhydrous Na 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give (S)-tert-butyl 3-(((S)-1 -(tert-butoxy)-3-methyl-1 -oxobutan-2-yl)(methyl)carbamoyl)pyrrolidine-1 -carboxylate (3.2 g, 82% yield) as an oil. LCMS (ESI) m/z. [M + Na] calcd for C 20 H 36 N 2 O 5 Na : 407.25; found 407.2.

Step 2. Synthesis of (S)-tert- butyl 3-methyl-2-((S)-N-methylpyrrolidine-3-carboxamido)butanoate

A mixture of (S)-tert- butyl 3-(((S)-1 -(tert-butoxy)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamoyl)pyrrolidine-1 -carboxylate (3.2 g, 8.4 mmol) in DCM (13 mL) and TFA (1.05 g,

9.2 mmol) was stirred at room temperature for 5 h. The mixture was concentrated under reduced pressure to give (S)-tert- butyl 3-methyl-2-((S)-N-methylpyrrolidine-3-carboxamido)butanoate (2.0 g, 84% yield) as an oil. LCMS (ESI) m/z. [M + H] calcd for C 15 H 28 N 2 O 3 : 285.21 ; found 285.2. Intermediate 5. Synthesis of ferf- butyl ((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin- 3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triίsopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa- 1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate.

Step 1: Synthesis of methyl (3S)-1 -[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[1 -ethyl-3-(3- hydroxy-2,2-dimethylpropyl)-2-[2-[(1S)-1-methoxyethyl]pyridi n-3-yl]indol-5-yl]-5- [(triisopropylsilyl)oxy]phenyl]propanoyl]-1,2-diazinane-3-ca rboxylate

To a stirred mixture of 3-(5-bromo-1 -ethyl-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]indol-3-yl)-2,2- dimethylpropan-1-ol (30 g, 67 mmol) and methyl (3S)-1-[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)-5-[(triisopropylsilyl)o xy]phenyl]propanoyl]-1,2-diazinane-3- carboxylate (55.8 g, 80.8 mmol) in 1 ,4-dioxane (750 mL) at room temperature under an atmosphere of Ar was added Na 2 CO 3 (17.9 g, 168.4 mmol), Pd(DtBPF)Cl 2 (4.39 g, 6.7 mmol), and H 2 O (150 mL) in portions. The mixture was heated to 85 °C and stirred for 3 h, cooled, diluted with H 2 O (2 L), and extracted with EtOAc (3 x 1 L). The combined organic layers were washed with brine (2 x 500 mL), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give methyl (3S)-1-[(2S)-2-[(tert- butoxycarbonyl)amino]-3-[3-[1-ethyl-3-(3-hydroxy-2,2-dimethy lpropyl)-2-[2-[(1S)-1-methoxyethyl]pyridin-3- yl]indol-5-yl]-5-[(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3-carboxylate (50 g, 72% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for C 52 H 77 N 5 O 8 Si: 928.56; found 928.8.

Step 2. Synthesis of (3S)-1-[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[1-ethyl-3- (3-hydroxy-2,2- dimethylpropyl)-2-[2-[(1S)-1-methoxyethyl]pyridin-3-yl]indol -5-yl]-5- [(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3-carboxylic acid

To a stirred mixture of methyl (3S)-1-[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[1-ethyl-3- (3- hydroxy-2,2-dimethylpropyl)-2-[2-[(1S)-1-methoxyethyl]pyridi n-3-yl]indol-5-yl]-5- [(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3-carboxylate (50 g, 54 mmol) in DCE (500 mL) at room temperature was added trimethyltin hydroxide (48.7 g, 269 mmol) in portions. The mixture was heated to 65 °C and stirred for 16 h, then filtered and the filter cake washed with DCM (3 x 150 mL). The filtrate was concentrated under reduced pressure to give (3S)-1-[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[3- [1 -ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-[2-[(1 S)-1 -methoxyethyl]pyridin-3-yl]indol-5-yl]-5- [(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3-carboxylic acid (70 g, crude), which was used directly in the next step without further purification. LCMS (ESI) m/z: [M + H] calcd for CsiHysNsOeSi: 914.55; found 914.6.

Step 3: Synthesis of tert-butyl ((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate

To a stirred mixture of (3S)-1 -[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[1 -ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]indol-5-yl]-5-

[(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3-carboxylic acid (70 g) in DCM (5 L) at 0 °C under an atmosphere of N 2 was added DIPEA (400 mL, 2.3 mol), HOBT (51.7 g, 383 mmol) and EDCI (411 g, 2.1 mol) in portions. The mixture was warmed to room temperature and stirred for 16 h, then diluted with DCM (1 L), washed with brine (3 x 1 L), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give tert-butyl ((6 3 S,4S)-1 '-ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5 ,7-dioxo-2 5 - ((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 8 -hexahydro-1 1 H-8-oxa-l (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate (36 g, 42% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for C 51 H 73 N 5 O 7 Si : 896.54; found 896.5.

Intermediate 6. Synthesis of tert- butyl ((6 3 S,4S)-1 2 -lodo-10.10-dlmethyl-5,7-dloxo-2 5 - ((trlisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 -H-8-oxa-1 (5,3)-lndola-6(1 ,3)-pyridazlna-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate.

Step 1: Synthesis of 3-(5-bromo-1 H-indol-3-yl)-2,2-dimethylpropan-1-ol This reaction was undertaken on five batches in parallel on the scale illustrated below. Into a 2L round-bottom flask were added 5-bromo-3-[3-[(tert-butyldiphenylsilyl)oxy]-2,2-dimethylprop yl]-1 H- indole (100 g, 192 mmol) and TBAF (301 .4 g, 1.15 mol) in THF (1.15 L) at room temperature. The resulting mixture was heated to 50 °C and stirred for 16 h, then the mixture was concentrated under reduced pressure.

At this stage the residues from all five batches were combined, diluted with H 2 O (5 L), and extracted with EtOAc (3 x 2 L). The combined organic layers were washed with brine (2 x 1.5 L), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 3-(5-bromo-1 H-indol-3-yl)-2,2- dimethylpropan-1 -ol (310 g, crude) as a solid. LCMS (ESI) m/z [M + H] calcd for C 13 H 16 BrNO: 282.05 and 284.05; found 282.1 and 284.1.

Step 2. Synthesis of 3-(5-bromo-1 H- indol-3-yl)-2,2-dimethylpropyl acetate This reaction was undertaken on two batches in parallel in accordance with the procedure below. To a stirred mixture of 3-(5-bromo-1 H- indol-3-yl)-2,2-dimethylpropan-1-ol (135 g, 478 mmol) and EtaN (200 mL, 1 .44 mol) in DCM (1.3 L) at 0 °C under an atmosphere of N 2 was added Ac 2 O (73.3 g, 718 mmol) and DMAP (4.68 g, 38.3 mmol) in portions. The resulting mixture was stirred for 10 min at 0 °C, then washed with H 2 O (3 x 2 L).

At this stage, the organic layers from both batches were combined and washed with brine (2 x 1 L), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by column chromatography to give 3-(5-bromo-1 H- indol-3-yl)-2,2-dimethylpropyl acetate (304 g, 88% yield) as a solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ 11.16 - 11.11 (m, 1H), 7.69 (d, J= 2.0 Hz, 1H), 7.32 (d, J= 8.6 Hz, 1H), 7.19 - 7.12 (m, 2H), 3.69 (s, 2H), 2.64 (s, 2H), 2.09 (s, 3H), 0.90 (s, 6H).

Step 3: Synthesis of methyl (2S)-3-(3-[3-[3-(acetyloxy)-2,2-dimethylpropyl]-1 H- indol-5-yl]-5- [(triisopropylsilyl)oxy]phenyl)-2-[(tert-butoxycarbonyl)amin o]propanoate

This reaction was undertaken on four batches in parallel in accordance with the procedure below. Into a 2L round-bottom flasks were added methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-(4, 4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)-5-[(triisopropylsilyl)oxy]phenyl]pro panoate (125 g, 216 mmol), 1 ,4- dioxane (1 L), H 2 O (200 mL), 3-(5-bromo-1 H- indol-3-yl)-2,2-dimethylpropyl acetate (73.7 g, 227 mmol), K2CO3 (59.8 g, 433 mmol), and Pd(DtBPF)Cl 2 (7.05 g, 10.8 mmol) at room temperature under an atmosphere of Ar. The resulting mixture was heated to 65 °C and stirred for 2 h, then diluted with H 2 O (10 L) and extracted with EtOAc (3 x 3 L). The combined organic layers were washed with brine (2 x 2 L), dried over anhydrous Na 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure.

At this point the residue from all four batches was combined and purified by column chromatography to give methyl (2S)-3-(3-[3-[3-(acetyloxy)-2,2-dimethylpropyl]-1 H-indol-5-yl]-5- [(triisopropylsilyl)oxy]phenyl)-2-[(tert-butoxycarbonyl)amin o]propanoate (500 g, 74% yield) as an oil. LCMS (ESI) m/z [M + Na] calcd for C 39 H 58 N 2 SiNa: 717.39; found 717.3.

Step 4: Synthesis of methyl (2S)-3-(3-[3-[3-(acetyloxy)-2,2-dimethylpropyl]-2-iodo-1 H- indol-5-yl]- 5-[(triisopropylsilyl)oxy]phenyl)-2-[(tert-butoxycarbonyl)am ino]propanoate

This reaction was undertaken on three batches in parallel in accordance with the procedure below. To a stirred mixture of methyl (2S)-3-(3-[3-[3-(acetyloxy)-2,2-dimethylpropyl]-1 H- indol-5-yl]-5- [(triisopropylsilyl)oxy]phenyl)-2-[(tert-butoxycarbonyl)amin o]propanoate (150 g, 216 mmol) and NaHCO 3 (21.76 g, 259 mmol) in THF (1.5 L) was added AgOTf (66.5 g, 259 mmol) in THF dropwise at 0 °C under an atmosphere of nitrogen. I2 (49.3 g, 194 mmol) in THF was added dropwise over 1 h at 0 °C and the resulting mixture was stirred for an additional 10 min at 0 °C. The combined experiments were diluted with aqueous Na 2 S 2 O 3 (5 L) and extracted with EtOAc (3 x 3 L). The combined organic layers were washed with brine (2 x 1.5 L), dried over anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure to a residue.

At this stage, the residue from all three batches was combined and purified by column chromatography to give methyl (2S)-3-(3-[3-[3-(acetyloxy)-2,2-dimethylpropyl]-2-iodo-1 H- indol-5-yl]-5- [(triisopropylsilyl)oxy]phenyl)-2-[(te/i-butoxycarbonyl)amin o]propanoate (420 g, 71% yield) as an oil.

LCMS (ESI) m/z. [M + Na] calcd for C 39 H 57 lN 2 O 7 SiNa: 843.29; found 842.9.

Step 5: Synthesis of methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[3-(3-hydroxy-2,2- dimethylpropyl)-2-iodo-1 H- indol-5-yl]-5-[(triisopropylsilyl)oxy]phenyl]propanoate

This reaction was undertaken on three batches in parallel in accordance with the procedure below. To a 2L round-bottom flask were added methyl (2S)-3-(3-[3-[3-(acetyloxy)-2,2-dimethylpropyl]-2- iodo-1 H-indol-5-yl]-5-((triisopropylsilyl)oxy]phenyl)-2-[(tert-but oxycarbonyl)amino]propanoate (140 g,

171 mmol), MeOH (1.4 L), and K3PO4 (108.6 g, 512 mmol) at 0 °C. The mixture was warmed to room temperature and stirred for 1 h, then the combined experiments were diluted with H 2 O (9 L) and extracted with EtOAc (3 x 3 L). The combined organic layers were washed with brine (2 x 2 L), dried over anhydrous Na 2 SO 4 , filtered, and the filtrate was concentrated under reduced pressure.

At this stage the residue from all three batches was combined to give methyl (2S)-2-[(tert- butoxycarbonyl)amino]-3-[3-[3-(3-hydroxy-2,2-dimethylpropyl) -2-iodo-1 H- indol-5-yl]-5- [(triisopropylsilyl)oxy]phenyl]propanoate (438 g, crude) as a solid. LCMS (ESI) m/z. [M + Na] calcd for C 37 H 55 lN 2 O 6 SiNa: 801.28; found 801.6.

Step 6: Synthesis of (2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[3-(3-hydroxy-2,2-d imethylpropyl)-2- iodo-1 H- indol-5-yl]-5-[(triisopropylsilyl)oxy]phenyl]propanoic acid

This reaction was undertaken on three batches in parallel in accordance with the procedure below. To a stirred mixture of methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[3-(3-hydroxy-2,2- dimethylpropyl)-2-iodo-1 H- indol-5-yl]-5-[(triisopropylsilyl)oxy]phenyl]propanoate (146 g, 188 mmol) in THF (1.46 L) was added LiOH (22.45 g, 937 mmol) in H 2 O (937 mL) dropwise at 0 °C. The resulting mixture was warmed to room temperature and stirred for 1.5 h [note: LCMS showed 15% de-TIPS product]. The mixture was acidified to pH 5 with 1 M HCI (1 M) and the combined experiments were extracted with EtOAc (3 x 3 L). The combined organic layers were washed with brine (2 x 2 L), dried over anhydrous Na 2 SO 4 , filtered, and the filtrate was concentrated under reduced pressure.

At this stage the residue from all three batches was combined to give (2S)-2-[(tert- butoxycarbonyl)amino]-3-[3-[3-(3-hydroxy-2,2-dimethylpropyl) -2-iodo-1 H- indol-5-yl]-5- [(triisopropylsilyl)oxy]phenyl]propanoic acid (402 g, crude) as a solid. LCMS (ESI) m/z. [M + Na] calcd for C36H53lN 2 0eSiNa: 787.26; found 787.6.

Step 7: Synthesis of methyl (3S)-1-[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[3-(3-hydro xy-2,2- dimethylpropyl)-2-iodo-1 H-indol-5-yl]-5-[(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3- carboxylate

To a stirred mixture of (2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[3-(3-hydroxy-2,2-d imethylpropyl)- 2-iodo-1 H- indol-5-yl]-5-[(triisopropylsilyl)oxy]phenyl]propanoic acid (340 g, 445 mmol) and methyl (3S)-

1.2-diazinane-3-carboxylate (96.1 g, 667 mmol) in DCM (3.5 L) was added NMM (225 g, 2.2 mol), EDCI (170 g, 889 mmol), and HOBt (12.0 g, 88.9 mmol) portionwise at 0 °C. The mixture was warmed to room temperature and stirred for 16 h, then washed with H 2 O (3 x 2.5 L), brine (2 x 1 L), dried over anhydrous N 32804, and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by column chromatography to give methyl (3S)-1-[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[3-(3-hydro xy-

2.2-dimethylpropyl)-2-iodo-1 H- indol-5-yl]-5-[(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3- carboxylate (310 g, 62% yield) as an oil. LCMS (ESI) m/z [M + H] calcd for C42H63IN4O7S1: 891.36; found 890.8. Step 8: Synthesis of (3S)-1 -[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[3-(3-hydroxy-2,2 - dimethylpropyl)-2-iodo-1 H- indol-5-yl]-5-[(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3-carboxylic acid

This reaction was undertaken on three batches in parallel in accordance with the procedure below. To a stirred mixture of methyl (3S)-1 -[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[3-(3-hydroxy-2,2 - dimethylpropyl)-2-iodo-1 H-indol-5-yl]-5-[(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3- carboxylate (85.0 g, 95.4 mmol) in THF (850 mL) was added LiOH (6.85 g, 286 mmol) in H 2 O (410 mL) dropwise at 0 °C under an atmosphere of N 2 . The mixture was stirred at 0 °C for 1.5 h [note: LCMS showed 15% de-TIPS product], then acidified to pH 5 with 1 M HCI

At this stage the mixtures from all three batches was combined and extracted with EtOAc (3 x 2 L). The combined organic layers were washed with brine (2 x 1.5 L), dried over anhydrous Na 2 SO 4 , filtered, and the filtrate was concentrated under reduced pressure to give (3S)-1-[(2S)-2-[(tert- butoxycarbonyl)amino]-3-[3-[3-(3-hydroxy-2,2-dimethylpropyl) -2-iodo-1 H- indol-5-yl]-5- [(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3-carboxylic acid (240 g, crude) as a solid. LCMS (ESI) m/z\ [M + H] calcd for C 41 H 61 IN 4 O 7 Si: 877.35; found 877.6.

Step 9: Synthesis of tert-butyl ((6 3 S,4S)-1 2 -iodo-10,10-dimethyl-5,7-dioxo-2 5 - ((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 ' -H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate

This reaction was undertaken on two batches in parallel in accordance with the procedure below. To a stirred mixture of (3S)-1 -[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[3-(3-hydroxy-2,2 -dimethylpropyl)- 2-iodo-1 H-indol-5-yl]-5-[(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3-carboxylic acid (120 g,

137 mmol) in DCM (6 L) was added DIPEA (357 mL, 2.05 mol), EDCI (394 g, 2.05 mol), and HOST (37 g, 274 mmol) in portions at 0 °C under an atmosphere of N 2 . The mixture was warmed to room temperature and stirred overnight.

At this stage the solutions from both batches were combined and washed with H 2 O (3 x 6 L), brine (2 x 6 L), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by column chromatography to give tert-butyl ((6 3 S,4S)-1 2 -iodo- 10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 -H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (140 g, 50% yield) as a solid. LCMS (ESI) m/z. [M + H] calcd for C 41 H 59 IN 4 O 6 Si: 859.33; found 858.3.

Intermediate 7. Synthesis of (6 3 S, 4S)-4-amlno-1 1 -ethyl-2 5 - hydroxy-1 2 -(4- (methoxymethyl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-5,7-dione

Step 1: Synthesis of 4-(methoxymethyl)-3-(4,4,5,5-tetramethyl-1 ,3 ,2-d ioxaborolan-2- yl)pyridine To a mixture of 3-bromo-4-(methoxymethyl)pyridine (1.0 g, 5.0 mmol), 4,4,5,5-tetramethyl-2- (tetramethyl-1 ,3,2-dioxaborolan-2-yl)-1 ,3,2-dioxaborolane (1.51 g, 5.9 mmol) and KOAc (1.21 g, 12.3 mmol) in toluene (10 mL) at room temperature under an atmosphere of Ar was added Pd(dppf)Cl 2 (362 mg, 0.5 mmol). The mixture was heated to 110 °C and stirred overnight, then concentrated under reduced pressure to give 4-(methoxymethyl)-3-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)pyridine, which was used directly in the next step directly without further purification. LCMS (ESI) m/z. [M + H] calcd for C13H20BNO3: 250.16; found 250.3.

Step 2. Synthesis of give tert-butyl ((6 3 S,4S)-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl- 5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 '-H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate

To a mixture of 4-(methoxymethyl)-3-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridine (290 mg, 1.16 mmol), K3PO4 (371 mg, 1.75 mmol) and tert-butyl ((6 3 S,4S)-1 2 -iodo-10,10-dimethyl-5,7- dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 -H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (500 mg, 0.58 mmol) in 1 ,4-dioxane (5 mL) and H 2 O (1 mL) at room temperature under an atmosphere of Ar was added Pd(dppf)Cl 2 (43 mg, 0.06 mmol). The mixture was heated to 70 °C and stirred for 2 h, then H 2 O was added and the mixture extracted with EtOAc (2 x 10 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give tert-butyl ((6 3 S,4S)-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10- dimethyl-5, T-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 - H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (370 mg, 74% yield) as a foam. LCMS (ESI) m/z. [M + H] calcd for C 48 H 67 N 5 O 7 Si: 854.49; found 854.6.

Step 3: Synthesis of tert-bu tyl ((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 - H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate

A mixture of tert-butyl ((6 3 S,4S)-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-di oxo-2 5 - ((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 -H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate (350 mg, 0.41 mmol), Cs 2 CO 3 (267 mg, 0.82 mmol), and Etl (128 mg, 0.82 mmol) in DMF (4 mL) was stirred at 35 °C overnight. H 2 O was added and the mixture was extracted with EtOAc (2 x 15 mL). The combined organic layers were washed with brine (15 mL), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give tert-butyl ((6 3 S,4S)-1 1 -ethyl-1 2 -(4- (methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 '-H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (350 mg, 97% yield) as an oil. LCMS (ESI) m/z. [M + H] calcd for C 50 H 71 N 5 O 7 5Si: 882.52; found 882.6.

Step 4: Synthesis of tert-butyl ((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)-

10.10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate

A mixture of tert-butyl ((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7- dioxo-2 5 -((triisopropylsilyl)oxy)-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 -/+8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (350 mg, 0.4 mmol) and 1 M TBAF in THF (0.48 mL, 0.480 mmol) in THF (3 mL) at 0 °C under an atmosphere of Ar was stirred for 1 h. The mixture was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give tert-butyl ((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7- dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate (230 mg, 80% yield) as an oil. LCMS (ESI) m/z. [M + H] calcd for C41H51N5O7: 726.39; found 726.6.

Step 5: Synthesis of (6 3 S,4S)-4-amino-1 1 -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)-

10.10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione

To a mixture of tert-butyl N-[(8S,14S)-22-ethyl-4-hydroxy-21-[4- (methoxymethyl)pyridin-3-yl]- 18,18-dimethyl-9,15-dioxo-16-oxa-10,22,28-triazapentacyclo[1 8.5.2.1 A [2,6].1 A [10,14].0 A [23,27]]nonacosa- 1 (26),2,4,6(29),20,23(27),24-heptaen-8-yl]carbamate (200 mg, 0.28 mmol) in 1 ,4-dioxane (2 mL) at 0 °C under an atmosphere of Ar was added 4M HCI in 1 ,4-dioxane (2 mL, 8 mmol). The mixture was allowed to warm to room temperature and was stirred overnight, then concentrated under reduced pressure to give (6 3 S,4S)-4-amino-1 1 -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 e -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane- 5,7-dione (200 mg). LCMS (ESI) m/z. [M + H] calcd for C36H43N5O5: 626.34; found 626.5. Intermediate 8. Synthesis of (6 3 ¾4S)-4-amino-1 '-ethyH 2 -(4-(methoxymethyl)pyrldin-3-yl)- 10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 /fS-oxa-1 (5,3>-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione

Step 1: Synthesis of methyl (S)-3-(3-bromophenyl)-2-((fe/t-butoxycarbonyl)amino)propanoa te

To a solution of (2S)-3-(3-bromophenyl)-2-[(tert-butoxycarbonyl)amino]propano ic acid (100 g, 290 mmol) in DMF (1 L) at room temperature was added NaHCOa (48.8 g, 581.1 mmol) and Mel (61.9 g, 435.8 mmol). The reaction mixture was stirred for 16 h and was then quenched with H 2 O (1 L) and extracted with EtOAc (3 x 1 L). The combined organic layers were washed with brine (3 x 500 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (13% EtO Ac/pet. ether) to afford the desired product (109 g, crude). LCMS (ESI) m/z: [M + Na] calcd for CisHaoBrNO*: 380.05; found 380.0.

Step 2. Synthesis of methyl (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(4,4,5,5-tetramethyl -1 ,3,2- dioxaborolan-2-yl)phenyl)propanoate

To a stirred solution of methyl (2S)-3-(3-bromophenyl)-2-[(tert-butoxycarbonyl)amino]propano ate (108 g, 301.5 mmol) and bis(pinacolato)diboron (99.53 g, 391.93 mmol) in 1 ,4-dioxane (3.2 L) was added KOAc (73.97 g, 753.70 mmol) and Pd(dppf)Cl 2 (22.06 g, 30.15 mmol). The reaction mixture was heated to 90 °C for 3 h and was then cooled to room temperature and extracted with EtOAc (2 x 3 L). The combined organic layers were washed with brine (3 x 800 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (5% EtOAc/pet. ether) to afford the desired product (96 g, 78.6% yield). LCMS (ESI) m/z. [M + Na] calcd for C21H32BNO6: 428.22; found 428.1.

Step 3: Synthesis of methyl (S)-3-(3-(3-(3-acetoxy-2,2-dimethylpropyl)-1 H-indol-5-yl)phenyl)-2- ((tert-butoxycarbonyl)amino)propanoate

To a mixture of methyl (2S)-2-[(fe/t-butoxycarbonyl)amino]-3-[3-(4,4,5,5-tetramethy l-1 ,3,2- dioxaborolan-2-yl)phenyl]propanoate (94 g, 231.9 mmol) and 3-(5-bromo-1 H-indol-3-yl)-2,2- dimethylpropyl acetate (75.19 g, 231.93 mmol) in 1 ,4-dioxane (1.5 L) and H 2 O (300 mL) was added «2003(64.11 g, 463.85 mmol) and Pd(DtBPF)Cl 2 (15.12 g, 23.19 mmol). The reaction mixture was heated to 70 °C and stirred for 4 h. The reaction mixture was extracted with EtOAc (2 x 2 L) and the combined organic layers were washed with brine (3 x 600 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (20% EtOAc/pet. ether) to afford the desired product (130 g, crude). LCMS (ESI) m/z. [M + H] calcd for θ3οΗ3βΝ2θβ: 523.28; found 523.1.

Step 4: Synthesis of methyl (S)-3-(3-(3-(3-acetoxy-2,2-dimethylpropyl)-2-iodo-1 H-indol-5- yl)phenyl)-2-((tert-butoxycarbonyl)amino)propanoate

To a solution of methyl (2S)-3-(3-[3-[3-(acetyloxy)-2,2-dimethylpropyl]-1 H-indol-5-yl]phenyl)-2- [(tert-butoxycarbonyl)amino]propanoate (95.0 g, 181.8 mmol) and iodine (36.91 g, 145.41 mmol) in THF (1 L) at -10 °C was added AgOTf (70.0 g, 272.7 mmol) and NaHCOs (22.9 g, 272.65 mmol). The reaction mixture was stirred for 30 min and was then quenched by the addition of sat. NazSaOs (100 mL) at 0 °C. The resulting mixture was extracted with EtOAc (3 x 1 L) and the combined organic layers were washed with brine (3 x 500 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (50% EtOAc/pet. ether) to afford the desired product (49.3 g, 41 .8% yield). LCMS (ESI) m/z. [M + H] calcd for C30H37IN 2 O6: 649.18; found 649.1.

Step 5: Synthesis of (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(3-(3-hydroxy-2,2-di methylpropyl)-2- iodo-1 H-indol-5-yl)phenyl)propanoic acid

To a solution of methyl (2S)-3-(3-[3-[3-(acetyloxy)-2,2-dimethylpropyl]-2-iodo-1 H-indol-5- yl]phenyl)-2-[(te/1-butoxycarbonyl)amino]propanoate (60 g, 92.5 mmol) in THF (600 mL) was added a solution of LiOHeHzO (19.41 g, 462.5 mmol) in H 2 O (460 mL). The resulting solution was stirred overnight and then the pH was adjusted to 6 with HCI (1 M). The resulting solution was extracted with EtOAc (2 x 500 mL) and the combined organic layers was washed with sat. brine (2 x 500 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (45 g, 82.1% yield).

LCMS (ESI) m/z. (M + Na] calcd for C27H33IN 2 O5: 615.13; found 615.1.

Step 6: Synthesis of methyl (S)-1 -((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(3-(3-hydroxy-2,2- dimethylpropyl)-2-iodo-1H-indol-5-yl)phenyl)propanoyl)hexahy dropyridazine-3-carboxylate

To a solution of (2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[3-(3-hydroxy-2,2-d imethylpropyl)-2- iodo-1 H-indol-5-yl]phenyl]propanoic acid (30 g, 50.6 mmol) and methyl (3S)-1 ,2-diazinane-3-carboxylate (10.9 g, 75.9 mmol) in DCM (400 mL) was added NMM (40.97 g, 405.08 mmol), HOBT (2.05 g, 15.19 mmol), and EDCI (19.41 g, 101.27 mmol). The reaction mixture was stirred overnight and then the mixture was washed with sat. NH4CI (2 x 200 mL) and sat. brine (2 x 200 mL), and the mixture was dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (14 g, 38.5% yield). LCMS (ESI) m/z. [M + H] calcd for Cs3H43lN40e: 718.23; found 719.4. Step 7: Synthesis of (S)-1-((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(3-(3-hydroxy -2,2- dimethylpropyl)-2-iodo-1H-indol-5-yl)phenyl)propanoyl)hexahy dropyridazine-3-carboxylic acid

To a solution of methyl (S)-1-((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(3-(3-hydroxy -2,2- dimethylpropyl)-2-iodo-1 H-indol-5-yl)phenyl)propanoyl)hexahydropyridazine-3-carboxyl ate (92 g, 128.0 mmol) in THF (920 mL) at 0 °C was added a solution of LiOHel-teO (26.86 g, 640.10 mmol) in H 2 O (640 mL). The reaction mixture was stirred for 2 h and was then concentrated under reduced pressure to afford the desired product (90 g, crude). LCMS (ESI) m/z: [M + H] calcd for C32H41IN4O6: 705.22; found 705.1).

Step 8: Synthesis of tert-butyl ((6 3 S,4S)-1 2 -iodo-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate To a solution of of (3S)-1-[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[3-(3-hydro xy-2,2- dimethylpropyl)-2-iodo-1H-indol-5-yl]phenyl]propanoyl]-1 ,2-diazinane-3-carboxylic acid (90 g, 127.73 mmol) in DCM (10 L) at 0 °C was added HOBt (34.52 g, 255.46 mmol), DIPEA (330.17 g, 2554.62 mmol) and EDCI (367.29 g, 1915.96 mmol). The reaction mixture was stirred for 16 h and was then concentrated under reduced pressure. The mixture was extracted with DCM (2 x 2 L) and the combined organic layers were washed with brine (3 x 1 L), dried over NazSCX filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (50% EtOAc/pet. ether) to afford the desired product (70 g, 79.8% yield). LCMS (ESI) m/z: [M + H] calcd for C32H39IN4O5: 687.21 ; found 687.1.

Step 9: Synthesis of tert-butyl ((6 3 S,4S)-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7- dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H-8-oxa-l (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate

To a solution of tert-butyl ((6 3 S,4S)-1 2 -iodo-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro- 1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (22.0 g,

32.0 mmol) in toluene (300.0 mL) was added Pdz(dba)3 (3.52 g, 3.85 mmol), S-Phos (3.95 g, 9.61 mmol), and KOAc (9.43 g, 96.13 mmol) followed by 4,4,5,5-tetramethyM ,3,2-dioxaborolane (26.66 g, 208.3 mmol), dropwise. The resulting solution was heated to 60 °C and stirred for 3 h. The reaction mixture was then cooled to room temperature, filtered, the filter cake was washed with EtOAc, and the filtrate was concentrated under reduced pressure. The residue was purified by normal phase chromatography to afford the desired product (22 g, 90% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for C38H51BN4O7: 687.39; found 687.3.

Step 10·. Synthesis of tert-butyl ((6 3 S,4S)-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7- dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 8 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate

To a mixture of tert-butyl ((6 3 S,4S)-10,10-dimethyl-5,7-dioxo-12-(4,4,5,5-tetramethyl -1 ,3,2- dioxaborolan-2-yl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate (3.0 g, 4.37 mmol) and 3-bromo-4-(methoxymethyl)pyridine (1.766 g, 8.74 mmol) in dioxane/HzO (5/1 ) at 60 °C was added K2CO3 (2.415 g, 17.48 mmol) and Pd(DTBPF)Cl 2 (0.5695 g, 0.874 mmol). The reaction mixture was stirred for 4 h. The reaction mixture was cooled to room temperature and was extracted with EtOAc (300 mL). The solution was washed with brine (3 x 100 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by normal phase chromatography (50% EtOAc/pet. ether) to afford the desired product (1.96 g, 65.8% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for C39H47N5O6: 682.36; found 682.7.

Step 11: Synthesis of tert-butyl ((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate

To a solution of tert-butyl ((6 3 S,4S)-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-di oxo- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane- 4-yl)carbamate (1.96 g, 2.88 mmol) and ethyl iodide (0.347 mL, 4.31 mmol) in DMF (20.0 mL) was added CS2CO3 (2.342 g, 7.19 mmol). The resulting mixture was stirred at room temperature for 5 h and then diluted with EtOAc (200 mL). The mixture was washed with H 2 O (3 x 100 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by normal phase chromatography (50% EtOAc/pet. ether) to afford the desired product (1.24 g, 61% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for CAIHSINSOB: 710.39; found 710.7.

Step 12. Synthesis of (6 3 S,4S)-4-amino-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10- dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione

To a solution of tert-butyl ((6 3 S,4S)-1 , -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl- 5,7-dioxo-6\6 2 , 6 3 , 6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate (1.09 g, 1.54 mmol) in DCM (1.5 mL) at 0 °C was added TFA (1.50 mL). The reaction mixture was stirred for 1 h, concentrated under reduced pressure, and then azeotroped with toluene (3 x 20 mL) to afford the desired crude product (1.09 g) as a solid. LCMS (ESI) m/z: [M + H] calcd for C36H43N5O4: 610.34; found 610.4.

Intermediate 9. Synthesis of fert-butyl ((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl) pyrkJin- 3-yl)-10,10-dimethyl-5,7-dtoxo-6 1 ,8 2 ,6 3 ,6 4 ,6 6 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-

2(1 ,3)-benzenacycloundecaphane-4-yl) carbamate

Step 1: Synthesis of tert-butyl ((6 3 S,4S)-1 2 -(2-((S)-1 -methoxyethyl) pyridin-3-yl)-10,10-dimethyl- 5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 8 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl) carbamate

To a solution of tert-butyl ((6 3 S,4S)-1 2 -iodo-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro- 1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (13 g, 18.93 mmol) and 2-[(1 S)-1-methoxyethyl]-3-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridine (14.95 g, 56.8 mmol) in dioxane (130 mL) and H 2 O (26 mL) was added K2CO3 (5.23 g, 37.9 mmol) and Pd(dppf)Cl 2 (1.39 g, 1.89 mmol). The reaction mixture was stirred for 4 h at 70 °C. The mixture was cooled to room temperature, filtered, and washed with EtOAc (3 x 100 mL). The filtrate was washed with brine (2 x 100 mL), dried over Na 2 SO 4 . filtered, and concentrated under reduced pressure. The residue was purified by prep-TLC (10% MeOH/DCM) to afford the desired product (21 g, 85.3% yield). LCMS (ESI) m/z: [M + H] calcd for CwhUaNsOe: 696.38; found 696.4.

Step 2: Synthesis of tert-butyl ((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl) pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl) carbamate

To a solution of tert-butyl ((6 3 S,4S)-1 2 -(2-((S)-1 -methoxyethyl) pyridin-3-yl)-10,10-dimethyl-5,7- dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl) carbamate (20 g, 28.7 mmol) and CS2CO3 (18.7 g, 57.5 mmol) in DMF (150 mL) at 0 °C was added a solution of ethyl iodide (13.45 g, 86.22 mmol) in DMF (50 mL). The resulting mixture was stirred overnight at 35 °C and was then diluted with H 2 O (500 mL). The mixture was extracted with EtOAc (2 x 300 mL) and the combined organic layers were washed with brine (3 x 100 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (10%→50% EtOAc/pet. ether) to afford the desired product (4.23 g, 18.8% yield) and the atropisomer (5.78 g, 25.7% yield). LCMS (ESI) m/z: [M + H] calcd for C42H53N5O8: 724.41 ; found 724.4.

Intermediate 10. Synthesis of (2S)-N- ((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyrldin-3-yl)- 10,10-dimethyl-5,7-dloxo-2 5 -((trllsopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- lndola-6(1 ,3)-pyrldazlna-2(1 ,3)-benzenacycloundecaphane-4-yl)-3-methyl-2- (methylamlno)butanamlde

Step 1: Synthesis of (6 3 S,4S)-4-amino-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10- dimethyl-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-5,7-dione

A mixture of tert-butyl ((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7- dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (880 mg, 1.2 mmol), DCM (10 mL), and TFA (5 mL) was stirred at 0 °C for 30 min. The mixture was concentrated under reduced pressure to afford the desired product, which was used directly in the next step without further purification. LCMS (ESI) m/z: [M + H] calcd for C 4 sH 63 N s OsSi: 782.47; found 782.7.

Step 2. Synthesis of tert-butyl ((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamate

To a mixture of (6 3 S,4S)-4-amino-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-2 5 - ((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 W-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione (880 mg, 1.13 mmol) and N-(tert-butoxycarbonyl)-N-methyl-L- valine (521 mg, 2.3 mmol) in DMF (8.8 mL) at 0 °C was added DIPEA (1.95 mL, 11.3 mmol) and COMU (88 mg, 0.21 mmol). The mixture was stirred at 0 °C for 30 min, then diluted with H 2 O (100 mL) and extracted with EtOAc (3 x 100 mL). The combined organic layers were washed with brine (3 x 100 mL), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by prep-TLC to afford the desired product (1 g, 89% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for CseHeaNeOeSi: 995.61 ; found 995.5.

Step 3: Synthesis of (2S)-N- ((6 3 S,4S)-1 '-ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl- 5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)bu tanamide

A mixture of tert-butyl ((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10- dimethyl-SJ-dioxo^S-iitriisopropylsilyOoxyJ-e 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan- 2-yl)(methyl)carbamate (1.0 g, 1.0 mmol), DCM (10 mL) and TFA (5 mL) was stirred for 30 min. The mixture was concentrated under reduced pressure and the residue was basified to pH ~8 with sat. NaHCOs, then extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over Na 2 SO 4 , filtered, and the filtrate concentrated under reduced pressure to afford the desired product (880 mg, 98% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for CsiH74NeOeSi: 895.55; found 895.5.

Intermediate 11. Synthesis of (2S)- W-((63S,4S)-1 1 -ethyl-2 5 - hydroxy-1 ^(Φ- (methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1,3)-pyridazina-2(1,3)-benzenacyclounc^ecaphane-4-y l)-3-methy^2-(N-■methyl·2- (methylamino)acetamido)butanamide

JX o j o j

HATU, DIPEA BOO LiOH

DMF MeOH Step 1: Synthesis of methyl W-(W-(tert-butoxycarbonyl)-W-methylglycyl)-A/-methyl-L-valin ate To a solution of methyl methyl-L-valinate hydrochloride (2.0 g, 11.01 mmol) and N-{tert- butoxycarbonyl)-N-methylglycine (3.12 g, 16.51 mmol) in DMF (60.0 mL) at 0 °C was added DIPEA (9.58 mL, 55.01 mmol) and HATU (8.37 g, 22.02 mmol). The reaction mixture was stirred overnight and was then quenched with H 2 O (100 mL). The mixture was extracted with EtOAc (3 x 100 mL) and the combined organic layers were washed with brine (100 mL), dried over NaaSOA, and concentrated under reduced pressure. The residue was purified by reverse phase chromatography (40→60% MeCN/H 2 O) to afford the desired product (2.9 g, 83% yield) as an oil. LCMS (ESI) m/z: [M + H] calcd for C15H28N 2 O5:

317.21 ; found 317.4.

Step 2. Synthesis of A/-(A/-(tert-butoxycarbonyl)-A/-methylglycyl)-W-methyl-Z.-va line To a solution of methyl A/-(A/-(terf-butoxycarbonyl)-A/-methylglycyl)-W-methyl-L-val inate (3.70 g,

11.69 mmol) in THF (37.0 mL) was added a solution of LiOHeH 2 O (1.96 g, 46.71 mmol) in H 2 O (47.0 mL). The reaction mixture was stirred for 4 h, and then 1 M HCI was added until the pH was adjusted to 5. The resulting solution was extracted with EtOAc (3 x 100 mL) and the combined organic layers were washed with brine (3 x 50 mL), dried over Na 2 SO 4 , and concentrated under reduced pressure. The residue was purified by reverse phase chromatography (60→60% MeCN/H 2 O) to afford the desired product (1 .47 g,

41.6% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for C14H26N 2 O5: 303.19; found 303.4.

Step 3: Synthesis of tert-butyl (2-(((2S)-1-(((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)(methyl)carbamate

To a solution of (6 3 S,4S)-4-amino-1 ’-ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-2 s - ((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 'H-8-oxa-l (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione (300.0 mg, 0.384 mmol) and A/-(N-(tert-butoxycarbonyl)-N- methylglycyl)-N-methyl-L-valine (173.9 mg, 0.575 mmol) in DMF (3.0 mL) at 0 °C was added DIPEA (0.534 mL, 3.069 mmol) and PyBOP (399.2 mg, 0.767 mmol). The reaction mixture was stirred for 2 h and was then diluted with H 2 O (30 mL). The resulting mixture was extracted with EtOAc (3 x 20 mL) and the combined organic layers were washed with brine (3 x 20 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by normal phase chromatography (25% EtOAc/pet. ether) to afford the desired product (300 mg, 73% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for CseHeyNyOgSi: 1066.64; found 1067.4.

Step 4: Synthesis of tert-butyl (2-(((2S)-1-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(4- (methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)(methyl)carbamate

To a solution of tert-butyl (2-(((2S)-1-(((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan- 2-yl)(methyl)amino)-2- oxoethyl)(methyl)carbamate (355.0 mg) in THF (4.0 mL) at 0 °C was added TBAF (1.0 mL). The reaction mixture was stirred for 1 h and was then concentrated under reduced pressure. The residue was purified by normal phase chromatography (25% EtO Ac/pet. ether) to afford the desired product (280 mg, 92% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for C50H67N7O9: 910.51 ; found 911.0.

Step 5: Synthesis of (2S)-N- ((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-3-methyl-2-(N-methyl-2-(methyl amino)acetamido)butanamide

To a solution of tert-butyl (2-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-2®-hydroxy-1 2 -(4-(methoxymethyl)pyridin- 3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan- 2-yl)(methyl)amino)-2- oxoethyl)(methyl)carbamate (150.0 mg, 0.165 mmol) in DCM (2.0 mL) at 0 °C was added TFA (0.70 mL). The reaction mixture was stirred for 1 h and was then concentrated under reduced pressure to afford the desired crude product (150 mg) as a solid. LCMS (ESI) m/z: [M + H] calcd for C45H59N7O7: 810.46; found 810.4.

Intermediate 12. Synthesis of (3S)-N-((2S)-1 -(((6 3 S,4S)-1 1 -ethyF2 5 -hydroxy-1 2 -(4- (methoxymethyl)pyridln-3-yl)-10,10-dimethyl-5,7-dloxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)- lndola-6(1 ,3)-pyrldazlna-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)- N-methylpyrrolldlne-3-carboxamlde

Step 1: Synthesis of benzyl (S)-3-(((S)-1 -methoxy-3-methyl-1 -oxobutan-2- yl)(methyl)carbamoyl)pyrrolidine-1 -carboxylate

To a solution of methyl methyl-L-valinate hydrochloride (2.0 g, 13.8 mmol) and (S)-1- ((benzyloxy)carbonyl)pyrrolidine-3-carboxylic acid (4.12 mg, 16.5 mmol) in DMF (20.0 mL) at 0 °C was added DIPEA (12 mL, 68.870 mmol). The reaction mixture was stirred for 0.5 h, and then HATU (7.856 mg, 20.66 mmol) was added. The resulting mixture was warmed to room temperature and stirred for 1 h. The reaction mixture was then diluted with EtO Ac (800 mL) and was washed with sat. NH4CI (500 mL) and brine (3 x 350 mL). The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by normal phase chromatography (0→80% EtOAc/pet. ether) to afford the desired product (3.8 g, 73% yield) as an oil. LCMS (ESI) m/z:

[M + H] calcd for C20H28N 2 O5: 377.21 ; found 377.2.

Step 2. Synthesis of N-((S)-1 -((benzyloxy)carbonyl)pyrrolidine-3-carbonyl)-N-methyl-L-val ine

To a solution of benzyl (S)-3-(((S)-1 -methoxy-3-methyl-1 -oxobutan-2- yl)(methyl)carbamoyl)pyrrolidine-1-carboxylate (1 .125 g, 2.99 mmol) in MeOH (10.0 mL) was added a solution of LiOH (180.0 mg, 7.52 mmol) in H 2 O (2 mL). The reaction mixture was stirred for 4 h and was then quenched with sat. aq. NH4CI. The mixture with extracted with EtOAc (3 x 60 mL) and the combined organic layers were concentrated under reduced pressure to afford the desired product. LCMS (ESI) m/z: [M + H] calcd for C19H26N 2 O5: 363.19; found 363.2.

Step 3: Synthesis of tert-butyl ((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate

To a solution of tert-butyl ((6 3 S,4S)-1 '-ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl- 5,7-dioxo-2 5 -((triίsopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (1.70 g, 1.93 mmol) in THE (20 mL) at 0 °C was added TBAF (755.7 mg, 2.89 mmol). The reaction mixture was stirred for 2 h and was then quenched with H 2 O (200 mL). The resulting mixture was extracted with EtOAc (3 x 200 mL) and the combined organic layers were washed with brine (3 x 200 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by normal phase chromatography (17% EtOAc/pet. ether) to afford the desired product (1.1 g, 70% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for C41H51N5O7: 726.39; found 726.7.

Step 4: Synthesis of (6 3 S,4S)-4-amino-1 , -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)- 10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione

To a solution of tert-butyl ((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 e -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate (500.0 mg, 0.689 mmol) in DCM (10.0 mL) at 0 °C was added TEA (0.527 mL, 6.888 mmol). The resulting mixture was stirred for 1 h and then was concentrated under reduced pressure to afford the desired crude product (500 mg) as a solid. LCMS (ESI) m/z: [M + H] calcd for C36H43N5O5: 626.34; found 626.4.

Step 5: Synthesis of benzyl (3S)-3-(((2S)-1-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(4- (methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamoyl)pyrrolidine-1-carboxylate

To a solution of AA((S)-1-((benzyloxy)carbonyl)pyrrolidine-3-carbonyl)-N-meth yl-L-valine (676.4 mg, 6.31 mmol) in MeCN (10.0 mL) at 0 °C was added COMU (432.5 mg, 1.01 mmol). The reaction mixture was stirred for 5 min followed by the addition of (6 3 S,4S)-4-amino-1 1 -ethyl-2 5 -hydroxy-1 2 -(4- (methoxymethyl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 ' H-8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-5,7-dione (395.0 mg, 0.631 mmol). The reaction mixture was warmed to room temperature and stirred for 20 h. The mixture was then concentrated under reduced pressure, taken up in EtOAc (100 mL), and washed with brine (3 x 5 mL). The organic layer was dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by normal phase chromatography to afford a crude solid (0.81 g), which was then purified by reversed phase chromatography (MeCN/H 2 O) to afford the desired product (174 mg, 29% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for C55H67N7O9: 970.51 ; found 970.8.

Step 6: Synthesis of (3S)-N- ((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-2 ® -hydroxy-1 2 -(4-(methoxymethyl)pyridin- 3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan- 2-yl)-N-methylpyrrolidine-3- carboxamide

To a solution of benzyl (3S)-3-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(4- (methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamoyl)pyrrolidine-1-carboxylate (174.0 mg, 0.179 mmol) in MeOH (20.0 mL) was added Pd/C (87.0 mg, 0.08 mmol) followed by 2% aq. HCI (one drop). The reaction mixture was stirred at room temperature under a H2 atmosphere (1 atm) for 14 h, at which point the reaction mixture was purged with Na, filtered, and concentrated under reduced pressure to afford the crude product (130 mg, 86.7% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for C47H61N7O7: 836.47; found 836.5.

Intermediate 13. Synthesis of (2S)-2-(3-amlno-AAmethylpropanamldo)-A#-((6 3 S,4S)-1 1 -ethyl- 1 2 -(2-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dl oxo-2 s -((triisopropylsllyl)oxy)-

6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-3-methylbutanamide

Step 1: Synthesis of (2S)-N- ((6 3 S,4S)-1 1 -ethyl-1 2 -(2-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl- 5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-

2(1 ,3)-benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)bu tanamide

To a solution of tert-butyl ((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-(methoxymethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan- 2-yl)(methyl)carbamate (212.4 mg, 212 pmol) in DCM (500 pL) at 0 °C was added TFA (500 pL, 6.52 mmol). After 2 h, the reaction was diluted with DCM (10 mL) and H 2 O (10 mL), and then sat. aq. NaHCOswas added until the solution was pH 9. The aqueous layer was extracted with DCM (10 mL) and the combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the crude product (194 mg, 103% yield). LCMS (ESI) m/z: [M + H] calcd for CsihUNeOeSi: 895.55; found 895.7.

Step 2: Synthesis of tert-butyl (3-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-(methoxymethyl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-3-oxopropyl)carbamate

To a mixture of (2S)-N-((6 3 S,4S)-1 1 -ethyl-1 2 -(2-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7- dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)bu tanamide (150 mg, 167 pmol), COMU (88.5 mg, 206 pmol), and 3-((tert-butoxycarbonyl)amino)propanoic acid (39.6 mg, 209 μιτιοΙ) in MeCN (1 .66 mL) was added 2,6-lutidine (77.7 pL, 668 pmol). The reaction was stirred for 18 h at room temperature and then for 1 h at 55 °C. The reaction mixture was cooled to room temperature and was concentrated under reduced pressure. The crude residue was purified by reverse phase chromatography (20→60% MeCN/H 2 O) to afford the product (132 mg, 67% yield). LCMS (ESI) m/z: [M + H] calcd for C59He7N 7 09Si: 1066.64; found 1066.7.

Step 3: Synthesis of (2S)-2-(3-amino-N-methylpropanamido)-N-((6 3 S,4S)-1 1 -ethyl-1 2 -(2- (methoxymethyOpyridin-S-yO-10.10-dimethyl-S^-dioxo^-titriiso propylsilyOoxyl-e'.e 2 ^ 3 ^ 4 ^ 5 ^- hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)-3- methylbutanamide

To a solution of tert-butyl (3-(((2S)-1-(((6 3 S,4S)-1 , -ethyl-1 2 -(2-(methoxymethyl)pyridin-3-yl)-10,10- dimethyl-S^-dioxo^s-^triisopropylsilyOoxyi-e' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan- 2-yl)(methyl)amino)-3- oxopropyl)carbamate (120 mg, 112 pmol) in DCM (560 pL) at 0 °C was added TEA (560 pL, 7.30 mmol). After 40 min, the reaction was diluted with DCM (10 mL) and then sat. aq. NaHCGj was added. The organic layer was dried over Na 2 SO 4 , filtered, and then concentrated under reduced pressure to afford the product (106 mg, 98% yield), which was used in the next step without purification. LCMS (ESI) m/z:

[M + H] calcd for CsAHygNyOySi: 966.59; found 966.8.

Intermediate 14. Synthesis of (2S)-2-cyclo pentyl· W-((6 3 S,4S)-1 1 -ethy F2 s -hydroxy-1 2 -(2-((S)- 1 -methoxyethyl)pyridin-3-yl)-10,10-dimethy^5,7-d ioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 6 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1,3)-pyridazina-2(1,3)-benzenacycloundecaphane-4-yl )-2-(methylamino)acetamide

Step 1: Synthesis of tert- butyl ((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3- yl)-10,10-d imethy I-5 ,7-d ioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 ® -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate

To a stirred solution of tert-butyl ((6 3 S,4S)-1 '-ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (18.0 g, 20.1 mmol) in THF (180 mL) at 0 °C was added a 1 M solution of TBAF in THF (24.1 mL, 24.1 mmol). The mixture was stirred at 0 °C for 1 h, then diluted with brine (1.5 L), and extracted with EtOAc (3 x 1 L). The combined organic layers were washed with brine (2 x 500 mL), dried over anhydrous NaaStX filtered, and concentrated under reduced pressure. Purification by normal phase chromatography afforded the desired product (11.5 g, 69% yield). LCMS (ESI) m/z. [M + H] calcd for C42H53N5O7: 740.40; found 740.4.

Step 2. Synthesis of (6 3 S,4S)-4-amino-1 , -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)- 10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 , 6 5 ,6 ® -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione

To a stirred solution of tert-butyl ((6 3 S,4S)-1 '-ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 -methoxyethyl)pyridin- 3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 ® -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (11.5 g, 15.5 mmol) in DCM (120 mL) at 0 °C was added TFA (60 mL, 808 mmol). The mixture was stirred at 0 °C for 1 h, then concentrated under reduced pressure and the residue again concentrated under reduced pressure with toluene (3 x 20 mL) to afford the desired crude product (12 g). LCMS (ESI) m/z. [M + H] calcd for C37H45N5O5: 640.35; found 640.6.

Step 3: Synthesis of benzyl ((1 S)-1 -cydopentyl-2-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 ® -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-2-oxoethyl)(methyl)c arbamate

To a stirred solution of (6 3 S,4S)-4-amino-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3- yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 ® -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione (400.0 mg, 0.63 mmol) in DMF (4.0 mL) at 0 °C was added DIPEA (1.09 mL, 6.25 mmol) and (S)-2-(((benzyloxy)carbonyl)(methyl)amino)-2-cyclopentylacet ic acid (255.0 mg, 0.88 mmol) followed by COMU (347.8 mg, 0.81 mmol). The resulting mixture was stirred at 0 °C for 1 h and was then diluted with H 2 O (40 mL). The aqueous layer was extracted with EtOAc (3 x 15 mL) and the combined organic layers were washed with brine (2 x 10 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by prep-TLC (25% EtO Ac/pet. ether) to afford the desired product (510 mg, 80% yield). LCMS (ESI) m/z. [M + H] calcd for CsaHewNeOe: 913.49; found 913.6.

Step 4: Synthesis of (2S)-2-cyclopentyl-N-((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 6 3 ,6 4 ,6 5 ,6 ® -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-

6(1,3)-pyridazina-2(1,3)-benzenacycloundecaphane-4-yl)-2- (methylamino)acetamide

To a stirred solution of benzyl ((1 S)-1 -cyclopentyl-2-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1,3)-pyridazina-2(1,3)-benzenacycloundecaphane-4-yl)amino) -2-oxoethyl)(methyl)carbamate (480.0 mg, 0.53 mmol), in MeOH (25 mL) was added Pd/C (200.0 mg, 1.88 mmol). The resulting mixture was placed under an atmosphere of Hz (1 atm) and stirred for 2 h. The mixture was filtered, the filter cake was washed with MeOH (3 x 10 mL), and the filtrate was concentrated under reduced pressure to afford the desired crude product (440 mg). LCMS (ESI) m/z. [M + H] calcd for C+sHseNeO®: 779.45; found 779.4.

Intermediate 15. Synthesis of (2S)- N-((6 3 S,4S)-1 1 -ethyl-2 5 - hydroxy-1 2 -(4- (methoxymethyl)pyrkJin-3-yl)-10,10-dimethyl-5,7-dloxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- Ιη<1οΐ8-6(1,3)-ρνΓΐΰ8ζΙη8-2(1,3)-Ιιβηζβ η8θνοΙουη<Ιβοβρήβηβ-4-γΙ)-3-ηβΙήγ Ι-2-(/ν·ηβΙήγΙ-3- (methylamino)propanamldo)butanamlde Step 1: Synthesis of methyl N-(3-((fe/t-butoxycarbonyl)(methyl)amino)propanoyl)-N-methyl -L· valinate

To a solution of methyl methyl-L-valinate hydrochloride (1.0 g, 6.89 mmol) in DMF (20.0 mL) at 0 °C was added DIPEA (5.92 mL, 0.034 mmol), 3-((tert-butoxycarbonyl)(methyl)amino)propanoic acid (2.10 g, 0.010 mmol), and COMU (3.54 g, 8.27 mmol). The resulting mixture was stirred for 30 min and then quenched with H 2 O (20 mL). The aqueous layer was extracted with EtOAc (3 x 20 mL) and the combined organic layers were washed with brine (3 x 20 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by reverse phase chromatography (0→100% MeCN/ΗϋΟ) to afford the desired product (2 g, 87.9% yield). LCMS (ESI) m/z: (M + H] calcd for C16H30N 2 O5: 331.22; found 331.2.

Step 2. Synthesis of AA(3-((tert-butoxycarbonyl)(methyl)amino)propanoyl)-N-methyl -L-valine To a solution of N-(3-((terf-butoxycarbonyl)(methyl)amino)propanoyl)-N-methyl -L-valinate (1.0 g, 3.03 mmol) in THE (20.0 mL) and H 2 O (4.0 mL) was added LiOH (0.14 g, 6.05 mmol). The resulting mixture was stirred for 3 h at room temperature. The mixture was acidified to pH 3 with HCI (1 N) and was then extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (3 x 20 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired crude product (800 mg, 83.6% yield). LCMS (ESI) m/z: [M + H] calcd for C15H2BN 2 O5: 317.21 ; found 317.2.

Step 3: Synthesis of tert-butyl (3-(((2S)-1-(((6 3 S,4S)-1 1 -ethyl-2 s -hydroxy-1 2 -(4- (methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-3-oxopropyl)(methyl)carbamate

To a solution of (6 3 S,4S)-4-amino-1 , -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10- dimethyl-6 1 ,6 2 , 6 3 , 6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione (600.0 mg, 0.96 mmol) in DMF (6.0 mL) at 0 °C was added DIPEA (1.67 mL, 9.59 mmol), N-(3-((tert-butoxycarbonyl)(methyl)amino)propanoyl)-N-methyl -L-valine (455.1 mg,

1.44 mmol), and COMU (492.5 mg, 1.15 mmol). The resulting mixture was stirred for 30 min and was then quenched with H 2 O (60 mL). The aqueous layer was extracted with EtOAc (3 x 60 mL) and the combined organic layers were washed with brine (3 x 60 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by reverse phase chromatography (0→100% MeCN/H20) to afford the desired product (650 mg, 73.4% yield). LCMS (ESI) m/z: [M + H] calcd for CSIHWNTOS: 924.52; found 924.6.

Step 4: Synthesis of (2S)-N- ((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-3-methyl-2-(N-methyl-3-(methyl amino)propanamido)butanamide

To a solution of tert-butyl (3-(((2S)-1-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin- 3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan- 2-yl)(methyl)amino)-3- oxopropyl)(methyl)carbamate (650.0 mg) in DCM (7.0 mL) at 0 °C was added TFA (3.5 mL). The resulting mixture was stirred for 30 min and was then concentrated under reduced pressure. The resulting residue was diluted with toluene (3 x 10 mL) and concentrated under reduced pressure to afford the desired crude product. LCMS (ESI) m/z: [M + H] calcd for C46H61N7O7: 824.47; found 824.6. Intermediate 16. Synthesis of (2S)-2-cyclo pentyl· W-((6 3 S,4S)-1 1 -ethy L2 s -hydroxy-1 2 -(2-((S)- 1 -methoxyethyl)pyridin-3-yl)-10,1 Ο-^ΙίιηβΙΙιγΜ,Τ-^Ιίοχο-β 1 ,6 2 ,6 3 ,6 4 ,6 6 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)-2-(N-methyl-2- (methylamino)acetamido)acetamide

Step 1: Synthesis of tert-butyl (2-(((1 S)-1 -cyclopentyl-2-(((6 3 S,4S)-1 '-ethyl-2 5 -hydroxy-1 2 -(2-((S)- 1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-2-oxoethyl)(methyl)a mino)-2- oxoethyl)(methyl)carbamate

To a mixture of (2S)-2-cyclopentyl-N-((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)-2-(methylamino)acetamide (300.0 mg, 0.385 mmol), DIPEA (0.657 mL, 3.851 mmol), and A/-(tert-butoxycarbonyl)-A/-methylglycine (109.30 mg, 0.578) in DMF (3.0 mL) at 0 °C was added HATU (175.72 mg, 0.462 mmol). The resulting mixture was stirred at 0 °C for 30 min and was then diluted with H 2 O (30 mL). The resulting mixture was extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (3 x 30 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by prep-TLC (50% EtO Ac/pet. ether) to afford the desired product (300 mg, 82.0% yield). LCMS (ESI) m/z. [M + H] calcd for C53H71N7O9: 950.54; found 950.4.

Step 2: Synthesis of (2S)-2-cyclopentyl-N-((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)-2-(N-methyl-2- (methylamino)acetamido)acetamide

To a mixture of tert-butyl (2-(((1 S)-1 -cyclopentyl-2-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 ® -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-2-oxoethyl)(methyl)a mino)-2- oxoethyl)(methyl)carbamate (300.0 mg, 0.316 mmol) in DCM (3.0 mL) at 0 °C was added TFA (1 .50 mL). The resulting mixture was stirred at 0 °C for 30 min and was then concentrated under reduced pressure to afford the desired crude product. LCMS (ESI) m/z: [M + H] calcd for CWHMNTO?: 850.49; found 850.5.

Intermediate 17. Synthesis of PHSfll-AHpSH-ttte^SM^thyH^-MSM- methoxyethyl)pyridin-3-yl)-1 Ο,ΙΟ-^ΙιηβΙΙιγΜΛΰΙοχο^-ΜΜΙβορΜΐρ ΙβΙΙγΙΙοχνΗΐ 1 ,6 z ,6 3 ,6 4 ,6 e ,6 8 - hexahydro-1 1 H- 8-oxa-1 (5,3)-lndola-6(1 ,3)-pyrkJazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amlno)- 3-methyl-1-oxobutan-2-yl)-jV,5-dlmethylpyrrolldlne-2-cait> ;oxamkle

Step 1: Synthesis of (6 3 S,4S)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10- dimethyl-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 ' H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-5,7-dione

To a solution of tert-butyl ((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (20.0 g, 22.315 mmol) in DCM (150.0 mL) at 0 °C was added TFA (50.0 mL). The resulting mixture was warmed to room temperature and stirred for 2 h and then concentrated under reduced pressure. The residue was dissolved in EtOAc (100 mL) and the solution was neutralized to pH 8 with sat. aq. NaHCOa. The solution was extracted with EtOAc (3 x 150 mL) and the combined organic layers were washed with brine (100 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (17.86 g, crude). LCMS (ESI) m/z. [M + H] calcd for CwHroNsOsSi: 796.49; found 795.5

Step 2: Synthesis of benzyl ((2S)-1 -(((6 3 S,4S)-1 1 -ethyl- 1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-

6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamate

To a solution of (6 3 S,4S)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl- 2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione (17.86 g, 22.433 mmol) and (2S)-2-

[[(benzyloxy)carbonyl](methyl)amino]-3-methylbutanoic acid (8.93 g, 33.65 mmol) in DMF (150.0 mL) at 0 °C was added DIPEA (19.5 mL, 112.17 mmol) and HATU (17.06 g, 44.87 mmol). The resulting mixture was warmed to room temperature and stirred for 2 h. The reaction mixture was cooled to 0 °C and was quenched by the addition of H 2 O (500 mL). The mixture was extracted with EtOAc (3 x 150 mL) and the combined organic layers were washed with brine (200 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by normal phase chromatography (25% EtOAc/pet. ether) to afford the desired product (19.0 g, 81 .2% yield). LCMS (ESI) m/z. [M + H] calcd for CeoHeaNeOeSi: 1043.61 ; found 1042.6

Step 3: Synthesis of (2S)-N- ((6 3 S,4S)-1 '-ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10- dimethyl-5, 7-dioxo-2 5 -((triisopropylsilyl)oxy)-6' ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)bu tanamide

To a solution of benzyl ((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 e -hexahydro-1 ' H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan- 2-yl)(methyl)carbamate (1.20 g, 1.150 mmol) in MeOH (1.2 mL) and toluene (1.2 mL) was added Pd/C (10%, 240 mg). The resulting mixture was placed under an atmosphere of Ha (1 atm) and stirred overnight. The mixture was filtered and concentrated under reduced pressure to afford the desired product (1.05 g, 97.4% yield). LCMS (ESI) m/z. [M + H] calcd for CsHyeNeOeSi: 909.57; found 909.3.

Step 4: Synthesis of tert-butyl (2/?,5fl)-2-(((2S)-1-(((6 3 S,4S)-1 '-ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro- 1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 - oxobutan-2-yl)(methyl)carbamoyl)-5-methylpyrrolidine-1-carbo xylate

To a solution of (2S)-N-((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl- 5,7-dioxo-2 s -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 ® -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)bu tanamide (500 mg, 0.550 mmol) in DMF (5 mL) at 0 °C was added DIPEA (0.94 mL, 5.499 mmol) and (2fl,5fl)-1 -(tert-butoxycarbonyl)-5- methylpyrrolidine-2-carboxylic acid (504.29 mg, 2.199 mmol) followed by HATU (627.23 mg, 1.650 mmol) in portions. The resulting mixture was warmed to room temperature and stirred for 1 h. Purification by reverse phase chromatography (0→100% MeCN/H 2 O) afforded the desired product (147 mg, 22.2% yield). LCMS (ESI) m/z. [M + H] calcd for CesHgaNyOgSi: 1120.69; found 1120.6.

Step 5: Synthesis of (2f?,5f?)-N-((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)- 10,10-dimethyl-S^-dioxo^-iitriisopropylsilyOoxyi-e 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)-/V,5- dimethylpyrrolidine-2-carboxamide

To a solution of tert-butyl (2/?,5/¾-2-(((2S)-1 -(((6 3 S,4S)-1 '-ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin- 3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamoyl)-5-methylpyrrolidine-1 -carboxylate (150.0 mg, 0.134 mmol) in DCM at 0 °C was added TFA (1.50 mL, 13.155 mmol) dropwise. The resulting mixture was warmed to room temperature and stirred for 2 h and was then basified to pH 8 with sat. NaHCOs. The resulting mixture was extracted with EtOAc (3 x 5 mL) and the combined organic layers were washed with brine (2 x 5 mL), dried with Na 2 SO 4 . filtered, and concentrated under reduced pressure to afford the desired product (85 mg, 54.1% yield). LCMS (ESI) m/z. [M + H] calcd for CseHnsNyOSi: 1020.64; found 1020.4.

Intermediate 18. Synthesis of (2fl)-Af-((2S)-1-(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 1 Wt-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)- 3-methyl-1 -oxobutan-2-yl)-i-methylpyrrolidine-2-carboxamlde Step 1: Synthesis of (tert-butoxycarbonyl)-D-proline To a solution of D-proline (5.0 g, 43.43 mmol) in 1 ,4-dioxane (50 mL) and sat. NaHCOa (50 mL) at 0 °C was added B0C2O (14.217 g, 65.143 mmol) in portions. The resulting mixture was stirred for 2 h at room temperature and was then extracted with EtOAc (100 mL). The aqueous layer was acidified to pH 6 with HCI and was then extracted into EtOAc (3 x 100 mL). The combined organic layers were washed with H 2 O (2 x 100 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product which was used without further purification. LCMS (ESI) m/z. [M - H] calcd for C10H17NO4: 214.11 ; found 214.0.

Step 2: Synthesis of tert-butyl (2fl)-2-(((2S)-1-(((6 3 S,4S)-1'-ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro- 1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 - oxobutan-2-yl)(methyl)carbamoyl)pyrrolidine-1-carboxylate

To a solution of (2S)-N-((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl- 5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6' ,6 2 ,6 3 ,6 4 -6 5 ,6 6 -hexahydro-1 ' H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)bu tanamide (142.03 mg, 0.660 mmol) in DMF was added DIPEA (0.710 mL, 5.499 mmol) followed by HATU (250.89 mg, 0.660 mmol) in portions. The resulting mixture was heated to 40 °C and stirred for 2 h. Purification by reverse phase chromatography (0→100% MeCN/H 2 O) afforded the desired product (350 mg, 54.6% yield). LCMS (ESI) m/z. [M + H] calcd for CeaHaiNyOgSi: 1106.67; found 1106.8.

Step 3: Synthesis of (2fl)-N-((2S)-1 -(((6 3 S,4S)-1 '-ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-2 s -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)-i- methylpyrrolidine-2-carboxamide

To a solution of tert-butyl (2fl)-2-(((2S)-1 -(((6 3 S,4S)-1 '-ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3- yl)-10,10-d imethyl-5 ,7-d ioxo-2 5 -((triisopropylsily l)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-

6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamoyl)pyrrolidine-1-carboxylate (350.0 mg, 0.325 mmol) in DCM (4 mL) at 0 °C was added TFA (2.0 mL). The resulting mixture was stirred for 30 min at 0 °C and then was concentrated under reduced pressure. The residue was dissolved in toluene (5 mL) then concentrated under reduced pressure three times to afford the desired product which was used without further purification. LCMS (ESI) m/z. [M + H] calcd for CsyMesNyOSi: 1006.62; found 1006.4.

Intermediate 19. Synthesis of (2fl)-Af-((2S)-1-(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)- 3-methyl-1-oxobutan-2-yl)-N-methylazetldine-2-carboxamlde Step 1: Synthesis of tert-butyl (2fl)-2-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro- 1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1- oxobutan-2-yl)(methyl)carbamoyl)azetidine-1-carboxylate

To a mixture of (2S)-N-((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl- 5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)bu tanamide (1.0 g, 1.10 mmol), {Ft)- 1- (tert-butoxycarbonyl)azetidine-2-carboxylic acid (0.33 g, 1.650 mmol) and HATU (1.25 g, 3.299 mmol) in MeCN (20 mL) at 0 °C was added DIPEA (0.94 mL, 5.499 mmol). The resulting mixture was stirred at 0 °C for 3 h and then was concentrated under reduced pressure. Purification by prep-TLC (10%

MeOH/DCM) afforded the desired product (800 mg, 59.9% yield). LCMS (ESI) m/z: [M + H] calcd for CeiHeeNTCfeSi: 1092.65; found 1092.6.

Step 2. Synthesis of (2fl)-N-((2S)-1-(((6 3 S,4S)-1 ' -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)-N- methylazetidine-2-carboxamide

To a mixture of tert-butyl (2f?)-2-(((2S)-1-(((6 3 S,4S)-1 '-ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3- yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamoyl)azetidine-1 -carboxylate (400.0 mg, 0.366 mmol) in DCM (8.0 mL) at 0 °C was added TFA (4.0 mL). When the reaction was complete the mixture was concentrated under reduced pressure to afford the desired product (400 mg, crude). LCMS (ESI) m/z: [M + H] calcd for CseHeiNyOySi: 992.61 ; found 992.4.

Intermediate 20. Synthesis of N-(sec-butyl)-5-((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-10,10-dlmethyl- 4-((S)-3-methyl-2-(N-methyl-2-(methylamino)acetamido)butanam ldo)-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 e - hexahydro-1 1 H4l·· oxa-1 (5,3)-lndola-6(1 ,3)-pyrldazlna-2(1 ,3)-benzenacycloundecaphane-1 2 -yl)-AA methylnicotlnamide

Step 1: Synthesis of 5-bromo-N-(sec-butyl)-N-methylnicotinamide

To a solution of 5-bromonicotinic acid (2.0 g, 9.901 mmol) and HATU (5.65 g, 14.851 mmol) in DMF (40 mL) at 0 °C was added DIPEA (5.2 mL 9.9 mmol). The resulting mixture was stirred for 30 min at 0 °C and then N-methylbutan-2-amine (0.91 g, 10.396 mmol) was added. The resulting mixture was warmed to room temperature and stirred overnight, then diluted with H 2 O (40 mL). The mixture was extracted with EtOAc (3 x 30 mL) and the combined organic layers were washed with brine (50 mL), dried over Na 2 SO 4 . filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (50% EtOAc/pet. ether) to afford the desired product (1.96 g, 73.2% yield). LCMS (ESI) m/z. [M + H] calcd for CnHisBrNaO: 271.04; found 271.1.

Step 2. Synthesis of tert-butyl ((6 3 S,4S)-25-(benzyloxy)-1 2 -(5 -{see- butyl(methyl)carbamoyl)pyridin-3-yl)-10,10-dimethyl-5,7-diox o-6',6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa- 1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate

To a solution of 5-bromo-N-(sec-butyl)-N-methylnicotinamide (800.0 mg, 2.95 mmol) and K3PO3

(1.565 g, 7.376 mmol) in 1 ,4-dioxane (30.0 mL) and H 2 O (6.0 mL) was added tert-butyl ((6 3 S,4S)-2 5 - (benzyloxy)-l 0,10-dimethyl-5,7-dioxo-1 2 -(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-6' hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (2.81 g, 3.540 mmol) and Pd(dppf)Cl 2 (215.87 mg, 0.295 mmol). The resulting mixture was heated to 85 °C and stirred for 3 h. The mixture was then cooled to room temperature, quenched with H 2 O, and extracted with EtOAc (3 x 100 mL). The combined organic layers were washed with H 2 O (100 mL), dried over Na 2 SO 4 , filtered, and concentrated. The residue was purified by silica gel chromatography (10% MeOH/DCM) to afford the desired product (2.2 g, crude). LCMS (ESI) m/z. [M + H] calcd for CsoHeoNeO?: 857.46; found 857.5. Step 3: Synthesis of tert-butyl ((6 3 S,4S)-2 5 -(benzyloxy)-1 2 -(5-(sec-butyl(methyl)carbamoyl)pyridin- 3-yl)-1 '-ethyl-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 e -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate

To a solution of tert-butyl ((6 3 S,4S)-25-(benzyloxy)-1 2 -(5-(sec-butyl(methyl)carbamoyl)pyridin-3- yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate (2.10 g, 2.450 mmol) and CsaCOs (2.39 g, 7.351 mmol) in DMF (20.0 mL) was added ethyl iodide (0.57 g, 3.675 mmol). The resulting mixture was stirred for 3 h at room temperature and was then quenched with H 2 O (200 mL). The resulting mixture was extracted with EtOAc (3 x 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. The residue was purified by silica gel chromatography (10% MeOH/DCM) to afford the desired product (800 mg, 36.9% yield). LCMS (ESI) m/z. [M + H] calcd for CsaHwNeOy: 885.49; found 885.5.

Step 4: Synthesis of tert-butyl ((6 3 S,4S)-1 2 -(5-(seo-butyl(methyl)carbamoyl)pyridin-3-yl)-1 1 -ethyl- 2 5 -hydroxy-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate

To a solution of tert-butyl ((6 3 S,4S)-2 5 -(benzyloxy)-1 2 -(5-(sec-butyl(methyl)carbamoyl)pyridin-3- yl)-1 '-ethyl-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (770.0 mg, 0.870 mmol) in tert-BuOH (20.0 mL) was added Pd(OH)2/C (24.42 mg, 0.174 mmol). The resulting suspension was stirred overnight at 50 °C under a hydrogen atmosphere (1 atm). The mixture was then cooled to room temperature, filtered and the filter cake was washed with MeOH (3 x 30 mL). The filtrate was concentrated under reduced pressure to afford the desired product (810 mg, crude). LCMS (ESI) m/z. [M + H] calcd for CAsHseNeO?: 795.44; found 795.5.

Step 5: Synthesis of tert-butyl ((6 3 S,4S)-1 2 -(5-(seobutyl(methyl)carbamoyl)pyridin-3-yl)-1 1 -ethyl- 10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-

6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate

To a solution of tert-butyl ((6 3 S,4S)-1 2 -(5-(sec-butyl(methyl)carbamoyl)pyridin-3-yl)-1 ’-ethyl-2 5 - hydroxy-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (800.0 mg, 1 .0 mmol) and DIPEA (0.876 mL, 5.031 mmol) in MeCN (10.0 mL) was added chlorotris(propan-2-yl)silane (291.02 mg, 1.509 mmol). The resulting mixture was stirred for 3 h and was then quenched with H 2 O. The resulting mixture was extracted with EtOAc (3 x 50 mL) and the combined organic layers were washed with H 2 O (3 x 30 mL), dried over NaaSOA, filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (10% MeOH/DCM) to afford the desired product (800 mg, 83.6% yield). LCMS (ESI) m/z. [M + H] calcd for Cs+HyeNeOySi: 951.58; found 950.8.

Step 6: Synthesis of 5-((6 3 S,4S)-4-amino-1 1 -ethyl-10,10-dimethyl-5,7-dioxo-2 5 - ((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-1 2 -yl)-N-(sec-butyl)-N-methylnicotinamide

To a solution of tert-butyl ((6 3 S,4S)-1 2 -(5-(seobutyl(methyl)carbamoyl)pyridin-3-yl)-1 1 -ethyl-10,10- dimethyl-5, 7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 ' H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (720.0 mg, 0.757 mmol) in DCM (10.0 mL) at 0 °C was added TFA (3.0 mL, 40.4 mmol). The resulting mixture was stirred for 2 h and was then concentrated under reduced pressure. The residue was cooled to at 0 °C and neutralized with sat. aq. NaHCOs. The resulting mixture was extracted with EtOAc (3 x 100 mL) and the combined organic layers were washed with H 2 O (3 x 30 mL), dried over Na2SC>4, filtered, and concentrated under reduced pressure to afford the desired product (540 mg, crude). LCMS (ESI) m/z. [M + H] calcd for CtgHyoNeOsSi: 851.53; found 851.8.

Step 7\ Synthesis of benzyl ((2S)-1-(((6 3 S,4S)-1 2 -(5-(sec-butyl(methyl)carbamoyl)pyridin-3-yl)-1 1 - ethyl-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamate

To a solution of 5-((6 3 S,4S)-4-amino-1 1 -ethyl-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 e -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane- 1 2 -yl)-N-(sec-butyl)-/\Amethylnicotinamide (530.0 mg, 0.623 mmol) and A/-((benzyloxy)carbonyl)-N-methyl- L-valine (198.23 mg, 0.747 mmol) in DMF (10.0 mL) were added HATU (473.49 mg, 1.245 mmol) and DIPEA (0.542 mL, 3.113 mmol). The resulting mixture was stirred for 2 h and was then quenched with H 2 O and extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with H 2 O (3 x 30 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (10% MeOH/DCM) to afford the desired product (720 mg, crude). LCMS (ESI) m/z. [M + H] calcd for CeaHe/NTOeSi: 1098.65; found 1098.7.

Step 8: Synthesis of N-(seo-butyl)-5-((6 3 S,4S)-1 1 -ethyl-10,10-dimethyl-4-((S)-3-methyl-2- (methylamino)butanamido)-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6®-hexahydro-1 1 H- 8-oxa-

1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-1 2 -yl)-N-methylnicotinamide

To a solution of benzyl ((2S)-1-(((6 3 S,4S)-1 2 -(5-(sec-butyl(methyl)carbamoyl)pyridin-3-yl)-1 1 -ethyl- 10,10-dimethyl-5,7-dioxo-2 s -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamate (670.0 mg, 0.610 mmol) in toluene (10.0 mL) and MeOH (1.0 mL) was added Pd/C (12.98 mg, 0.122 mmol). The suspension was stirred overnight under a hydrogen atmosphere (1 atm) and was then filtered, and the filter cake washed with MeOH (3 x 50 mL). The filtrate was concentrated under reduced pressure to afford the desired product (600 mg, crude). LCMS (ESI) m/z. [M + H] calcd for CssHeiNyOeSi: 964.61 ; found 964.8.

Step 9·. Synthesis of tert-butyl (2-(((2S)-1-(((6 3 S,4S)-1 2 -(5-(seo-butyl(methyl)carbamoyl)pyridin-3- yl)-1 1 -ethyl-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa- 1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)(methyl)carbamate

To a solution of N-(sec-butyl)-5-((6 3 S,4S)-1 1 -ethyl-10,10-dimethyl-4-((S)-3-methyl-2- (methylamino)butanamido)-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-

1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-1 2 -yl)-A/-methylnicotinamide (490.0 mg, 0.508 mmol) and N-(tert-butoxycarbonyl)-N-methylglycine (114.4 mg, 0.610 mmol) in DMF (10.0 mL) was added HATU (386.39 mg, 1 .016 mmol) and DIPEA (0.443 mL, 2.540 mmol). The resulting mixture was stirred for 2 h and was then quenched with H 2 O and extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with H 2 O (3 x 30 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (560 mg, 79.3% yield). LCMS (ESI) m/z. [M + H] calcd for C63H94NBO9S1: 1135.70; found 1136.3. Step 10: Synthesis of tert-butyl (2-(((2S)-1 -(((6 3 S,4S)-1 2 -(5-(seobutyl(methyl)carbamoyl)pyridin- 3-yl)-1 ’-ethyl-2 5 -hydroxy-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)(methyl)carbamate

To a solution of tert-butyl (2-(((2S)-1-(((6 3 S,4S)-1 2 -(5-(seobutyl(methyl)carbamoyl)pyridin-3-yl)-

1 1 -ethyl-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)(methyl)carbamate (540.0 mg, 0.476 mmol) in DMF (10.0 mL) was added CsF (288.94 mg, 1.90 mmol). The resulting mixture was stirred for 2 h and was then quenched with H 2 O and extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with H 2 O (3 x 30 mL), dried over Na 2 SO 4 . filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (10% MeOH/DCM) to afford the desired product (430 mg, crude). LCMS (ESI) m/z. [M + H] calcd for Cs^NeOs: 979.57; found 980.0.

Step 11: Synthesis of N-(seo-butyl)-5-((6 3 S,4S)-1 '-ethyl-2 5 -hydroxy-10,10-dimethyl-4-((S)-3- methyl-2-(N-methyl-2-(methylamino)acetamido)butanamido)-5,7- dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 /+8- oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-1 2 -yl)-N-methylnicotinamide

To a solution of tert-butyl (2-(((2S)-1-(((6 3 S,4S)-1 2 -(5-(sec-butyl(methyl)carbamoyl)pyridin-3-yl)- 1 1 -ethyl-2 s -hydroxy-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 ' H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan- 2-yl)(methyl)amino)-2- oxoethyl)(methyl)carbamate (400.0 mg, 0.408 mmol) in DCM (10.0 mL) at 0 °C was added TFA (3.0 mL, 40.4 mmol). The reaction was stirred for 1 h and was thenquenched with sat. aq. NaHCOa. The mixture was extracted with EtOAc (3 x 50 mL) and the combined organic layers were washed with h½0 (3 x 30 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (380 mg, crude). LCMS (ESI) m/z. [M + H] calcd for Ο^ΗββΝβΟ?: 879.51 ; found 879.5.

Intermediate 21. Synthesis of (2Q-2-(2-amlno-N-methylacetamido)-Af-((6 3 ¾4S)-1 1 -ethyl-2 e - hydroxy-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 e - hexahydro-1 1 M-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3>-benzenacyclou ndecaphane-4-y l)-3- methylbutanamide

Step 1: Synthesis of benzyl (2-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-

10.10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)carbamate

To a solution of (2S)-N-((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl- 5,7-dioxo-2 s -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 ® -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)bu tanamide (2.50 g, 2.75 mmol) and ((benzyloxy)carbonyl)glycine (690 mg, 3.30 mmol) in DMF (25 mL) at 0 °C was added HATU (2.10 g, 5.50 mmol) followed by DIPEA (1.5 mL, 8.25 mmol). The reaction mixture was stirred for 2 h and was then quenched with H 2 O and extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with H 2 O (3 x 10 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by silica gel column chromatography (50% EtOAC/hexanes) afforded desired product (2.0 g, 72% yield). LCMS (ESI) m/z. [M + H] calcd for CeaHestoOgSi: 1100.63; found 1100.7.

Step 2. Synthesis of benzyl (2-(((2S)-1-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-

6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)carbamate

To a solution of benzyl (2-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-

10.10-dimethyl-5,7-dioxo-2 s -((triisopropylsilyl)oxy)- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 ® -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)carbamate (400 mg, 0.36 mmol) in DMF at 0 °C was added CsF (220 mg,

1.5 mmol). The reaction mixture was stirred for 2 h and was then quenched with HzO and extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with h½0 (3 x 10 mL), dried with Na 2 SO 4 . filtered, and concentrated under reduced pressure to afford the desired product (300 mg, 87% yield). LCMS (ESI) m/z. [M + H] calcd for C53H65N7O9: 944.49; found 944.4. Step 3: Synthesis of (2S)-2-(2-amino-N-methylacetamido)-N-((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2- ((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1,3)-pyridazina-2(1,3)-benzenacycloundecaphane-4-yl )-3-methylbutanamide To a solution of benzyl (2-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-

6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)carbamate (300 mg, 0.32 mmol) in toluene (10 mL) and MeOH (1 mL) was added Pd/C (50 mg, 0.47 mmol). The suspension was stirred overnight under an atmosphere of hydrogen (1 atm). The reaction mixture was then was filtered and the filter cake was washed with EtOAc (3 x 10 mL). The filtrate was concentrated under reduced pressure to afford the desired product (180 mg, 43% yield). LCMS (ESI) m/z. [M + H] calcd for CASHMNTO?: 810.46; found 810.5.

Intermediate 22. (3S,4fl)-M L ((2S)-1 -(((6 3 S,4S)-1 1 -ethyl- 1 *-(2-((S)-1 -methoxyethyl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-2 6 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)- lndola-6(1 ,3)-pyrldazlna-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)-

/V,4-dlmethylpyrrolldlne-3-carboxamlde

Step 1: Synthesis of (fl)-3-(but-2-ynoyl)-4-phenyloxazolidin-2-one

To a solution of 2-butynoic acid (5.0 g, 59.47 mmol) in THF (100 mL) at -78 °C was added pivalic acid chloride (7.39 g, 61.26 mmol) and EtsN (6.2 mL, 61.85 mmol) and then the mixture was stirred for 15 min and then warmed to 0 °C and stirred for 45 min. In a second flask, to a solution of (4fl)-4-phenyl-1 ,3- oxazolidin-2-one (9.70 g, 59.47 mmol) in THF (100 mL) at -78 °C was added n-BuLi (2.5 M in hexane, 25 mL, 62.5 mmol). The mixture was stirred at -78 °C for 15 min and was then added to the initial mixture. The combined solutions were warmed to room temperature and stirred overnight. The reaction solution was quenched with sat. NFUCI (200 mL) and then the mixture was extracted with EtOAc (3 x 100 mL). The combined organic layers were washed with brine (200 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by normal phase chromatography (20% EtOAc/pet. ether) afforded the desired product (6.0 g, 44.0% yield). LCMS (ESI) m/z. [M + H] calcd for C13H11NO3: 230.08; found 229.9.

Step 2: Synthesis of (f?,Z)-3-(but-2-enoyl)-4-phenyloxazolidin-2-one

To a solution of (fl)-3-(but-2-ynoyl)-4-phenyloxazolidin-2-one (6.0 g, 26.17 mmol) in pyridine (6.0 mL) and toluene (60.0 mL) at 0 °C was added Lindlar Pd catalyst (594.57 mg, 2.88 mmol). The resulting mixture was stirred for 30 min at 0 °C under a hydrogen atmosphere (1 atm). The mixture was filtered, and the filter cake was washed with toluene (10.0 mL). The filtrate was concentrated under reduced pressure to afford the desired product (5.5 g, crude). LCMS (ESI) m/z: [M + H] calcd for C13H13NO3: 232.10; found 231.9.

Step 3: (fl)-3-((3S,4fl)-1 -benzyl-4-methylpyrrolidine-3-carbonyl)-4-phenyloxazolidin-2 -one

To a solution of (R,2)-3-(but-2-enoyl)-4-phenyloxazolidin-2-one (3.0 g, 12.97 mmol) and benzyl(methoxymethyl)[(trimethylsilyl)methyl]amine (3.70 g, 15.57 mmol) in toluene (20.0 mL) at 0 °C was added TFA (1.30 mL, 0.87 mmol). The resulting mixture was warmed to room temperature and stirred overnight. The mixture was then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (20% EtOAc/pet. ether) to afford the desired product (2 g, 42.3% yield). LCMS (ESI) m/z: [M + H] calcd for C22H24N 2 O3: 365.19; found 365.2.

Step 4: Synthesis of (3S,4fl)-1-benzyl-4-methylpyrrolidine-3-carboxylic acid A solution of LiOHeH 2 O (0.16 g, 6.860 mmol) and H 2 O2 (0.13 g, 3.76 mmol) in H 2 O (5 mL) was added to a solution of (fl)-3-((3S,4fl)-1-benzyl-4-methylpyrrolidine-3-carbonyl)-4- phenyloxazolidin-2-one (1.0 g, 2.74 mmol) in THE (15.0 mL) at 0 °C. The resulting mixture was stirred for 2 h and was then quenched with H 2 O (30 mL) and sodium sulfite (0.69 g, 5.48 mmol) and the solution was extracted with EtOAc (2 x 50 mL). The aqueous phase was adjusted to pH 4 with NaHaPCUeH 2 O and 10% HCI, and the brine was added. The solution was extracted with APrOH/DCM (1 :3, 5 x 50 mL) and the combined organic layers were washed with brine (40 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (400 mg, crude). LCMS (ESI) m/z·. [M + H] calcd for C13H17NO2: 220.14; found 220.2.

Step 5: Synthesis of (3S,4fl)-1 -benzyl-AA((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 , ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro- 1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 - oxobutan-2-yl)-/V,4-dimethylpyrrolidine-3-carboxamide

To a mixture of (2S)-N-((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl- 5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-

2(1 ,3)-benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)bu tanamide (414.67 mg, 0.456 mmol) and (3S,4fl)-1-benzyl-4-methylpyrrolidine-3-carboxylic acid (200.0 mg, 0.912 mmol) in DMF (5.0 mL) at 0 °C was added HATU (693.58 mg, 1.824 mmol) and DIPEA (0.794 mL, 4.560 mmol). The resulting mixture was warmed to room temperature and stirred for 2 h. The reaction was quenched with the addition of sat. aq. NH4CI (40 mL) and then extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (2 x 20 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by prep-TLC (9% MeOH/DCM) to afford the desired product (350 mg, 34.6% yield). LCMS (ESI) m/z: [M + H] calcd for CesHeiNyO/Si: 1110.68; found 1110.9.

Step 6: (3S,4fl)-N-((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10- dimethyl-5, 7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan- 2-yl)-/V,4- dimethylpyrrolidine-3-carboxamide

To a solution of (3S,4fl)-1-benzyl-AA((2S)-1-(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin- 3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)-/V,4- dimethylpyrrolidine-3-carboxamide (300.0 mg, 0.270 mmol) in f-BuOH (10.0 mL) was added Pd/C (60.08 mg, 0.565 mmol). The resulting suspension was stirred overnight under a hydrogen atmosphere (1 atm). The mixture was then filtered, the filter cake was washed with MeOH (2 x 5 mL), and the filtrate was concentrated under reduced pressure to afford the desired product (280 mg, crude). LCMS (ESI) m/z. [M + H] calcd for CseHroNyOySi: 1020.64; found 1020.8.

Intermediate 23. Synthesis of (2S)- N-((6 3 S,4S)-1 1 -ethyl-2 5 - hydroxy-1 ^-((SH- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 e -hexahydro-1 1 H-8-oxa-1 (5,3)- indola-6(1,3)-pyridazlna-2(1,3)-benzenacycloundecaphane-4-yl )-3-methyl-2-((S}-N- methyl-2- (methylamino)propanamldo)butanamide

? v HO ο γ 0ν>

A O H ΝΗ T T A Η ,Boc

MeO HATU, DIPEA MeO

N •5s, DMF

(/ mps N-

(/ // OTIPS

N' N'

<v

° Y Ov

A H > o H u I =

NBoc NH

TBAF MeO I TFA MeO I

THF N sW DCM ·¾ ¾ . f OH ΌΗ

(/ f

N' N'

Step 1: Synthesis of tert-butyl ((2S)-1 -(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro- 1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 - oxobutan-2-yl)(methyl)amino)-1-oxopropan-2-yl)(methyl)carbam ate

To a solution of (2S)-N-((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl- 5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-

2(1 ,3)-benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)bu tanamide (500.0 mg, 0.55 mmol), DIPEA (480 mL, 2.75 mmol) and (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]propanoic acid (167.63 mg, 0.825 mmol) in DMF (5.0 mL) at 0 °C was added HATU (271.80 mg, 0.715 mmol). The mixture was warmed to room temperature and stirred for 4 h. The reaction was then quenched with H 2 O and extracted with EtOAc (2 x 10 mL). The combined organic layers were washed with brine (5 mL), dried over Na 2 SO 4 . filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (50% EtOAc/pet. ether) to afford the desired product (550 mg, 91.4% yield). LCMS (ESI) m/z. [M + H] calcd for CeiHgiNTOgSi: 1094.67; found 1094.5.

Step 2: Synthesis of tert-butyl ((2S)-1 -(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-

6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-1-oxopropan-2-yl)(methyl)carbamate

To a solution of tert-butyl ((2S)-1 -(((2S)-1 -(((6 3 S,4S)-1 '-ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3- yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-1-oxopropan-2-yl)(methyl)carbamate (540 mg, 0.493 mmol) in THF (5.0 mL) at 0 °C was added TBAF (1 M in THF, 0.59 mL, 0.592 mmol). The mixture was warmed to room temperature and stirred for 30 min. The reaction was quenched with H 2 O and was then extracted with EtOAc (2 x 10 mL). The combined organic layers were washed with brine (10 mL), dried over Na 2 SO 4 , filtered, After filtration, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (20% EtO Ac/pet. ether) to afford the desired product (320 mg, 69.1% yield). LCMS (ESI) m/z. [M + H] calcd for C52H71N7O9: 938.534; found 938.4.

Step 3: Synthesis of (2S)-N- ((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-3-methyl-2-((S)-N-methyl-2-(me thylamino)propanamido)butanamide To a solution of tert-butyl ((2S)-1-(((2S)-1-(((6 3 S,4S)-1 1 -ethyl-2 s -hydroxy-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-1-oxopropan-2-yl)(methyl)carbamate (300.0 mg, 0.320 mmol) in DCM (3.0 mL) at 0 °C and was added TFA (1.0 mL). The mixture was warmed to room temperature and stirred for 2 h. The mixture was concentrated under reduced pressure to afford the desired product (300 mg, crude). LCMS (ESI) m/z\ [M + H] calcd for C47H63N7O7: 838.49; found 838.4.

Intermediate 24. Synthesis of tert-butyl ((6 3 S,4S,2)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 e -hexahydro-1 1 H-8-oxa-2(4,2)- thiazola-1 (5,3)-indola-6(1 ,3)-pyrldazlnacycloundecaphane-4-yl)carbamate

Step 1: Synthesis of (S)-3-(4-bromothiazol-2-yl)-2-((tert-butoxycarbonyl)amino)pr opanoic acid To a solution of methyl (2S)-3-(4-bromo-1 ,3-thiazol-2-yl)-2-[(tert- butoxycarbonyl)amino]propanoate (110 g, 301.2 mmol) in THF (500 mL) and H 2 O (200 mL) at room temperature was added LiOH (21.64 g, 903.6 mmol). The resulting solution was stirred for 1 h and was then concentrated under reduced pressure. The resulting residue was adjusted to pH 6 with 1 M HCI and then extracted with DCM (3 x 500 mL). The combined organic layers were, dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (108 g, crude). LCMS (ESI) m/z: [M + H] calcd for CnHisBrN^S: 351.00; found 351.0.

Step 2: Synthesis of methyl (S)-1-((S)-3-(4-bromothiazol-2-yl)-2-((tert- butoxycarbonyl)amino)propanoyl)hexahydropyridazine-3-carboxy late

To a solution of (S)-3-(4-bromothiazol-2-yl)-2-((tert-butoxycarbonyl)amino)pr opanoic add (70 g,

199.3 mmol) in DCM (500 mL) at 0 °C was added methyl (3S)-1 ,2-diazinane-3-carboxylate bis(trifluoroacetic acid) salt (111.28 g, 298.96 mmol), NMM (219.12 mL. 1993.0 mmol), EDCI (76.41 g, 398.6 mmol) and HOBt (5.39 g, 39.89 mmol). The resulting solution was warmed to room temperature and stirred for 1 h. The reaction was then quenched with H 2 O (500 mL) and was extracted with EtOAc (3 x 500 mL). The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressured. The residue was purified by silica gel chromatography (0→50% EtOAc/pet. ether) to afford the desired product (88.1 g, 92.6% yield). LCMS (ESI) m/z: [M + H] calcd for Ci7H2sBrN40sS: 477.08; found 477.1.

Step 3: Synthesis of (S)-3-(1-ethyl-2-(2-(1-methoxyethyl)pyridin-3-yl)-5-(4,4,5,5 -tetramethyl-1 ,3,2- dioxaborolan-2-yl)-1 H- indol-3-yl)-2,2-dimethylpropan-1 -ol

To a solution of 3-(5-bromo-1-ethyl-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1 H- indol-3-yl)-2,2- dimethylpropan-1 -ol (60 g, 134.7 mmol) in toluene (500 mL) at room temperature was added bis(pinacolato)diboron (51.31 g, 202.1 mmol), Pd(dppf)Cl 2 (9.86 g, 13.48 mmol) and KOAc (26.44 g,

269.4 mmol). Then reaction mixture was then heated to 90 °C and stirred for 2 h. The reaction solution was then cooled to room temperature and concentrated under reduced pressure. Purification by silica gel chromatography (0→50% EtOAc/pet. ether) afforded the desired product (60.6 g, 94.0% yield). LCMS (ESI) m/z: [M + H] calcd for C29H41 BN 2 O4: 493.32; found 493.3.

Step 4: Synthesis of methyl (S)-1 -((S)-2-((tert-butoxycarbonyl)amino)-3-(4-(1 -ethyl-3-(3-hydroxy- 2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1 /findol-5-yl)thiazol-2- yl)propanoyl)hexahydropyridazine-3-carboxylate

To a solution of (S)-3-(1-ethyl-2-(2-(1-methoxyethyl)pyridin-3-yl)-5-(4,4,5,5 -tetramethyl-1 ,3,2- dioxaborolan-2-yl)-1 H- indol-3-yl)-2,2-dimethylpropan-1 -ol (30 g, 60.9 mmol) in toluene (600 mL), dioxane (200 mL), and H 2 O (200 mL) at room temperature was added methyl (S)-1-((S)-3-(4-bromothiazol-2-yl)-2- ((tert-butoxycarbonyl)amino)propanoyl)hexahydropyridazine-3- carboxylate (43.62 g, 91 ,4mmol), K3PO4 (32.23 g, 152.3 mmol) and Pd(dppf)Cl 2 (8.91 g, 12.18 mmol). The resulting solution was heated to 70 °C and stirred overnight. The reaction mixture was then cooled to room temperature and was quenched with H 2 O (200 mL). The resulting mixture was extracted with EtOAc (3 x 1000 mL) and the combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (0→90% EtOAc/pet. ether) to afford the desired product (39.7 g, 85.4% yield). LCMS (ESI) m/z: [M + H] calcd for C40H54N6O7S: 763.39; found 763.3.

Step 5: Synthesis of (S)-1-((S)-2-((tert-butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3 -hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1 -methoxyethyl)pyridin-3-yl)-1 H-indol-5-yl)thiazol-2- yl)propanoyl)hexahydropyridazine-3-carboxylic acid To a solution of methyl (S)-1-((S)-2-((tert-butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3 -hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1 H- indol-5-yl)thiazol-2- yl)propanoyl)hexahydropyridazine-3-carboxylate (39.7 g, 52.0 mmol) in THF (400 mL) and H 2 O (100 mL) at room temperature was added LiOHeH 2 O (3.74 g, 156.2 mmol). The resulting mixture was stirred for 1.5 h and was then concentrated under reduced pressure. The residue was acidified to pH 6 with 1 M HCI and extracted with DCM (3 x 1000 mL). The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (37.9 g, crude). LCMS (ESI) m/z: [M + H] calcd for CsgHsaNeOyS: 749.37; found 749.4.

Step 6: Synthesis of tert-butyl ((6 3 S,4S,2)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10- dimethyl-5, 7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 ® -hexahydro-1 1 H-8-oxa-2(4,2)-thiazola-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-4-yl)carbamate

To a solution of (S)-1-((S)-2-((tert-butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3 -hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1 H- indol-5-yl)thiazol-2- yl)propanoyl)hexahydropyridazine-3-carboxylic acid (37.9 g, 50.6 mmol ), HOBt (34.19 g, 253.0 mmol) and DIPEA (264.4 mL, 1518 mmol) in DCM (4 L) at 0 °C was added EDCI (271.63 g, 1416.9 mmol). The resulting mixture was warmed to room temperature and stirred overnight. The reaction mixture was then quenched with H 2 O and washed with 1 M HCI (4 x 1 L). The organic layer was separated and concentrated under reduced pressure. The residue was purified by silica gel chromatography (0→70% EtOAc/pet. ether) to afford the desired product (30 g, 81.1% yield). LCMS (ESI) m/z: [M + H] calcd for CsgHsoNeOeS: 731.36; found 731.3.

Intermediate 25. Synthesis of (6 3 $4S)-4-amino-1 1 -ethyl-2 9 -hydroxy-1 2 -(2-((S)-1- methoxyethyl)-5-(1 -methylplperldln-4-yl)pyridln-3-yl)-10,10-dimethyl-6 1 ,6 z ,6 3 ,6 4 ,6 s ,6 6 -hexahydro- 1 1 Wt-oxa-1 (5,3)-lndola-6(1 ,3)-pyridazlna-2(1 ,3)-benzenacyclou ndecaphane-5,7-dlone Step 1: Synthesis of benzyl (S)-5-bromo-6-(1-methoxyethyl)-3 , ,6 , -dihydro-[3,4'-bipyridine]-1 , (2 , H)- carboxylate

To a solution of (S)-3-bromo-5-iodo-2-(1-methoxyethyl)pyridine (6.0 g, 17.55 mmol) and benzyl 4- (4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-3,6-dihydropyridine-1 (2H) -carboxylate (7.23 g, 21.05 mmol) in dioxane (70 mL) and H 2 O (14 mL) was added K2CO3 (6.06 g, 43.86 mmol) and Pd(dppf)Cl 2 (1.28 g,

1.76 mmol). The reaction mixture was heated to 60 °C and stirred for 3 h. The mixture was diluted with H 2 O (50 mL) then extracted into EtOAc (3 x 100 mL). The combined organic layers were washed with brine (3 x 50 mL), dried over Na 2 SO 4 . filtered, and concentrated under reduced pressure. Purification by silica gel column chromatography (25% EtOAc/pet. ether) afforded the desired product (7.1 g, 94% yield). LCMS (ESI) m/z [M + H] calcd for CaiHaaBrNaOs: 431.10; found 431.1.

Step 2. Synthesis of tert-butyl ((6 3 S,4S)-2 s -(benzyloxy)-10,10-dimethyl-5,7-dioxo-1 2 -(4,4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate

To a solution of tert-butyl ((6 3 S,4S)-2 5 -(benzyloxy)-1 2 -iodo-10,10-dimethyl-5,7-dioxo- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-

4-yl)carbamate (5.0 g, 6.31 mmol), Pda(dba)3 (690 mg, 757 pmol), S-Phos (0.78 g, 1.89 mmol), and KOAc (2.17 g, 22.08 mmol) in toluene (75 mL) was added 4,4,5,5-tetramethyM ,3,2-dioxaborolane (5.65 g,

44.15 mmol). The reaction mixture was heated to 60 °C and stirred for 3 h. The reaction was quenched with H 2 O at 0 °C then extracted into EtOAc (3 x 100 mL). The combined organic layers were washed with brine (3 x 30 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by silica gel column chromatography (50% EtOAc/pet. ether) afforded the desired product (4.5 g, 90% yield). LCMS (ESI) m/z. [M + H] calcd for C75H57BN4O8: 793.43; found 793.4.

Step 3: Synthesis of benzyl 5-((6 3 S,4S)-2 5 -(benzyloxy)-4-((tert-butoxycarbonyl)amino)-10,10- dimethyl-5,7-dioxo-6 , ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-1 2 -yl)-6-((S)-1 -methoxyethyl)-3',6 , -dihydro-[3,4 , -bipyridine]-1 '(2'H) -carboxylate

To a solution of tert-butyl ((6 3 S,4S)-2 5 -(benzyloxy)-10,10-dimethyl-5,7-dioxo-1 2 -(4,4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 8 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (4.0 g, 5.05 mmol) and benzyl (S)-5-bromo- 6-(1-methoxyethyl)-3',6 , -dihydro-[3,4 , -bipyridine]-1 , (2 , H) -carboxylate (2.61 g, 6.06 mmol) in dioxane (50 mL) and H 2 O (10 mL) was added K2CO3 (1.74 g, 12.6 mmol) and Pd(dtbpf)Cla (330 mg, 505 pmol). The reaction mixture was heated to 70 °C. After 3 h the reaction was diluted with H 2 O (40 mL) and extracted into EtOAc (3 x 100 mL). The combined organic layers were washed with brine (2 x 50 mL), dried over Na 2 SO 4 . filtered, and concentrated under reduced pressure. Purification by silica gel column chromatography (50% EtOAc/pet. ether) afforded the desired product (4.1 g, 80% yield). LCMS (ESI) m/z. [M + H] calcd for CeoHeeNeOg: 1017.51 ; found 1017.4.

Step 4: Synthesis of benzyl 5-((6 3 S,4S)-2 5 -(benzyloxy)-4-((/erf-butoxycarbonyl)amino)-1 1 -ethyl- 10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-1 2 -yl)-6-((S)-1-methoxyethyl)-3 , ,6'-dihydro-[3,4 , -bipyridine]-1 , (2’H)-carboxylate To a solution of benzyl 5-((6 3 S,4S)-2 5 -(benzyloxy)-4-((tert-butoxycarbonyl)amino)-10,10-dime thyl- 5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 8 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-1 2 -yl)-6-((S)-1-methoxyethyl)-3 , ,6'-dihydro-[3,4 , -bipyridine]-1 , (2’H)-carboxylate (4.0 g, 3.93 mmol) and CS2CO3 (3.84 g, 11.80 mmol) in DMF (30 mL) at 0 °C was added iodoethane (2.45 g, 15.73 mmol). The reaction mixture was warmed to room temperature. After 3 h the reaction mixture was diluted with H 2 O (100 mL) and extracted into EtOAc (3 x 200 mL). The combined organic layers were washed with brine (3 x 100 mL), dried over Na 2 SO 4 . filtered, and concentrated under reduced pressure. Purification by silica gel column chromatography (66% EtOAc/pet. ether) afforded the desired product (1.4 g, 34% yield). LCMS (ESI) m/z. [M + H] calcd for CeaHyaNeOg: 1045.54; found 1045.5.

Step 5: Synthesis of tert-butyl ((6 3 S,4S)-1 1 -ethyl-2 s -hydroxy-1 2 -(2-((S)-1-methoxyethyl)-5-(1- methylpiperidin-4-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate

A solution of benzyl 5-((6 3 S,4S)-2 5 -(benzyloxy)-4-((tert-butoxycarbonyl)amino)-1 1 -ethyl-10,10- dimethyl-5, 7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6®-hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-1 2 -yl)-6-((S)-1-methoxyethyl)-3 , ,6 , -dihydro-[3,4'-bipyridine]-1 , (2 , H)-carboxylate (1.29 g, 1.23 mmol) and Pd/C (700 mg) in MeOH (30 mL) was stirred for 72 h at room temperature under Ha atmosphere. The reaction mixture was then filtered with MeOH (3 x 50 mL). The filtrate was concentrated under reduced pressure which afforded the desired product (850 mg, crude). LCMS (ESI) m/z. [M + H] calcd for CwHwNeO?: 837.49; found 837.7.

Step 6: Synthesis of (6 3 S,4S)-4-amino-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1-methoxyethyl)-5-(1- methy I pi pe r id i n -4-y l)py r id i n -3-y I)- 10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6®-hexahydro-1 ' H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-5,7-dione

To a solution of tert-butyl ((6 3 S,4S)-1 , -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1-methoxyethyl)-5-(1- methyl pi pe r id i n -4-y l)py r id i n -3-y I)- 10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6®-hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (840 mg, 1.00 mmol) in DCM (10 mL) at 0 °C was added TFA (3.0 mL, 40.4 mmol). The reaction mixture was warmed to room temperature. After 2 h the reaction was cooled to 0 °C, quenched with sat. at. NaHCOs, and extracted into EtOAc (3 x 50 mL). The combined organic layers were washed with brine (2 x 30 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure which afforded product (670 mg, crude). LCMS (ESI) m/z. [M + H] calcd for C43H56N6O5: 737.44; found 737.3.

Intermediate 26. Synthesis of (6 3 S, 4S)-4-amino-1 1 -ethy^2 5 -hydroxy-1 2 -(2-((S)-1- methoxyethyl)-5-(4-methylpiperazin-1 -yl)pyridine-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro- 1 1 M-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacyclou ndecaphane-5,7-dione

Step 1: Synthesis of ( S)-(5-bromo-6-(1 -methoxyethyl)pyridin-3-yl)boronic acid

To a solution of (S)-3-bromo-2-(1-methoxyethyl)pyridine (40 g, 185 mmol) and bis(pinacolato)diboron (70.5 g, 278 mmol) in THF (1.6 L) at 75 °C was added 4,4'-di-tert-butyl-2,2'- bipyridine (7.45 g, 27.7 mmol) and [lr(cod)CI]2 (1.24 mg, 1.85 mmol). After 16 h the mixture was concentrated under reduced pressure and the residue diluted with H 2 O (1 L). The aqueous layer extracted with DCM/MeOH (2 L, 5:1), dried with NazSO*, filtered, and concentrated under reduced pressure. Following purification by reverse phase chromatography (10→50% MeCN/H 2 O, 0.1% HCOaH) the combined product fractions were partially concentrated under reduced pressure. The aqueous layer was extracted with DCM/MeOH (3000 mL, 5:1), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (35.0 g, 65.5% yield). LCMS (ESI) m/z. [M + Na] calcd for CeHnBBrNOs: 282.00; found 281.1.

Step 2. Synthesis of (S)-3-bromo-5-iodo-2-(1-methoxyethyl)pyridine

To a solution of (S)-(5-bromo-6-(1-methoxyethyl)pyridin-3-yl)boronic acid (35.0 g, 135 mmol) in MeCN (100 mL) was added AFiodosuccinimide (60.6 g, 269 mmol). The resulting reaction mixture was stirred overnight and then concentrated under reduced pressure. Purification by normal phase chromatography (10% EtO Ac/pet. ether) afforded the desired product (40.0 g, 78.1% yield). LCMS (ESI) m/z. [M + H] calcd for CsHgBrlNO: 341.90; found 341.8.

Step 3: Synthesis of benzyl (S)-4-(5-bromo-6-(1 -methoxyethyl)pyridin-3-yl)piperazine-1 - carboxylate

To a solution of (S)-3-bromo-5-iodo-2-(1-methoxyethyl)pyridine (7.0 g, 20.5 mmol) and benzyl piperazine-1 -carboxylate (9.0 g, 40.8 mmol) in toluene (70 mL) were added Pda(dba)3 (375 mg, 0.409 mmol), Xantphos (1.18 g, 2.05 mmol) and sodium tert-butoxide (2.29 g, 24.6 mmol). The resulting mixture was heated to 120 °C and stirred for 16 h then cooled to room temperature and concentrated under reduced pressure. Purification by normal phase chromatography (25% EtOAc/pet. ether) afforded the desired product (5.0 g, 50.6% yield). LCMS (ESI) m/z. [M + H] calcd for C2oH24BrN3<D3: 434.11 ; found 434.0.

Step 4: Synthesis of benzyl 4-(5-((6 3 S,4S)-2 5 -(benzyloxy)-4-((tert-butoxycarbonyl)amino)-10,10- dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-1 2 -yl)-6-((S)-1 -methoxyethyl)pyridin-3-yl)piperazine-1 -carboxylate

To a solution of benzyl (S)-4-(5-bromo-6-(1-methoxyethyl)pyridin-3-yl)piperazine-1 -carboxylate (3.29 g, 7.56 mmol) and tert-butyl ((6 3 S,4S)-2 5 -(benzyloxy)-10,10-dimethyl-5,7-dioxo-1 2 -(4, 4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (50 g, 6.31 mmol) dioxane (40 mL) and H 2 O (10 mL) were added K2CO3 (1.74 g, 12.614 mmol) and Pd(dtbpf)Cl 2 (822 mg, 1.26 mmol) and the resulting mixture was heated to 80 °C for 2 h. The reaction mixture was then concentrated under reduced pressure and diluted with H 2 O (1 L). The aqueous layer was extracted with EtOAc (3 x 200 mL) and the combined organic layers were washed with H 2 O, dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by normal phase chromatography (50% EtOAc/pet. ether) afforded the desired product (5.0 g, 73.8% yield). LCMS (ESI) m/z. [M + H] calcd for C59H69N7O9: 1020.54; found 1020.6.

Step 5: Synthesis of benzyl 4-(5-((6 3 S, 4S)-2 5 -(benzyloxy) -4-((tert-butoxycarbonyl)amino)-1 ethyl-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-1 2 -yl)-6-((S)-1 -methoxyethyl)pyridin-3-yl)piperazine-1 -carboxylate To a stirred solution of benzyl 4-(5-((6 3 S,4S)-2 5 -(benzyloxy)-4-((tert-butoxycarbonyl)amino)- 10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-1 2 -yl)-6-((S)-1 -methoxyethyl)pyridin-3-yl)piperazine-1 -carboxylate (5.0 g, 5 mmol) in DMF (50 mL) at 0 °C was added CszCOs (3.19 g, 9.80 mmol) and ethyl iodide (1.53 g, 10 mmol). The resulting mixture was stirred for 2 h at room temperature and then diluted with H 2 O (200 mL). The aqueous layer was extracted with EtOAc (3 x 100 mL) and the combined organic layers were washed with H 2 O, dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by normal phase chromatography (33% EtO Ac/pet. ether) afforded the desired product (1.8 g, 35% yield). LCMS (ESI) m/z. [M + H] calcd for C61H73N7O9: 1048.56; found 1048.4.

Step 6: Synthesis of tert-butyl ((6 3 S, 4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 -methoxyethyl)-5- (piperazin-1 -yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-61 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate

To a stirred solution of benzyl 4-(5-((6 3 S, 4S)-2 5 -(benzyloxy) -4-((tert-butoxycarbonyl)amino)-1 1 - ethyl-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6®-hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-1 2 -yl)-6-((S)-1 -methoxyethyl)pyridin-3-yl)piperazine-1 -carboxylate (1.80 g, 1.72 mmol) in MeOH (20 mL) was added Pd/C (900 mg). The resulting mixture was stirred for 2 h at room temperature under a hydrogen atmosphere, filtered, and the filter cake washed with MeOH. The filtrate was concentrated under reduced pressure to afford the crude desired product which was used without further purification. LCMS (ESI) m/z. [M + H] calcd for CWHBINTO?: 824.47; found 824.3. Step 7\ Synthesis of tert-butyl((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 -methoxyethyl)-5-(4- methylpiperazin-1 -yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 e -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate

To a stirred solution of tert-butyl ((6 3 S, 4S)-1 '-ethyl-2 5 -hydroxy-1 2 -(2-((S)-1-methoxyethyl)-5- (piperazin-1 -yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-

6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate carbamate (590 mg, 0.716 mmol) and HCHO (129 mg, 1.43 mmol, 37 wt% in H 2 O) in MeOH (6 ml ) at 0 °C were added CH3COOH (122 mg, 2.02 mmol) and NaBHaCN (85.3 mg, 1.35 mmol). The resulting mixture was warmed to room temperature and stirred for 2 h. The reaction mixture was then concentrated under reduced pressure and diluted with H 2 O (100 mL). The aqueous layer was extracted with EtOAc (3 x 100 mL) and the combined organic layers were washed with H 2 O, dried with Na 2 SO 4 , filtered, concentrated under reduced pressure, pressure to afford the crude desired product which was used without further purification. LCMS (ESI) m/z. [M + H] calcd for C47H63N7O9: 838.49; found 838.4.

Step 8: Synthesis of (6 3 S, 4S)-4-amino-1 1 -ethyl-2 s -hydroxy-1 2 -(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1 -yl)pyridine-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-

6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-5,7-dione

To a stirred solution of tert-butyl((6 3 S,4S)-1 1 -ethyl-2 s -hydroxy-1 2 -(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1 -yl)pyridin-3-yl)-10,10-dimethyl-S^-dioxo-e 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (590 mg, 0.704 mmol) in DCM (6 mL) at 0 °C was added TEA (3.0 mL). The resulting mixture was stirred for 30 min and then concentrated under reduced pressure to afford the crude desired product which was used without further purification. LCMS (ESI) m/z. [M + H] calcd for C42H55N7O5: 738.44; found 738.4.

Intermediate 27. Synthesis of (6 3 $4S,2)-4-amlno-1 1 -ethyl-1 z -(2-((S)-1 -methoxyethyl)-5-(4- methylpiperazin-1 -yl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 e ,6 6 -hexahydro-1 1 W*-oxa-2(4,2)- thiazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-5,7-dione

.OTBDPS

.Br

MeO

N- cf. Brfrin* Pd(dppf)CI 2 M(dppf)CI 2 N- .Br toluene (HoxaiM/HsG

O H

Cte Cte O'

Cte'

Step 1: Synthesis of benzyl (S)-4-(5-bromo-6-(1 -methoxyethyl)pyridin-3-yl)piperazine-1 - carboxylate

Into a 3-L 3-necked round-bottom flask purged and maintained with an inert atmosphere of argon, was placed benzyl 4-[5-bromo-6-[(1 S)-1 -methoxyethyl]pyridin-3-yl]piperazine-1 -carboxylate (135 g, 310.821 mmol), bis(pinacolato)diboron (86.82 g, 341.903 mmol), Pd(dppf)Cl 2 (22.74 g, 31.082 mmol), KOAc (76.26 g, 777.052 mmol), and toluene (1 L). The resulting solution was stirred for 2 days at 90 °C in an oil bath. The reaction mixture was cooled to room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by neutral alumina column chromatography (25% EtOAc/hexanes) to afford the desired product (167 g, crude). LCMS (ESI) m/z. [M + H] calcd for CMHMBNSOS: 481.3; found 482.1.

Step 2. Synthesis of benzyl (S)-4-(5-(5-bromo-3-(3-((tert-butyldiphenylsilyl)oxy)-2,2- dimethylpropyl)-1 H-indol-2-yl)-6-(1 -methoxyethyl)pyridin-3-yl)piperazine-1 -carboxylate

Into a 3-L 3-necked round-bottom flask purged and maintained with an inert atmosphere of argon, was placed (S)-4-(6-(1-methoxyethyl)-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-3- yl)piperazine-1 -carboxylate (167 g, 346.905 mmol), 5-bromo-3-[3-[(tert-butyldiphenylsilyl)oxy]-2,2- dimethylpropyl]-2-iodo-1 H- indole (224.27 g, 346.905 mmol), Pd(dppf)Cl 2 (25.38 g, 34.69 mmol), dioxane (600 mL), H 2 O (200 mL), K3PO4 (184.09 g, 867.262 mmol), and toluene (200 mL). The resulting solution was stirred for overnight at 70 °C in an oil bath. The reaction mixture was cooled to room temperature after reaction completed. The resulting mixture was concentrated under reduced pressure. The residue was purified by normal phase column chromatography (50% EtOAc/hexanes) to afford the desired product (146 g, 48.2% yield). LCMS (ESI) m/z. [M + H] calcd for C49Hs7BrN404Si: 872.3; found 873.3.

Step 3: Synthesis of benzyl (S)-4-(5-(5-bromo-3-(3-((tert-butyldiphenylsilyl)oxy)-2,2- dimethylpropyl)-1 -ethyl-1 H-indol-2-yl)-6-(1 -methoxyethyl)pyridin-3-yl)piperazine-1 -carboxylate To a stirred mixture of benzyl (S)-4-(5-(5-bromo-3-(3-((tert-butyldiphenylsilyl)oxy)-2,2- dimethylpropyl)-1 H- indol-2-yl)-6-(1 -methoxyethyl)pyridin-3-yl)piperazine-1 -carboxylate (146 g, 167.047 mmol) and CS2CO3 (163.28 g, 501.14 mmol) in DMF (1200 mL) was added C2HSI (52.11 g, 334.093 mmol) in portions at 0 °C under N 2 atmosphere. The final reaction mixture was stirred at room temperature for 12h. The resulting mixture was diluted with EtOAc (1 L) and washed with brine (3 x 1.5L). The organic layers were dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure to afford the desired product (143 g, crude). LCMS (ESI) m/z. [M + H] calcd for CsiH 6 iBrN404Si: 900.4; found 901.4.

Step 4: Synthesis of benzyl (S)-4-(5-(5-bromo-1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-1 H-indol- 2-yl)-6-(1 -methoxyethyl)pyridin-3-yl)piperazine-1 -carboxylate

To a stirred mixture of benzyl (S)-4-(5-(5-bromo-3-(3-((fe/t-butyldiphenylsilyl)oxy)-2,2- dimethylpropyl)-1 -ethyl-1 H- indol-2-yl)-6-(1 -methoxyethyl)pyridin-3-yl)piperazine-1 -carboxylate (143 g,

158.526 mmol) in DMF (1250 mL) was added CsF (72.24 g, 475.578 mmol). The reaction mixture was stirred at 60 °C for 2 days under N 2 atmosphere. The resulting mixture was diluted with EtOAc (1 L) and washed with brine (3 x 1 L). The organic phase was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to afford two atropisomers of benzyl (S)-4-(5-(5- bromo-1 -ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-1 H-indol-2-yl)-6-(1 -methoxyethyl)pyridin-3-yl)piperazine- 1 -carboxylate A (38 g, 36% yield, RT = 1.677 min in 3 min LCMS(0.1% FA)) and B (34 g, 34% yield, RT = 1.578 min in 3 min LCMS(0.1% FA)). LCMS (ESI) m/z. [M + H] calcd for C3sH43BrN404: 663.2; found 662.2.

Step 5: Synthesis of benzyl (S)-4-(5-(1 -ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-5-(4, 4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)-1 H- indol-2-yl)-6-(1 -methoxyethyl)pyridin-3-yl)piperazine-1 - carboxylate

Into a 500-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed benzyl (S)-4-(5-(5-bromo-1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-1 /+indol-2-yl)-6- (1 -methoxyethyl)pyridin-3-yl)piperazine-1 -carboxylate A (14 g, 21.095 mmol), bis(pinacolato)diboron (5.89 g, 23.205 mmol), Pd(dppf)Cl 2 (1.54 g, 2.11 mmol), KOAc (5.18 g, 52.738 mmol), and toluene (150 mL). The resulting solution was stirred for 5 h at 90 °C in an oil bath. The reaction mixture was then cooled to room temperature. The resulting mixture was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to afford the desired product (12 g, 76.0% yield). LCMS (ESI) m/z. [M + H] calcd for CAIHSSBN^: 710.4; found 711.3.

Step 6: Synthesis of methyl (S)-1-((S)-3-(4-(2-(5-(4-((benzyloxy)carbonyl)piperazin-1-yl )-2-((S)-1- methoxyethyl)pyridin-3-yl)-1-ethyl-3-(3-hydroxy-2,2-dimethyl propyl)-1H-indol-5-yl)thiazol-2-yl)-2-((te/1- butoxycarbonyl)amino)propanoyl)hexahydropyridazine-3-carboxy late Into a 250-mL round-bottom flask purged and maintained with an inert atmosphere of argon, was placed benzyl (S)-4-(5-(1 -ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-5-(4,4,5,5-tetrameth yl-1 ,3,2- dioxaborolan-2-yl)-1 H- indol-2-yl)-6-(1 -methoxyethyl)pyridin-3-yl)piperazine-1 -carboxylate (10.8 g, 15.196 mmol), methyl (3S)-1 -[(2S)-3-(4-bromo-1 ,3-thiazol-2-yl)-2-[(tert-butoxycarbonyl)amino]propanoyl]-1 ,2- diazinane-3-carboxylate (7.98 g, 16.716 mmol), Pd(dtbpf)Cl 2 (0.99 g, 1.52 mmol), K3PO4 (8.06 g, 37.99 mmol), toluene (60 mL), dioxane (20 mL), and H 2 O (20 mL). The resulting solution was stirred for 3 h at 70 °C in an oil bath. The reaction mixture was then cooled to room temperature. The resulting solution was extracted with EtOAc (2 x 50 mL) and concentrated under reduced pressure. The residue was purified by normal phase column chromatography to afford the desired product (8 g, 50.9% yield). LCMS (ESI) m/z : [M + H] calcd for CHHeeNeOgS: 980.5; found 980.9.

Step T. Synthesis of (S)-1-((S)-3-(4-(2-(5-(4-((benzyloxy)carbonyl)piperazin-1-yl )-2-((S)-1- methoxyethyl)pyridin-3-yl)-1-ethyl-3-(3-hydroxy-2,2-dimethyl propyl)-1 H- indol-5-yl)thiazol-2-yl)-2-((tert- butoxycarbonyl)amino)propanoyl)hexahydropyridazine-3-carboxy lic acid

To a stirred mixture of methyl (S)-1-((S)-3-(4-(2-(5-(4-((benzyloxy)carbonyl)piperazin-1-yl )-2-((S)- 1 -methoxyethyl)pyridin-3-yl)-1 -ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-1 H- indol-5-yl)thiazol-2-yl)-2-((tert- butoxycarbonyl)amino)propanoyl)hexahydropyridazine-3-carboxy late (12 g, 12.230 mmol) in THF (100 mL) and H 2 O (100 mL) was added LiOH (2.45 g, 61.148 mmol) under N 2 atmosphere and the resulting mixture was stirred for 2 h at room temperature. The resulting mixture was concentrated under reduced pressure and the pH of aqueous phase was acidified to 5 with HCI (1 N) at 0 °C. The aqueous layer was extracted with DCM (3 x 100 mL). The organic phase was concentrated under reduced pressure to afford the desired product (10 g, 84.5% yield). LCMS (ESI) m/z. [M + H] calcd for CsiHeeNeOgS: 966.5; found 967.0.

Step 8: Synthesis of benzyl 4-(5-((6 3 S,4S,Z)-4-((tert-butoxycarbonyl)amino)-1 ’-ethyl-10,10- dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-thiazola-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-1 2 -yl)-6-((S)-1 -methoxyethyl)pyridin-3-yl)piperazine-1 -carboxylate

Into a 3-L round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed (S)-1 -((S)-3-(4-(2-(5-(4-((benzyloxy)carbonyl)piperazin-1 -yl)-2-((S)-1 -methoxyethyl)pyridin-3-yl)-1 - ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-1 H- indol-5-yl)thiazol-2-yl)-2-((tert- butoxycarbonyl)amino)propanoyl)hexahydropyridazine-3-carboxy lic acid (18 g, 18.61 mmol), MeCN (1.8 L), DIPEA (96.21 g, 744.417 mmol), EDCI (107.03 g, 558.313 mmol), HOST (25.15 g, 186.104 mmol).

The resulting solution was stirred for overnight at room temperature. The resulting mixture was concentrated under reduced pressure after reaction completed. The resulting solution was diluted with DCM (1 L) and was washed with HCI (3 x 1 L, 1 N aqueous). The resulting mixture was washed with H 2 O (3 x 1 L) and then the organic layer was concentrated. The residue was purified by normal phase column chromatography (50% EtOAc/hexanes) to afford the desired product (10.4 g, 54.9% yield). LCMS (ESI) m/z. [M + H] calcd for CsiH&tNeOeS: 948.5; found 949.3.

Step 9: Synthesis of tert-butyl ((6 3 S,4S,Z)-1 f-ethyl-1 2 -(2-((S)-1 -methoxyethyl)-5-(piperazin-1 - yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-2(4,2)-thiazola-1 (5,3)- indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)carbamate

Into a 250-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed benzyl 4-(5-((6 3 S,4S,Z)-4-((tert-butoxycarbonyl)amino)-1 1 -ethyl-10,10-dimethyl-5,7-dioxo- 6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-2(4,2)-thiazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane- 1 2 -yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-c arboxylate (10.40 g, 10.957 mmol), Pd(0H)2/C (5 g, 46.984 mmol), and MeOH (100 mL). The resulting solution was stirred for 3 h at room temperature under a 2 atm Ha atmosphere. The solids were filtered out and the filter cake was washed with MeOH (3 x 100 mL). The combined organic phase was concentrated under reduced pressure to afford the desired product (8.5 g, 90.4% yield). LCMS (ESI) m/z: [M + H] calcd for CAsHseNeOeS: 814.4; found 815.3.

Step 10: Synthesis of tert-butyl ((6 3 S,4S,Z)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1 -yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-2(4,2)- thiazola-1(5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)carbamate

Into a 1000-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed tert-butyl ((6 3 S,4S,Z)-1 /-ethyl-1 2 -(2-((S)-1 -methoxyethyl)-5-(piperazin-1 -yl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-2(4,2)-thiazola-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-4-yl)carbamate (8.5 g, 10.429 mmol), MeOH (100 mL), AcOH (1.88 g,

31.286 mmol) and stirred for 15 min. Then HCHO (1 .88 g, 23.15 mmol, 37% aqueous solution) and NaBHsCN (788 mg, 12.5 mmol) was added at room temperature. The resulting solution was stirred for 3 hr. The resulting mixture was quenched with H 2 O (100 mL) and concentrated under reduced pressure to remove MeOH. The resulting solution was diluted with DCM (300 mL) and was washed with HzO (3 x 100 mL). The organic phase was concentrated under reduced pressure to afford the desired product (8.2 g, 90.1% yield). LCMS (ESI) m/z: [M + H] calcd for GwHeoNeOeS: 828.4; found 829.3.

Step 10: Synthesis of (6 3 S,4S,2)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1 -yl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 /+8-oxa-2(4,2)-thiazola- 1 (5,3)-indola-6(1 ,3)-pyridazinacydoundecaphane-5,7-dione

Into a 250-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed tert-butyl ((6 3 S,4S,Z)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)-5-(4-methylpiperazin-1 -yl)pyridin-3- yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-thiazola-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-4-yl)carbamate (8.20 g, 9.891 mmol) and dioxane (40 mL), followed by the addition of HCI in 1 ,4-dioxane (4M, 40 mL) at 0 °C. The resulting solution was stirred for 1 h at 0 °C. The mixture was then concentrated under reduced pressure. The resulting solution was diluted with DCM (600 mL) and sat. aq. NaHCOa (400 mL). The organic phase was then washed twice with brine (500 mL). The organic phase was concentrated under reduced pressure to afford the desired product (7.2 g, 94.9% yield).

Intermediate 28. Synthesis of (6 3 S,4S)-4-amino-2 s -(difluoromethyl)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyF6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-5,7-dione

Step 1: Synthesis of methyl (S)-3-(3-bromo-5-(difluoromethyl)phenyl)-2-((tert- butoxycarbonyl)amino)propanoate

Into a 1000 mL 3-necked round-bottom flask was added Zn powder (43.42 g, 663.835 mmol) and I2 (1.30 g, 5.106 mmol) in DMF (400 mL) at room temperature. To the above mixture was added a solution of methyl (2fl)-2-[(tert-butoxycarbonyl)amino]-3-iodopropanoate (36.42 g, 110.64 mmol) in DMF (10 mL). The mixture was heated to 30 °C for 10 min. To the mixture was then added a solution of methyl

(2fl)-2-[(tert-butoxycarbonyl)amino]-3-iodopropanoate (72.83 g, 221.28 mmol) in DMF (20 mL) dropwise at room temperature. The resulting mixture was stirred for 30 min. The resulting mixture was filtered and the solution was added to a mixture of 1-bromo-3-(difluoromethyl)-5-iodobenzene (85.0 g, 255.321 mmol), tris(furan-2-yl) phosphane (3.56 g, 15.319 mmol), and Pd2(dba)s (4.68 g, 5.106 mmol) in DMF (400 mL) at room temperature under argon atmosphere. The reaction mixture was heated to 60 °C for 10 min and was then removed from the oil bath and was stirred for 1 h until the temperature of the resulting mixture cooled down to 50 °C. The reaction was quenched with aq. NhLCI (3000 mL) and the aqueous layer was extracted with EtOAc (3 x 1000 mL). The combined organic layers were washed with brine (2 x 1000 mL), dried over anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (9% EtOAc/pet. ether) to afford the desired product (59 g, 56.6% yield). Step 2. Synthesis of methyl (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(difluoromethyl)-5-( 1 -ethyl-3- (3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyri din-3-yl)-1 H-indol-5-yl)phenyl)propanoate A mixture of methyl (2S)-3-[3-bromo-5-(difluoromethyl)phenyl]-2-[(tert-butoxycar bonyl)amino] propanoate (90.0 g, 220.459 mmol), (S)-3-(1-ethyl-2-(2-(1-methoxyethyl)pyridin-3-yl)-5-(4, 4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)-1 H- indol-3-yl)-2,2-dimethylpropan-1 -ol (1.50 g, 3.046 mmol),

Pd(dppf)CI 2 (16.13 g, 22.046 mmol) and K3PO4 (116.99 g, 551.148 mmol) in dioxane (600 mL), H 2 O (200 mL), and toluene(200 mL) was stirred for 2 h at 70 °C. The resulting mixture was concentrated under reduced pressure and then diluted with H 2 O (300 mL). The mixture was extracted with EtOAc (3 x 500 mL). The combined organic layers were washed with H 2 O (3 x 500 mL), dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography (50% EtOAc/pet. ether) to afford the desired product (128 g, 83.7% yield). LCMS (ESI) m/z. [M + H] calcd for C39H49F2N3O6: 694.37; found 694.2.

Step 3: Synthesis of (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(difluoromethyl)-5-( 1-ethyl-3-(3- hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin -3-yl)-1 H- indol-5-yl)phenyl)propanoic acid To a stirred solution of methyl (S)-2-((fe/t-butoxycarbonyl)amino)-3-(3-(difluoromethyl)-5-( 1 -ethyl- 3-(3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)py ridin-3-yl)-1 H- indol-5-yl)phenyl)propanoate (125.0 g, 180.159 mmol) in THF (800 mL) was added LiOHeH 2 O (11.48 g, 479.403 mmol) in H 2 O (200 mL) dropwise at 0 °C. The resulting mixture was stirred for 2 h at 0 °C. The mixture was acidified to pH 6 with 1 M HCI (aq.) and was then extracted with EtOAc (3 x 800 mL). The combined organic layers were washed with brine (2 x 200 mL), dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure to afford the desired product (125 g, crude). LCMS (ESI) m/z. [M + H] calcd for C38H47F2N3O6: 680.37; found 680.2.

Step 4: Synthesis of methyl (S)-1-((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(difluorometh yl)-5-(1- ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyet hyl)pyridin-3-yl)-1 H- indol-5- yl)phenyl)propanoyl)hexahydropyridazine-3-carboxylate

To a stirred solution of methyl (3S)-1 ,2-diazinane-3-carboxylate (39.77 g, 275.814 mmol) and NMM (185.98 g, 1838.760 mmol) in DCM (1500 mL) was (S)-2-((tert-butoxycarbonyl)amino)-3-(3- (difiuoromethyl)-5-(1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl) -2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1 H- indol-5-yl)phenyl)propanoic acid (125.0 g, 183.876 mmol), HOBt (12.42 g, 91.938 mmol) and EDCI (70.50 g, 367.752 mmol) in portions at 0 °C. The resulting mixture was stirred at room temperature for 16 h. The reaction mixture was then washed with 0.5 M HCI (2 x 1000 mL) and brine (2 x 800 mL), dried over anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (50% EtOAc/pet.ether) to afford the desired product (110 g, 74.2% yield). LCMS (ESI) m/z. (M + H] calcd for C44H57F2N5O7: 806.43; found 806.2.

Step 5: Synthesis of (S)-1-((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(difluorometh yl)-5-(1-ethyl-3- (3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyri din-3-yl)-1 H-indol-5- yl)phenyl)propanoyl)hexahydropyridazine-3-carboxylic acid

To a stirred solution of methyl (S)-1-((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(difluorometh yl)-5- (1 -ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1 -methoxyethyl)pyridin-3-yl)-1 H-indol-5- yl)phenyl)propanoyl)hexahydropyridazine-3-carboxylate (110.0 g, 136.482 mmol) in THF (800 mL) was added a solution of Li0H*H20 (17.18 g, 409.446 mmol) in H 2 O (200 mL) in portions at 0 °C. The resulting mixture was stirred for 2 h at 0 °C and was then neutralized to pH 6 with 0.5 M HCI. The resulting mixture was extracted with EtOAc (3 x 800 mL) and the combined organic layers were washed with brine (2 x 600 mL), dried over anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (100 g, crude). LCMS (ESI) m/z: [M + H] calcd for C43H55F2N5O7: 792.42; found 792.4. Step 6: Synthesis of tert-butyl ((6 3 S,4S)-2 5 -(difluoromethyl)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-

6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate

To a stirred solution of (S)-1 -((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(difluoromethyl)-5 -(1 -ethyl- 3-(3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)py ridin-3-yl)-1 H- indol-5- yl)phenyl)propanoyl)hexahydropyridazine-3-carboxylic acid (100.0 g, 126.273 mmol) in DCM (6000 mL) was added DIPEA (163.20 g, 1262.730 mmol), HOBt (85.31 g, 631 .365 mmol), and EDCI (363.10 g, 1894.095 mmol) dropwise at 0 °C. The resulting mixture was stirred overnight at room temperature. The mixture was then washed with 0.5 M HCI (2 x2000 mL) and brine (2 x 2000 mL), dried over anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (50% EtOAc/pet. ether) to afford the desired product (70 g, 71.6% yield). LCMS (ESI) m/z: [M + H] calcd for C43H53F2N5O6: 774.41 ; found 774.0.

Step 7: Synthesis of ^S^SH-amino^-idifluoromethyO-l'-ethyl-l^-^S)-!- methoxyethyl)pyridin-3-yl)-10,1 O-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-5,7-dione

To a stirred solution of tert-butyl ((6 3 S,4S)-2 5 -(difluoromethyl)-1 '-ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-

6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (202.0 mg, 0.261 mmol) in DCM (2 mL) was added TFA (1.0 mL) dropwise at 0 °C. The resulting mixture was stirred for 1.5 h at 0 °C and was then concentrated under reduced pressure to afford the desired product. LCMS (ESI) m/z. [M + H] calcd for CseH+sFaNsO^ 674.35; found 674.5.

Intermediate 29. Synthesis of (6 3 S,4S)-4-amino-1 1 -ethyl-2 5 -(fluoromethyl)-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyF6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-5,7-dione

Step 1: Synthesis of 1-bromo-3-(fluoromethyl)-5-iodobenzene

To a solution of (3-bromo-5-iodophenyl)methanol (175.0 g, 559.227 mmol) in DCM (2 L) was added BAST (247.45 g, 1118.454 mmol) dropwise at 0 °C. The resulting mixture was stirred for 16 h at room temperature. The reaction was quenched with sat. aq. NaHCOa at 0 °C. The organic layers were washed with H 2 O (3 x 700 mL) and dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (3% EtOAc/pet. ether) to afford the desired product (120 g, 68% yield).

Step 2. Synthesis of methyl (2S)-3-[3-bromo-5-(fluoromethyl)phenyl]-2-[(tert- butoxycarbonyl)amino]propanoate

Into a 1000 mL 3-necked round-bottom flask was added Zn powder (32.40 g, 495.358 mmol) in DMF (350.0 mL) and la (967.12 mg, 3.810 mmol). To the mixture was added a solution of methyl (2fl)-2- [(tert-butoxycarbonyl)amino]-3-iodopropanoate (27.0 g, 82.03 mmol) in DMF (10 mL). The mixture was heated to 30 °C for 10 min. To the mixture was then added a solution of methyl (2R)-2-[(tert- butoxycarbonyl)amino]-3-iodopropanoate (54.0 g, 164.07 mmol) in DMF (20 mL). The resulting mixture was stirred for 30 min at room temperature and was filtered. The resulting solution was added to a mixture of 1 -bromo-3-(fluoromethyl)-5-iodobenzene (60 g, 190.522 mmol), tris(furan-2-yl)phosphane (2.65 g, 11.431 mmol), and Pda(dba)3 (3.49 g, 3.810 mmol) in DMF (400 mL) at room temperature under argon atmosphere and the reaction mixture was heated to 60 °C for 10 min then removed the oil bath. The resulting mixture was stirred for about 1 h until the temperature cooled down to 50 °C. The reaction was quenched with aq. NhUCI (3000 mL) and the resulting mixture was extracted with EtOAc (3 x 1000 mL). The combined organic layers were washed with brine (2x 1000 mL) and dried over anhydrous NazSO*. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (9% EtOAc/pet. ether) to afford the desired product (45 g, 60% yield).

Step 3: Synthesis of methyl (S)-2-((te/†-butoxycarbonyl)amino)-3-(3-(1 -ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin -3-yl)-1H-indol-5-yl)-5-(fluoromethyl)phenyl)propanoate A mixture of methyl (2S)-3-[3-bromo-5-(fluoromethyl)phenyl]-2-[(tert- butoxycarbonyl)amino]propanoate (75.28 g, 192.905 mmol), (S)-3-(1-ethyl-2-(2-(1-methoxyethyl)pyridin- 3-yl)-5-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)-1 H-indol-3-yl)-2,2-dimethylpropan-1-ol (95 g,

192.905 mmol), Pd(dppf)Cl 2 (14.11 g, 19.291 mmol) and K2CO3 (53.32 g, 385.810 mmol) in dioxane (900 mL) and H 2 O (180 mL) was stirred for 2 h at 80 °C. The resulting mixture was concentrated under reduced pressure and was then diluted with H 2 O. The resulting mixture was extracted with EtOAc (3 x 1200 mL) and the combined organic layers were washed with H 2 O (3 x 500 mL) and dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (50% EtOAc/pet. ether) to afford the desired product (105 g, 80% yield). LCMS (ESI) m/z. [M + H] calcd for C39H50FN3O6: 676.38; found 676.1.

Step 4: Synthesis of (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1 -ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1 H- indol-5-yl)-5-(fluoromethyl)phenyl)propanoic acid

To a stirred solution of methyl (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1 -ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin -3-yl)-1H-indol-5-yl)-5-(fluoromethyl)phenyl)propanoate (108 g, 159.801 mmol) in THF (500 mL) was added a solution of LIOHeHzO (11.48 g, 479.403 mmol) in H 2 O (500 mL) at 0 °C. The resulting mixture was stirred for 2 h at 0 °C and was then acidified to pH 6 with 1 M HCI (aq.). The mixture was extracted with EtOAc (3 x 800 mL) and the combined organic layers were washed with brine (2x 200 mL) and dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure to afford the desired product (101 g, crude). LCMS (ESI) m/z. [M + H] calcd for CaeH^FNsOe: 662.36; found 662.1.

Step 5: Synthesis of methyl (S)-1 -((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1 -ethyl-3-(3-hydroxy- 2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1 W-indol-5-yl)-5- (f!uoromethyl)phenyl)propanoyl)hexahydropyridazine-3-carboxy late

To a stirred solution of (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1 -ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1 H-indol-5-yl)-5-(fluoromethyl)phenyl)propanoic acid (103 g, 155.633 mmol) and NMM (157.42 g, 1556.330 mmol) in DCM (1200 mL) was added methyl (3S)-1 ,2- diazinane-3-carboxylate (33.66 g, 233.449 mmol), HOBt (10.51 g, 77.816 mmol) and EDCI (59.67 g, 311.265 mmol) in portions at 0 °C. The resulting mixture was stirred a t room temperature for 16 h. The organic layers were then washed with 0.5 M HCI (2 x 1000 mL) and brine (2 x 800 mL), dried over anhydrous NaaSOA, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (50% EtOAc/pet. ether) to afford the desired product (103 g, 83% yield). LCMS (ESI) m/z. [M + H] calcd for C44HseFNs07: 788.44; found 788.1.

Step 6: Synthesis of (S)-1-((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3 -hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1 H-indol-5-yl)-5- (fluoromethyl)phenyl)propanoyl)hexahydropyridazine-3-carboxy lic acid

To a stirred solution of methyl (S)-1-((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3 - hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin -3-yl)-1 W-indol-5-yl)-5- (fluoromethyl)phenyl)propanoyl)hexahydropyridazine-3-carboxy late (103 g, 130.715 mmol) in THF (700 mL) was added a solution of LiOHehfeO (27.43 g, 653.575 mmol) in hfeO (700 mL) at 0 °C .The resulting mixture was stirred for 2 h at 0 °C and was then neutralized to pH 6 with 1 M HCI.The resulting mixture was extracted with EtOAc (3 x 800 mL) and the combined organic layers were washed with brine (2 x 600 mL), dried over anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (101 g, crude). LCMS (ESI) m/z. [M + H] calcd for CwHseFNsO: 774.43; found 774.1.

Step 7\ Synthesis of tert-butyl ((6 3 S,4S)-1 1 -ethyl-2 5 -(fluoromethyl)-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl) -10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate

To a stirred solution of (S)-1-((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3 -hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1 FFindol-5-yl)-5-

(fluoromethyl)phenyl)propanoyl)hexahydropyridazine-3-carb oxylic acid (101 g, 130.50 mmol) in DCM (5500 mL) was added DIPEA (227.31 mL, 1305.0 mmol) and HOBt (88.17 g, 652.499 mmol), and EDCI (375.26 g, 1957.498 mmol) at 0 °C. The resulting mixture was stirred at room temperature overnight. The mixture was then washed with 0.5 M HCI (2 x 2000 mL), brine (2 x 2000 mL), dried over anhydrous

NapSO*. filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (50% EtOAc/pet. ether) to afford the desired product (68 g, 65% yield). LCMS (ESI) m/z. [M + H] calcd for C43H54FN5O6: 756.42; found 756.4.

Step 8: Synthesis of (2S)-N- ((6 3 S,4S)-1 , -ethyl-2 5 -(fluoromethyl)-1 2 -(2-((S)-1-methoxyethyl)pyridin- 3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 6 5 ,6®-hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-

2(1 ,3)-benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)bu tanamide

To a stirred solution of tert-butyl ((6 3 S,4S)-1 '-ethyl-2 5 -(fluoromethyl)-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl) -10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (350 mg, 0.403 mmol) in DCM (4 mL) was added TFA (1.50 mL) at 0 °C. The resulting mixture was stirred at room temperature for 1 .5 h and was then concentrated under reduced pressure to afford the desired product (600 mg, crude). LCMS (ESI) m/z. [M + H] calcd for C38H46FN5O4: 656.36; found 656.4.

Intermediate A-1. Synthesis of N-methyl-N-((S)-1-((fl)-1-trltylazlrldlne-2- carbonyl)pyrrolldlne-3-carbonyl)-Z.-valine

Step 1: Synthesis of methyl A/-methyl-N-((S)-1 -((fl)-1 -tritylaziridine-2-carbonyl)pyrrolidine-3- carbonyl)-L-valinate

To a mixture of methyl N-methyl-N-((S)-pyrrolidine-3-carbonyl)-L-valinate (0.840 g, 3.47 mmol) and (fl)-1 -tritylaziridine-2-carboxylic acid (1.713 g, 5.2 mmol) in DMF (20 mL) at 0 °C was added DIPEA (3.0 mL, 17.33 mmol) and HATU (2.636 g, 6.93 mmol). The reaction mixture was stirred for 3 h, at which point the mixture was extracted with EtOAc (200 mL). The EtOAc layer was washed with brine (3 x 50 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The crude residue was purified by reverse phase chromatography (10→50% MeCN/H20) to afford the desired product (1.02 g, 53% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for C34H39N3O4: 554.30; found 554.3.

Step 2. Synthesis of N-methyl-N-((S)-1-((fl)-1-tritylaziridine-2-carbonyl)pyrroli dine-3-carbonyl)-L- valine

To a solution of methyl N-methyl-N-((S)-1-((fl)-1-tritylaziridine-2-carbonyl)pyrroli dine-3-carbonyl)- L-valinate (1.0 g, 1.81 mmol) in THF (10 mL) at 0 °C was added a solution of LiOHeH 2 O (0.3789 g,

9.03 mmol) in H 2 O (9.0 mL). After 3 h, the reaction solution was neutralized to pH 7 with sat. aq. NH4CI. The resulting mixture was extracted with EtOAc (3 x 50 mL) and the combined organic layers were washed with brine (3 x 20 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the crude product (740 mg, 75.9% yield) as a solid. LCMS (ESI) m/z: [M - H] calcd for C33H37N3O4: 538.27; found 538.2.

Intermediate A-2. Synthesis of N-methyl- jV-((S)-1 -((S)-1 -trltylazlrkflne-2- carbonyl)pyrrolldine-3-carbonyl)-L-valine

Step 1: Synthesis of methyl W-methyl-W-((S)-1 -((S)-1 -tritylaziridine-2-carbonyl)pyrrolidine-3- carbonyl)-L-valinate

To a mixture of methyl N-methyl-N-((S)-pyrrolidine-3-carbonyl)-Z.-valinate (0.800 g, 3.30 mmol) and (S)-1 -tritylaziridine-2-carboxylic acid (1.305 g, 3.96 mmol) in DMF (16 mL) at 0 °C was added DIPEA (2.9 mL, 16.5 mmol) and HATU (1.88 g, 4.9 mmol). The reaction mixture was warmed to room temperature and stirred for 1 h, at which point the mixture was diluted with EtOAc. The mixture was washed with sat. NH4CI and the resulting aqueous layer extracted with EtOAc. The combined organic layers were washed with brine, dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The crude residue was purified by reverse phase chromatography (10→80% MeCN/H 2 O) to afford the desired product (1.17 g, 64% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for C34H39N3O4: 554.30; found 554.3.

Step 2. Synthesis of N-methyl-N-((S)-1-((S)-1-tritylaziridine-2-carbonyl)pyrrolid ine-3-carbonyl)-L- valine

To a stirred solution of methyl N-methyl-N-((S)-1 -((S)-1 -tritylaziridine-2-carbonyl)pyrrolidine-3- carbonyl)-L-valinate (1.10 g, 1.99 mmol) in THF (10.0 mL) at 0 °C was added a 1 M solution of LiOH (9.93 mL, 9.93 mmol). The reaction mixture was warmed to room temperature and stirred for 16 h. The reaction mixture was cooled to 0 °C and quenched with sat. aq. NI-LCI to pH 6. The resulting mixture was extracted with EtOAc. The combined organic layers were washed with brine, dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired crude product (1.2 g). LCMS (ESI) m/z: [M + H] calcd for C33H37N3O4: 540.29; found 540.3. Intermediate A-3. Synthesis of W-methyl-N-(1-((fl)-1-tritylaziridine-2-carbonyl)piperidine- 4- carbonyl)-L-valine

Step 1: Synthesis of methyl N-methyl-N-(1 -((fl)-1 -tritylaziridine-2-carbonyl)piperidine-4-carbonyl)-

L-valinate

To a solution of (fl)-1 -tritylaziridine-2-carboxylic acid (1 .157 g, 3.51 mmol) and methyl N-methyl- N-(piperidine-4-carbonyl)-L-valinate (0.600 g, 2.34 mmol) in DMF (20 mL) at 0 °C was added DIPEA (0.204 mL, 11.70 mmol) and HATU (1.780 g, 4.68 mmol. After 3 h, the reaction mixture was extracted with EtOAc (200 mL). The combined organic layers were washed with brine (3 x 50 mL), dried over NazS04, filtered, and concentrated under reduced pressure. The residue was purified by reverse phase chromatography (10→50% MeCN/HzO) to afford the desired product (740 mg, 55.7% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for C35H41N3O4: 568.32; found 568.3.

Step 2. Synthesis of N-methyl-N-(1-((fl)-1-tritylaziridine-2-carbonyl)piperidine- 4-carbonyl)-/--valine To a solution of methyl N-methyl-N-(1 -((fl)-1 -tritylaziridine-2-carbonyl)piperidine-4-carbonyl)-L- valinate (0.700 g, 1.23 mmol) in THF (7.0 mL) at 0 °C was added a solution of ϋΟΗ·Η2θ (0.259 g,

6.17 mmol) in H 2 O (6.0 mL). The resulting solution was warmed to room temperature and stirred for 3 h. The reaction mixture was diluted with EtOAc (100 mL) and was washed with sat. brine (5 x 50 mL). The organic layer was dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the crude product (700 mg) as a solid. LCMS (ESI) m/z: [M - H] calcd for C34H39N3O4: 552.29; found 552.2.

Intermediate A-4. Synthesis of N-methyl-N-(1-((S)-1-trltylaziridine-2-carbonyl)piperidine-4 - carbonyl)-L-valine.

Step 1: Synthesis of methyl N-methyl-N-(1-((S)-1-tritylaziridine-2-carbonyl)piperidine-4 -carbonyl)-

L-valinate

To a solution of methyl N-methyl-N-(piperidine-4-carbonyl)-Z.-valinate (0.550 g, 2.15 mmol) and (S)-1 -tritylaziridine-2-carboxylic acid (0.848 g, 2.57 mmol) in DMF (10.0 mL) at 0 °C was added DIPEA (1.9 mL, 10.7 mmol) and HATU (1.2 g, 3.2 mmol). The reaction mixture was warmed to room temperature and stirred for 1 h. The reaction mixture was diluted with EtOAc (50 mL) and washed with sat. NhUCI (60 mL). The aqueous layer was extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (200 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by reverse phase chromatography (10→80% MeCN/H 2 O) to afford the desired product (1.2 g, 98.5% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for C35H41N3O4: 568.32; found 568.3.

Step 2. Synthesis of N-methyl-N-(1-((S)-1-tritylaziridine-2-carbonyl)piperidine-4 -carbonyl)-L-valine To a solution of methyl N-methyl-N-(1 -((S)-1 -tritylaziridine-2-carbonyl)piperidine-4-carbonyl)-L- valinate (1.20 g, 2.11 mmol) in THF (11.0 mL) at 0 °C was added 1 M LiOH (10.57 mL, 10.57 mmol). The resulting solution was warmed to room temperature and stirred for 16 h. The reaction mixture was cooled to 0 °C and quenched with sat. NH4CI until pH 6. The resulting mixture was extracted with EtOAc (3 x 50 mL) and the combined organic layers were washed with brine (3 x 50 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the crude product (900 mg). LCMS (ESI) m/z: [M - H] calcd for C34H39N3O4: 554.29; found 554.3.

Intermediate A-5. Synthesis of N-methyl-N-(1-((fl)-1-trltylazlrldlne-2-carbonyl)azetldlne-3 - carbonyl)-L-valine

Step 1: Synthesis of methyl N-methyl-N-(1 -((fl)-1 -tritylaziridine-2-carbonyl)azetidine-3-carbonyl)-

L-valinate

To a solution of methyl N-(azetidine-3-carbonyl)-N- methyl-L-valinate (0.410 g, 1.79 mmol) and (fl)-1 -tritylaziridine-2-carboxylic acid (0.887 g, 2.69 mmol) in DMF (10 mL) at 0 °C was added DIPEA (1.56 mL, 8.98 mmol) and HATU (1.37 g, 3.59 mmol). The reaction mixture was stirred for 1 h. The resulting mixture was then extracted with EtOAc (3 x 50 mL) and the combined organic layers were washed with brine (3 x 20 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by reverse phase chromatography (10→80% MeCN/H 2 O) to afford the desired product (650 mg, 67% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for C33H37N3O4: 540.29; found 540.3.

Step 2. Synthesis of AAmethyl-AF(1-((/?)-1-tritylaziridine-2-carbonyl)azetidine-3 -carbonyl)-L-valine To a solution of methyl N-methyl-/\A(1 -((fl)-1 -tritylaziridine-2-carbonyl)azetidine-3-carbonyl)-L- valinate (0.650 mg, 1.20 mmol) in THF (10 mL) at 0 °C was added a 1 M solution of LiOHeH 2 O (6.03 mL). The reaction mixture was stirred for 3 h. The resulting mixture was then quenched with sat. NhUCI until pH 7. The resulting mixture was extracted with EtOAc (3 x 20 mL) and the combined organic layers were washed with brine (3 x 20 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired crude product (588 mg) as a solid. LCMS (ESI) m/z: [M - H] calcd for C32H35N3O4: 526.27; found 526.3. Intermediate A-6. Synthesis of N-methyl-N-(1-((S)-1-tritylazirldine-2-carbonyl)azetldlne-3- carbonyl)-L-valine

Step 1: Synthesis of methyl A/-methyl-N-(1-((S)-1-tritylaziridine-2-carbonyl)azetidine-3 -carbonyl)- L-valinate

To a solution of methyl N-(azetidine-3-carbonyl)-N-methyl-L-valinate (0.550 g, 2.41 mmol) and (S)-1 -tritylaziridine-2-carboxylic acid (0.952 g, 2.89 mmol) in DMF (10 mL) at 0 °C was added DIPEA (2.1 mL, 12.05 mmol) and HATU (1.37 g, 3.61 mmol). The reaction mixture was warmed to room temperature and stirred for 1 h. The resulting mixture was diluted with EtOAc (50 mL) and washed with sat. NH4CI (60 mL). The aqueous layer was then extracted with EtOAc (3 x 50 mL) and the combined organic layers were washed with brine (200 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by reverse phase chromatography (10→80% MeCN/H 2 O) to afford the desired product (820 mg, 63% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for C33H37N3O4: 540.29; found 540.3.

Step 2. Synthesis of N-methyl-N-(1-((S)-1-tritylaziridine-2-carbonyl)azetidine-3- carbonyl)-L-valine To a solution of methyl N-methyl-N-(1 -((S)-1 -tritylaziridine-2-carbonyl)azetidine-3-carbonyl)-L- valinate (0.800 g, 1.48 mmol) in THF (8.0 mL) at 0 °C was added 1 M LiOH (7.41 mL, 7.41 mmol). The reaction mixture was warmed to room temperature and stirred for 16 h and was then cooled to 0 °C and quenched with sat. NH4CI until pH 6. The resulting mixture was extracted with EtOAc (3 x 50 mL) and the combined organic layers were washed with brine (150 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by reverse phase chromatography (10→80% MeCN/H 2 O+0.5% NH4HCO3) to afford the desired product (440 mg, 56% yield) as a solid. LCMS (ESI) m/z: [M - H] calcd for C32H35N3O4: 524.25; found 524.2.

Intermediate A-7. Synthesis of (2fl,3S)-3-phenylazirldlne-2-carboxyllc acid

Step 1: Synthesis of ethyl (2S,3fl)-2,3-dihydroxy-3-phenylpropanoate

To a solution of ethyl cinnamate (2.0 g, 11.4 mmol) in f-BuOH (35.0 mL) and H 2 O (35.0 mL) at 0 °C was added AD-mix-β (15.83 g, 20.32 mmol), and methanesulfonamide (1.08 g, 11.3 mmol). The reaction mixture was stirred at room temperature for 16 h. The reaction was cooled to 0 °C and quenched with aq. KHSCU. The resulting mixture was extracted with EtOAc (3 x 100 mL) and the combined organic layers were washed with brine (2 x 90 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by normal phase chromatography (50% EtO Ac/pet. ether) to afford the desired product (2.2 g, 82% yield) as a solid.

Step 2. Synthesis of ethyl (2S,3fl)-3-hydroxy-2-(((4-nitrophenyl)sulfonyl)oxy)-3-phenyl propanoate To a solution of ethyl (2S,3fl)-2,3-dihydroxy-3-phenylpropanoate (2.0 g, 9.5 mmol) and EtaN (3.97 mL, 28.5 mmol) in DCM (30.0 mL) at 0 °C was added 4-nitrobenzenesulfonyl chloride (2.11 g,

9.51 mmol). The resulting mixture was stirred for 1 h and was then diluted with H 2 O (300 mL). The mixture was extracted with DCM (3 x 100 mL) and the combined organic layers were washed with brine (2 x 100 mL), dried over Na 2 SO 4 . filtered, and concentrated under reduced pressure. The residue was purified by prep-TLC (50% EtOAc/pet. ether) to afford the desired product (2.8 g, 67% yield) as a solid. Step 3: Synthesis of ethyl (2fl,3fl)-2-azido-3-hydroxy-3-phenylpropanoate To a solution of ethyl (2S,3fl)-3-hydroxy-2-(((4-nitrophenyl)sulfonyl)oxy)-3-phenyl propanoate (2.80 g, 7.08 mmol) in THE (30 mL) at room temperature was added trimethylsilyl azide (1.63 g,

14.2 mmol) and TBAF (1 M in THF, 14.16 mL, 14.16 mmol). The reaction mixture was heated to 60 °C and was stirred for 16 h. The reaction mixture was then cooled to room temperature, diluted with H 2 O (150 mL), and extracted with EtO Ac (3 x 50 mL). The combined organic layers were washed with brine

(2 x 30 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by normal phase chromatography (50% EtOAc/pet. ether) to afford the desired product (1.2 g, 64% yield) as an oil.

Step 4: Synthesis of ethyl (2fl,3S)-3-phenylaziridine-2-carboxylate

To a solution of ethyl (2f?,3fl)-2-azido-3-hydroxy-3-phenylpropanoate (1.20 g, 5.10 mmol) in DMF (15.0 mL) was added PPhs (1.61 g, 6.12 mmol). The reaction mixture was stirred at room temperature for 30 min and then heated to 80 °C for an additional 16 h. The reaction mixture was then cooled to room temperature, diluted with H 2 O (100 mL), and extracted with EtO Ac (3 x 40 mL). The combined organic layers were washed with brine (20 mL), dried over NaaSO, filtered, and concentrated under reduced pressure. The residue was purified by normal phase chromatography (16% EtOAc/pet. ether) to afford the desired product (620 mg, 57% yield) as an oil. LCMS (ESI) m/z: [M + H] calcd for C11H13NO2: 192.10; found 192.0.

Step 5: Synthesis of (2/?,3S)-3-phenylaziridine-2-carboxylic acid

To a solution of ethyl (2f?,3S)-3-phenylaziridine-2-carboxylate (0.100 g, 0.523 mmol) in MeOH (0.70 mL) at 0 °C was added a solution of LiOH (18.8 mg, 0.784 mmol) in H 2 O (0.70 mL). The reaction mixture was stirred for 1 h. The mixture was then diluted with MeCN (10 mL), and the resulting precipitate was collected by filtration and washed with MeCN (2 x 10 mL) to afford the crude desired product (70 mg) as a solid. LCMS (ESI) m/z: [M + H] calcd for C9H9NO2: 164.07; found 164.0.

Intermediate A-8. Synthesis of (2S,3fl)-3-phenylaziridine-2-carboxylic acid Step 1: Synthesis of ethyl (2fl,3S)-2,3-dihydroxy-3-phenylpropanoate To a solution of ethyl cinnamate (2.0 g, 11.4 mmol) in f-BuOH (35.0 mL) and H 2 O (35.0 mL) at 0 °C was added AD-mix-a (15.83 g, 20.32 mmol), and methanesulfonamide (1.08 g, 11.3 mmol). The reaction mixture was stirred at room temperature for 16 h. The reaction was cooled to 0 °C and quenched with aq. KHSO4. The resulting mixture was extracted with EtOAc (3 x 100 mL) and the combined organic layers were washed with brine (2 x 80 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by normal phase chromatography (50% EtOAc/pet. ether) to afford the desired product (2.2 g, 82% yield) as a solid.

Step 2. Synthesis of ethyl (2R,3S)-3-hydroxy-2-(((4-nitrophenyl)sulfonyl)oxy)-3-phenylp ropanoate To a solution of ethyl (2fl,3S)-2,3-dihydroxy-3-phenylpropanoate (2.10 g, 9.99 mmol) and EtaN (4.18 mL, 29.9 mmol) in DCM (30.0 mL) at 0 °C was added 4-nitrobenzenesulfonyl chloride (2.21 g,

9.99 mmol). The resulting mixture was stirred for 1 h and was then diluted with H 2 O (200 mL). The mixture was extracted with DCM (3 x 80 mL) and the combined organic layers were washed with brine (2 x 80 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by prep-TLC (50% EtOAc/pet. ether) to afford the desired product (3.0 g, 68% yield) as a solid. Step 3: Synthesis of ethyl (2S,3S)-2-azido-3-hydroxy-3-phenylpropanoate To a solution of ethyl (2f?,3S)-3-hydroxy-2-(((4-nitrophenyl)sulfonyl)oxy)-3-phenyl propanoate (3.0 g, 7.59 mmol) in THF (30 mL) at room temperature was added trimethylsilyl azide (1.75 g, 15.2 mmol) and TBAF (1 M in THF, 15.18 mL, 15.18 mmol). The reaction mixture was heated to 60 °C and was stirred for 16 h. The reaction mixture was then cooled to room temperature, diluted with H 2 O (150 mL), and extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (2 x 30 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by normal phase chromatography (50% EtOAc/pet. ether) to afford the desired product (1.4 g, 70% yield) as an oil.

Step 4: Synthesis of ethyl (2S,3fl)-3-phenylaziridine-2-carboxylate

To a solution of ethyl (2S,3S)-2-azido-3-hydroxy-3-phenylpropanoate (1.40 g, 5.95 mmol) in DMF (20.0 mL) was added PPha (1.87 g, 7.14 mmol). The reaction mixture was stirred at room temperature for 30 min and then heated to 80 °C for an additional 16 h. The reaction mixture was then cooled to room temperature, diluted with H 2 O (150 mL), and extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (40 mL), dried over NaaSO, filtered, and concentrated under reduced pressure. The residue was purified by normal phase chromatography (16% EtOAc/pet. ether) to afford the desired product (720 mg, 56% yield) as an oil. LCMS (ESI) m/z: [M + H] calcd for C11H13NO2: 192.10; found 192.0.

Step 5: Synthesis of (2S,3fl)-3-phenylaziridine-2-carboxylic acid

To a solution of ethyl (2S,3fl)-3-phenylaziridine-2-carboxylate (0.100 g, 0.523 mmol) in MeOH (0.70 mL) at 0 °C was added a solution of LiOH (18.8 mg, 0.784 mmol) in H 2 O (0.70 mL). The reaction mixture was stirred for 1 h. The mixture was then diluted with MeCN (10 mL), and the resulting precipitate was collected by filtration and washed with MeCN (2 x 10 mL) to afford the crude desired product (68 mg) as a solid. LCMS (ESI) m/z: [M + H] calcd for CgHgNOa: 164.07; found 164.0. Intermediate A-9. Synthesis of N-(W-((fl)-1-benzylaziridine-2-cart>onyl)-N-methylglycyl) -W· methyl-i-valine

Step 1: Synthesis of methyl N-(N-(te/1-butoxycarbonyl)-N- methylglycyl)-N-methyl-L-valinate To a solution of methyl methyl-L-valinate hydrochloride (4.0 g, 22.0 mmol) and N-(tert- butoxycarbonyl)-N-methylglycine (5.0 g, 26.4 mmol) in DCM (100.0 mL) was added EtsN (9.2 mL,

66.1 mmol) and HATU (10.88 g, 28.63 mmol). The reaction mixture was stirred for 4 h. The reaction was then neutralized to pH 7 with sat. aq. NaHCOs. The mixture was extracted with DCM and the combined organic layers were washed with brine, dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by normal phase chromatography (50% EtOAc/pet. ether) to afford the desired product (6.2 g, 89% yield) as an oil. LCMS (ESI) m/z: [M + H] calcd for C15H28N 2 O5: 317.21 ; found 317.2.

Step 2. Synthesis of methyl N-methyl-N-(methylglycyl)-L-valinate hydrochloride To a solution of methyl AA-(N-(tert-butoxycarbonyl)-N-methylglycyl)-AAmethyl-L-valin ate (4.97 g, 15.7 mmol) in EtOAc (150.0 mL) at 0 °C was added HCI (4M in dioxane, 50.0 mL, 200 mmol). The reaction mixture was stirred for 3 h and then concentrated under reduced pressure to afford the desired crude product (4.26 g, 107% yield) as an oil. LCMS (ESI) m/z: [M + H] calcd for C10H20N 2 O3: 217.16; found 217.1.

Step 3: Synthesis of methyl N-methyl-N-(N-methyl-N- ((fl)-1 -tritylaziridine-2-carbonyl)glycyl)-L- valinate

To a solution of methyl N-methyl-N-(methylglycyl)-L-valinate hydrochloride (1.0 g, 3.9 mmol) and (fl)-1 -tritylazihdine-2-carboxylic acid (1.30 g, 3.94 mmol) in DCM (25.0 mL) was added EtaN (2.76 mL, 19.8 mmol) and HATU (1.81 g, 4.76 mmol). The reaction mixture was stirred for 1 h. The reaction was then neutralized to pH 7 with sat. aq. NaHCOs. The mixture was extracted with DCM and the combined organic layers were washed with brine, dried over NasSCX filtered, and concentrated under reduced pressure. The residue was purified by normal phase chromatography (50% EtOAc/pet. ether) to afford the desired product (1.1 g, 52.6% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for C32H37N3O4: 528.29; found 528.2.

Step 4: Synthesis of methyl N-(N-((fl)-aziridine-2-carbonyl)-AAmethylglycyl)-N-methyl-Z. -valinate To a solution methyl W-methyl-N-(N-methyl-N- ((fl)-1-tritylaziridine-2-carbonyl)glycyl)-L-valinate (1.0 g, 3.9 mmol) in DCM (6 mL) at 0 °C was added TFA (2 mL). The reaction mixture was warmed to room temperature and stirred for 1 h, then concentrated under reduced pressure to afford the desired crude product (250 mg) as an oil. LCMS (ESI) m/z: [M + H] calcd for C13H23N3O4: 286.18; found 286.1 . Step 5: Synthesis of methyl N-(N-((fl)-1-benzylaziridine-2-carbonyl)-N-methylglycyl)-W-m ethyl-L- valinate

To a solution of methyl N-(N-((fl)-aziridine-2-carbonyl)-N-methylglycyl)-N-methyl-L- valinate (220.0 mg, 0.771 mmol) in MeCN (2.0 mL) was added DIPEA (537 pL, 3.08 mmol) and benzyl bromide (101 pL, 0.848 mmol). The reaction mixture was stirred for 6 h. The reaction mixture was then concentrated under reduced pressure. The residue was purified by prep-TLC (9% MeOH/DCM) to afford the desired product (261 mg, 90% yield) as an oil. LCMS (ESI) m/z: [M + H] calcd for C20H29N3O4: 376.22; found 376.2.

Step 6: Synthesis of W-(N-((fl)-1-benzylaziridine-2-carbonyl)-N- methylglycyl)-N-methyl-L-valine To a solution of methyl AA(N-((fl)-1-benzylaziridine-2-carbonyl)-N-methylglycyl)-N-m ethyl-L- valinate (261.0 mg, 0.695 mmol) in THF (3.38 mL) was added a solution of LiOH (83.2 mg, 3.48 mmol) in H 2 O (3.50 mL). The reaction mixture was stirred for 1 h. The reaction was then quenched with sat. aq. NhUCI. The resulting mixture was extracted with EtOAc and the combined organic layers were washed with brine, dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by reverse phase chromatography (10→50% MeCN/H 2 O) to afford the desired product (230 mg, 91% yield) as an oil. LCMS (ESI) m/z: [M + H] calcd for C19H27N3O4: 362.21 ; found 362.2.

Intermediate A-10. Synthesis of N-(N-((fl)-1-benzylazirldlne-2-carbonyl)-AAmethylglycyl)-N- methyl-L-valine

O I O o o

BnBr, DIRE* LIOH

'°¾nV H HO 1 N N Ph

MeCN THF/H z O

Step 1: Synthesis of methyl N-(N-((S)-1-benzylaziridine-2-carbonyl)-N-methylglycyl)-N-me thyl-L- valinate

To a solution of methyl N-(N-((S)-aziridine-2-carbonyl)-N-methylglycyl)-N-methyl-L-v alinate (362.0 mg, 1.269 mmol) in MeCN (6.0 mL) at 0 °C was added DIPEA (883 pL, 5.08 mmol) and benzyl bromide (165 pL, 1.39 mmol). The reaction mixture was then warmed to room temperature and stirred overnight. The reaction mixture was then concentrated under reduced pressure. The residue was purified by prep-TLC (7% MeOH/DCM) to afford the desired product (287 mg, 60% yield) as an oil. LCMS (ESI) m/z: [M + H] calcd for C20H29N3O4: 376.22; found 376.2.

Step 2. Synthesis of N-(N-((S)-1-benzylaziridine-2-carbonyl)-N- methylglycyl)-N-methyl-Z.-valine To a solution of methyl N-(N-((S)-1-benzylaziridine-2-carbonyl)-N- methylglycyl)-N-methyl-Z.- valinate (270.0 mg, 0.719 mmol) in THF (3.6 mL) was added a solution of LiOH (86.1 mg, 3.59 mmol) in H 2 O (3.60 mL). The reaction mixture was stirred for 30 min. The reaction was then quenched with sat. aq. NH4CI. The resulting mixture was extracted with EtOAc (3 x 15 mL) and the combined organic layers were washed with brine, dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired crude product (240 mg, 92% yield) as an oil. LCMS (ESI) m/z: [M + H] calcd for C19H27N3O4: 362.21 ; found 362.2.

Intermediate A-11. Synthesis of N-methyl-W-(N-methyl-N-{(fl)-1-tritylaziridine-2- carbonyl)glycyl)-L-valine

To a solution of methyl N-methyl-N-(N-methyl-N-((fl)-1-tritylaziridine-2-carbonyl)gl ycyl)-Z--valinate (1.30 g, 2.46 mmol) in THF (10.0 mL) at 0 °C was added a solution of LIOH (177.0 mg, 7.39 mmol) in hfeO (7.40 mL). The resulting mixture was warmed to room temperature, stirred for 3 h, and was then acidified to pH 5 with HCI (aq). The resulting mixture was extracted with EtOAc (3 x 80 mL) and the combined organic layers were washed with brine (2 x 50 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired crude product (1 g, 71% yield). LCMS (ESI) m/z: [M + H] calcd for C31H35N3O4: 514.27; found 514.3.

Intermediate A-12. Synthesis of W-methyl-N-(4-((fl)-1-tritylaziridine-2-carbonyl)-1,4- diazepane-1 -carbonyl)-L-vallne

Step 1: Synthesis of benzyl (S)-4-((1 -methoxy-3-methyl-1 -oxobutan-2-yl)(methyl)carbamoyl)-1 ,4- diazepane-1 -carboxylate

To a solution of methyl N-methyl-L-valinate (2.50 g, 17.22 mmol) in DCM at 0 °C was added DIPEA (1.8 mL, 10.33 mmol) followed by triphosgene (2.55 g, 8.61 mmol). The resulting mixture was stirred for 3 h at 0 °C. To the mixture was then added benzyl 1 ,4-diazepane-1 -carboxylate (4.03 g,

17.20 mmol). The resulting mixture was warmed to room temperature and stirred overnight. The reaction was cooled to 0 °C and was quenched with NaHCOs. The aqueous layer was extracted with EtOAc (2 x 30 mL) and the combined organic layers were washed with brine (2 x 30 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by normal phase chromatography (25% EtOAc/pet. ether) to afford the desired product (3.5 g, 50.1% yield). LCMS (ESI) m/z. [M + H] calcd for C21H31N3O5: 406.23; found 406.5.

Step 2. Synthesis of methyl N-{ 1 ,4-diazepane-1-carbonyl)-N-methyl-L-valinate

To a solution of benzyl (S)-4-((1-methoxy-3-methyl-1-oxobutan-2-yl)(methyl)carbamoyl )-1 ,4- diazepane-1 -carboxylate (2.0 g, 4.93 mmol) in MeOH (20 mL) was added Pd/C (10%wt, 1 g). The mixture was placed under a hydrogen atmosphere (1 atm) and stirred for 2 h. The reaction mixture was filtered through a Celite and concentrated under reduced pressure to afford the desired crude product (1.3 g, 97.1 % yield). LCMS (ESI) m/z. [M + H] calcd for C13H25N3O3: 272.20; found 272.3.

Step 3: Synthesis of methyl N-methyl-N-(4-((fl)-1-tritylaziridine-2-carbonyl)-1 ,4-diazepane-1- carbonyl)-L-valinate

To a solution of methyl N-( 1 ,4-diazepane-1-carbonyl)-N- methyl-L-valinate (1.0 g, 3.69 mmol) and (fl)-1-tritylaziridine-2-carboxylic acid (1.46 g, 4.42 mmol) in DMF at 0 °C was added DIPEA (1.93 mL,

11.06 mmol) followed by HATU (2.10 g, 5.52 mmol). The resulting mixture was warmed to room temperature and stirred for 1 h. The reaction mixture was then diluted with H 2 O (15 mL) and the aqueous layer was extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (3 x 30 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by normal phase chromatography (25% EtOAc/pet. ether) to afford the desired product (1.6 g, 74.5% yield). LCMS (ESI) m/z. [M + H] calcd for C35H42N4O4: 583.33; found 583.5.

Step 4: Synthesis of N-methyl-N-(4-((f7)-1 -tritylaziridine-2-carbonyl)-1 ,4-diazepane-1 -carbonyl)-!- valine

To a solution of methyl N-methyl-N-(4-((fl)-1 -tritylaziridine-2-carbonyl)-1 ,4-diazepane-1 -carbonyl)- L-valinate (1.60 g, 2.75 mmol) in MeOH (10.0 mL) and H 2 O (5.0 mL) at 0 °C was added LiOH (0.66 g, 27.56 mmol). The resulting mixture was warmed to room temperature and stirred overnight. The reaction mixture was acidified to pH 5 with HCI (aq) and the aqueous layer was extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (3 x 30 mL), dried over Na 2 SO 4 . filtered, concentrated under reduced pressure to afford the desired crude product (1.4 g, 95.6% yield). LCMS (ESI) m/z. [M + H] calcd for C34H40N4O4: 569.31 ; found 569.5.

Intermediate A-13. Synthesis of W-methyl-W-(4-((S)-1-tritylaziridine-2-carbonyl)-1,4- diazepane-1-carbonyl)-L-vaHne

Step 1: Synthesis of methyl N-methyl-N-(4-((S)-1 -tritylaziridine-2-carbonyl)-1 ,4-diazepane-1 - carbonyl)-L-valinate

To a solution of methyl N-( 1 ,4-diazepane-1 -carbonyl)-N- methyl-L-valinate (1.16 g, 4.28 mmol) and (S)-1-tritylaziridine-2-carboxylic acid (1.69 g, 5.13 mmol) in DMF (10 mL) at 0 °C was added DIPEA (2.23 mL, 12.82 mmol) followed by HATU (2.44 g, 6.41 mmol). The resulting mixture was stirred for 1 h at 0 °C. The reaction mixture was then diluted with H 2 O (15 mL) and the aqueous layer was extracted with EtOAc (3 x 15 mL). The combined organic layers were washed with brine (3 x 15 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by normal phase chromatography (17% EtOAc/pet. ether) to afford the desired product (2 g, 80.3% yield). LCMS (ESI) m/z. [M + H] calcd for C35H42N4O4: 583.33; found 583.5. Step 2. Synthesis of N-methyl-N-(4-((S)-1-tritylaziridine-2-carbonyl)-1 ,4-diazepane-1 -carbonyl)-L- valine

To a solution of methyl N-methyl-N-(4-((S)-1-tritylaziridine-2-carbonyl)-1 ,4-diazepane-l-carbonyl)- L-valinate (1.0 g, 1.72 mmol) in MeOH (8.0 mL) and H 2 O (4.0 mL) at 0 °C was added LIOH (411 mg,

17.16 mmol). The resulting mixture was warmed to room temperature and stirred overnight. The reaction mixture was acidified to pH 5 with HCI (aq) and the aqueous layer was extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over NazSO*, filtered, concentrated under reduced pressure to afford the desired crude product (0.6 g, 61.5% yield). LCMS (ESI) m/z. [M + H] calcd for C34H40N4O4: 569.31 ; found 569.5.

Intermediate A-14. Synthesis of A#-methyl-Af-(5-((S)-1-trltylazlrldine-2- caitx>xamldo)picolinoyl)-L-vallne

Step 1: Synthesis of methyl N-methyl-N-(5-nitropicolinoyl)-L-valinate

To a solution of methyl N-methyl-L-valinate hydrochloride (190.0 mg, 1.31 mmol) and 5- nitropicolinic acid (200.0 mg, 1.19 mmol) in DMF (2 mL) at 0 °C was added HATU (678.6 mg, 1.79 mmol) and EtaN (0.332 mL, 2.38 mmol). The resulting mixture was warmed to room temperature and stirred for 2 h. The resulting mixture was then extracted with EtOAc (2 x 50 mL) and the combined organic layers were washed with H 2 O (20 mL) and brine (20 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by normal phase chromatography (33% EtOAc/pet. ether) to afford the desired product (210 mg, 59.8% yield). LCMS (ESI) m/z·. [M + H] calcd for C13H17N3O5: 296.12; found 296.0.

Step 2. Synthesis of methyl N-(5-aminopicolinoyl)-N-methyl-L-valinate

To a solution of methyl N-methyl-N-(5-nitropicolinoyl)-L-valinate (5.0 g, 16.93 mmol) in MeOH (50.0 mL) was added Pd/C (2.50 g). The reaction mixture was placed under a hydrogen atmosphere (1 atm) and was stirred for 2 h. The mixture was filtered, the filter cake was washed with MeOH (2 x 20 mL), and the filtrate was concentrated under reduced pressure to afford the desired crude product (5.3 g). LCMS (ESI) m/z. [M + H] calcd for C13H19N3O3: 266.15; found 266.0.

Step 3: Synthesis of methyl N-methyl-N-(5-((S)-1-tritylaziridine-2-carboxamido)picolinoy l)-L- valinate

To a solution (S)-1 -tritylaziridine-2-carboxylic acid (55.9 mg, 0.17 mmol) in DCM at 0 °C was added isobutyl chloroformate (21.7 pL, 0.23 mmol) and N-methylmorpholine (66.8 pL, 0.61 mmol). The resulting mixture was stirred for 1 h and then methyl N-(5-aminopicolinoyl)-W-methyl-L-valinate (30.0 mg, 0.11 mmol) was added. The resulting mixture was warmed to room temperature and stirred for an additional 5 h. The mixture was extracted with DCM (3 x 50 mL) and the combined organic layers were washed with sat. NaHCOs (30 mL) and brine, dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by normal phase chromatography (33% EtOAc/pet. ether) to afford the desired product (1.09 g, 66.9% yield). LCMS (ESI) m/z. [M + H] calcd for C35H36N4O4: 577.28; found 577.1.

Step 4: Synthesis of N-methyl-N-(5-((S)-1-tritylaziridine-2-carboxamido)picolinoy l)-L-valine To a solution methyl AAmethyl-N-(5-((S)-1-tritylaziridine-2-carboxamido)picolinoy l)-/.-valinate (100.0 mg, 0.17 mmol) in THE (0.5 mL) at 0 °C was added a solution of LiOH (20.76 mg, 0.87 mmol) in H 2 O (0.5 mL). The resulting mixture was warmed to room temperature and stirred for 6 h. The mixture was acidified to pH 5 with 1 M citric acid. The resulting mixture was extracted with EtOAc (3 x 20 mL) and the combined organic layers were washed with brine (5 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired crude product (76.8 mg, 78.7% yield). LCMS (ESI) m/z : [M + H] calcd for C34H34N4O4: 563.27; found 563.3.

Intermediate A-15. Synthesis of W-methyl-N-(5-((fl)-1-tritylaziridine-2- caitx>xamido)picolinoyl)-i.-valine

NHj isobutyl chloroformete H 09 H

N N IW. N.

NMM 'Tit UOH Tit

DCM THF/H 2 O

Step 1: Synthesis of methyl N-methyl-N-(5-((f7)-1-tritylaziridine-2-carboxamido)picolino yl)-L· valinate

To a solution (fl)-1 -tritylaziridine-2-carboxylic acid (1396.7 mg, 4.24 mmol) in DCM (8 mL) at 0 °C was added isobutyl chloroformate (440 pL, 3.39 mmol) and N-methylmorpholine (466 pL, 4.24 mmol). The resulting mixture was stirred for 1 h and then methyl N-(5-aminopicolinoyl)-N-methyl-L-valinate (750.0 mg, 2.83 mmol) was added. The resulting mixture was warmed to room temperature and stirred for an additional 5 h. The mixture was quenched by the addition of NaHCOa and the aqueous layer was extracted with DCM (2 x 100 mL). The combined organic layers were washed with brine (120 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by normal phase chromatography (50% EtOAc/pet. ether) to afford the desired product (580 mg, 35.6% yield). LCMS (ESI) m/z: [M + H] calcd for C35H36N4O4: 577.28; found 577.2.

Step 2. Synthesis of N-methyl-N-(5-((fl)-1-tritylaziridine-2-carboxamido)picolino yl)-L-valine To a solution methyl W-methyl-N-(5-((fl)-1-tritylaziridine-2-carboxamido)picolino yl)-/--valinate (558.0 mg, 0.97 mmol) in THF (14 mL) at 0 °C was added a solution of LiOH (115.9 mg, 4.84 mmol) in H 2 O (14 mL). The resulting mixture was warmed to room temperature and stirred for 6 h. The mixture was acidified to pH 5 with 1 M citric acid. The resulting mixture was extracted with EtOAc (3 x 100 mL) and the combined organic layers were washed with brine (30 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired crude product (580 mg, 78.7% yield). LCMS (ESI) m/z: [M + H] calcd for C34H34N4O4: 563.27; found 563.2.

Intermediate A-16. Synthesis of (2/?,3S)-1-((fl)-tert-butylsulfinyl)-3- (methoxycarbonyl)aziridine-2-carboxylic acid o

Step 1: Synthesis of methyl (fl,£)-2-((tert-butylsulfinyl)imino)acetate

To a solution of (fl)-2-methylpropane-2-sulfinamide (13.21 g, 109.01 mmol) and methyl 2- oxoacetate (8.0 g, 90.85 mmol) in DCM (130 mL) at room temperature was added MgSO* (54.67 g, 454.23 mmol). The resulting mixture was heated to 35 °C and stirred for 16 h. The resulting mixture was filtered, the filter cake washed with EtOAc (3 x 50 mL), and the filtrate was concentrated under reduced pressure. The residue was purified by normal phase chromatography (25% EtOAc/pet. ether) to afford the desired (5.8 g, 33.4% yield). LCMS (ESI) m/z. [M + H] calcd for C7H13NO3S: 192.07; found 191.9.

Step 2: Synthesis of 2-(tert-butyl) 3-methyl (2f?,3S)-1-((fl)-tert-butylsulfinyl)aziridine-2,3- dicarboxylate

To a solution of 1 M LiHMDS (61.40 mL, 61.40 mmol) in THE (300.0 mL) at -78 °C was added tert-butyl 2-bromoacetate (11.83 g, 60.65 mmol). The resulting mixture was stirred for 30 min. To the reaction mixture was then added methyl methyl (fl,E)-2-((tert-butylsulfinyl)imino)acetate (5.8 g, 30.33 mmol). The resulting mixture was warmed to -60 °C and stirred for 2.5 h. The reaction was warmed to 0 °C and quenched with sat. NH4CI (aq.). The resulting mixture was extracted with EtOAc (3 x 200 mL). The combined organic layers were washed with brine (500 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by reverse phase chromatography (10→50% MeCN/H 2 O) to afford the desired product (1 .34 g, 4.5% yield). LCMS (ESI) m/z. [M + H] calcd for CiaHaaNOsS: 306.14; found 306.2.

Step 3: Synthesis of (2fl,3S)-1-((fl)-tert-butylsulfinyl)-3-(methoxycarbonyl)azir idine-2-carboxylic acid

To a solution of 2-(tert-butyl) 3-methyl (2f?,3S)-1 -((fl)-tert-butylsulfinyl)aziridine-2,3-dicarboxylate (302.0 mg, 0.99 mmol) in DCM (3.0 mL) at 0 °C was added TFA (1.50 mL). The resulting mixture was stirred for 1 h and then concentrated under reduced pressure to afford the desired crude product (300 mg). LCMS (ESI) m/z. [M + H] calcd for C9H15NO5S: 250.07; found 250.1.

Intermediate A-17. Synthesis of (2/?,3S)-1-((S)-tort-butylsulflnyl)-3- (methoxycarbonyl)aziridine-2-carboxylic acid

Step 1: Synthesis of methyl (S,E)-2-((terf-butylsulfinyl)imino)acetate

To a solution of (S)-2-methylpropane-2-sulfinamide (9.81 g, 80.94 mmol) and methyl 2-oxoacetate (5.94 g, 67.45 mmol) in DCM (100 mL) at room temperature was added MgS04 (40.60 g, 337.26 mmol). The resulting mixture was heated to 35 °C and stirred for 16 h. The resulting mixture was filtered, the filter cake washed with EtOAc (3 x 50 mL), and the filtrate was concentrated under reduced pressure. The residue was purified by normal phase chromatography (25% EtOAc/pet. ether) to afford the desired (5.68 g, 44.0% yield). LCMS (ESI) m/z. [M + H] calcd for C7H13NO3S: 192.07; found 191.1 .

Step 2. Synthesis of 2-(tert-butyl) 3-methyl (2f?,3S)-1-((S)-tert-butylsulfinyl)aziridine-2,3- dicarboxylate

To a solution of 1 M LiHMDS (59.40 mL, 59.40 mmol) in THF (300.0 mL) at -78 °C was added tert-butyl 2-bromoacetate (11.59 g, 59.40 mmol). The resulting mixture was stirred for 30 min. To the reaction mixture was then added methyl methyl (S,E)-2-((tert-butylsulfinyl)imino)acetate (5.68 g, 29.70 mmol). The resulting mixture was warmed to -60 °C and stirred for 2.5 h. The reaction was warmed to 0 °C and quenched with sat. NhUCI (aq.). The resulting mixture was extracted with EtOAc (3 x 200 mL). The combined organic layers were washed with brine (500 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by reverse phase chromatography (10→50% MeCN/H 2 O) to afford the desired product (1 .26 g, 13.9% yield). LCMS (ESI) m/z. [M + H] calcd for CiaHaaNOsS: 306.14; found 306.1.

Step 3: Synthesis of (2f?,3S)-1-((S)-tert-butylsulfinyl)-3-(methoxycarbonyl)aziri dine-2-carboxylic acid

To a solution of 2-(tert-butyl) 3-methyl (2f?,3S)-1 -((S)-terf-butylsulfinyl)aziridine-2,3-dicarboxylate (457.0 mg, 1.50 mmol) in DCM (6.0 mL) at 0 °C was added TEA (3.0 mL). The resulting mixture was stirred for 1 h and then concentrated under reduced pressure to afford the desired crude product (450 mg). LCMS (ESI) m/z. [M + H] calcd for C9H15NO5S: 250.07; found 250.1.

Intermediate A-18. Synthesis of (2/?,3fl)-1-((fl)-tort-butylsulflnyl)-3-cyclopropylazlrldlne -2- carboxylic acid

Step 1: Synthesis of (/?,E)-N-(cyclopropylmethylene)-2-methylpropane-2-sulfinamid e To a solution of (fl)-2-methylpropane-2-sulfinamide (1.0 g, 8.25 mmol) and cyclopropanecarbaldehyde (1.16 g, 16.55 mmol) in DCM (50 mL) at room temperature was added CuSCU (3.95 g, 24.75 mmol). The resulting mixture was stirred overnight. The reaction mixture was then filtered, the filter cake washed with EtOAc, and the filtrate was concentrated under reduced pressure. The residue was purified by prep-TLC (17% EtOAc/pet. ether) to afford the desired product (1 .4 g, 97.9% yield).

LCMS (ESI) m/z. [M + H] calcd for CBHISNOS: 174.10; found 174.1.

Step 2. Synthesis of ethyl (2/?,3fl)-1-((fl)-tert-butylsulfinyl)-3-cyclopropylaziridine -2-carboxylate To a solution of 1 M LiHMDS (23 mL, 23 mmol) in THF (50.0 mL) at -78 °C was added ethyl bromoacetate (3.83 g, 22.95 mmol). The resulting mixture was warmed to -70 °C and stirred for 1 h. To the reaction mixture was then added (fl,E)-N-(cyclopropylmethylene)-2-methylpropane-2-sulfinamid e (2.0 g, 11.48 mmol). The resulting mixture was stirred for 1 h at -70 °C. The reaction mixture was warmed to 0 °C and quenched with HzO. The aqueous layer was extracted with EtOAc (3 x 100 mL). The combined organic layers were washed with brine, dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by prep-TLC (25% EtOAc/pet. ether) to afford the desired product (1.8 g, 60.5% yield). m/z. [M + H] calcd for C12H21NO3S: 306.14; found 260.13.

Step 3: Synthesis of fl)-tert-butylsulfinyl)-3-cyclopropylaziridine-2-carboxylic acid To a solution of ethyl (fl)-tert-butylsulfinyl)-3-cyclopropylaziridine-2-carboxylat e (900.0 mg, 3.47 mmol) in TH nd H 2 O (3.0 mL) at 0 °C was added LiOHeH 2 O (218.4 mg,

5.21 mmol). The resulting mixture was stirred for 1 h and was then quenched by H 2 O. The aqueous layer was extracted with EtOAc (3 x 50) and the combined organic layers were washed with brine, dried over N32S04, filtered, and concentrated under reduced pressure to afford the desired crude product (400 mg, 29.9% yield). LCMS (ESI) m/z. [M + H] calcd for C10H17NO3S: 232.10; found 232.1.

Intermediate A-19. Synthesis of (2/?,3fl)-1-((fl)-ferMxitylsulflnyl)-3-methylazlrldlne-2- carboxylic acid o o o

Step i: Synthesis of (f?,£)-N-ethylidene-2-methylpropane-2-sulfinamide To a solution of (fl)-2-methylpropane-2-sulfinamide (3.0 g, 24.75 mmol) and tetraethoxytitanium (1.7 g, 7.43 mmol) in THF (30 mL) at 0 °C was added acetaldehyde (218.1 mg, 4.95 mmol). The resulting mixture was stirred for 20 min and was then quenched with HzO (100 mL). The suspension was filtered, and the filter cake washed with EtO Ac (3 x 100 mL). The aqueous layer was extracted with EtOAc (3 x 100 mL) and the combined organic layers were washed with brine (3 x 100 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by normal phase chromatography (9% EtOAc/pet. ether) afforded desired product (3 g, 82% yield). LCMS (ESI) m/z. [M + H] calcd for CeHisNOS: 148.08; found 148.0.

Sfep 2. Synthesis of ethyl (2fl,3fl)-1-((fl)-tert-butylsulfinyl)-3-methylaziridine-2-ca rboxylate To a solution of 1 M LiHMDS (40.75 mL, 40.75 mmol) in THF (30.0 mL) at -78 °C was added ethyl bromoacetate (6.80 g, 40.75 mmol). The resulting mixture was stirred for 1 h. To the reaction mixture was then added (/?,E)-N-ethylidene-2-methylpropane-2-sulfinamide (3.0 g, 20.38 mmol). The resulting mixture was stirred for 2 h at -78 °C and then quenched with H 2 O (300 mL). The aqueous layer was extracted with EtOAc (3 x 300 mL) and the combined organic layers were washed with brine (3 x 100 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by reverse phase chromatography (10→50% MeCN/H 2 O) to afford the desired product (1.4 g, 29.5% yield). LCMS (ESI) m/z. [M + H] calcd for C10H19NO3S: 234.12; found 234.1.

Step 3: Synthesis of (2/?,3fl)-1-((fl)-tert-butylsulfinyl)-3-methylaziridine-2-ca rboxylic acid To a solution of ethyl (2fl,3fl)-1-((fl)-/erf-butylsulfinyl)-3-methylaziridine-2-ca rboxylate (1.0 g,

4.29 mmol) in THF (6.4 mL) and H 2 O (6.4 mL) at 0 °C was added LiOHeH 2 O (539.5 mg, 12.86 mmol).

The resulting mixture was warmed to room temperature and stirred for 2 h and was then neutralized to pH 5 with HCI (aq.) and sat. NH4CI (aq.). The aqueous layer was extracted with EtOAc (3 x 10 mL) and the combined organic layers were washed with brine, dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired crude product (489 mg, 55.6% yield). LCMS (ESI) m/z: [M + H] calcd for CeHisNOaS: 206.09; found 206.0.

Intermediate A-20. Synthesis of (2S,3S)-1-(S)-ferf-butylsulfinyl)-3-methylaziridine-2- carboxylic acid

Step 1: Synthesis of (S,£)-N-ethylidene-2-methylpropane-2-sulfinamide To a mixture of (S)-2-methylpropane-2-sulfinamide (5.0 g, 41.25 mmol) and tetraethoxytitanium (18.82 g, 82.51 mmol) at 0 °C was added acetaldehyde (3.63 g, 82.51 mmol). The resulting mixture was warmed to room temperature and stirred for 30 min and was then quenched with H 2 O (100 mL). The suspension was filtered, and the filter cake washed with EtOAc (3 x 100 mL). The aqueous layer was extracted with EtOAc (3 x 100 mL) and the combined organic layers were washed with brine (3 x 100 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford desired crude product (3.9 g, 64% yield). LCMS (ESI) m/z: [M + H] calcd for CeHisNOS: 148.08; found 148.2.

Sfep 2: Synthesis of ethyl (2S,3S)-1-((S)-tert-butylsulfinyl)-3-methylaziridine-2-carbo xylate To a solution of 1 M LiHMDS (40.75 mL, 40.75 mmol) in THF (30.0 mL) at -78 °C was added ethyl bromoacetate (6.80 g, 40.75 mmol). The resulting mixture was stirred for 1 h. To the reaction mixture was then added (S,E)-N-ethylidene-2-methylpropane-2-sulfinamide (3.0 g, 20.38 mmol). The resulting mixture was stirred for 2 h at -78 °C and then quenched with H 2 O. The aqueous layer was extracted with EtOAc (3 x 200 mL) and the combined organic layers were washed with brine (3 x 300 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by reverse phase chromatography (10→50% MeCN/H 2 O) to afford the desired product (2 g, 42% yield). LCMS (ESI) m/z: [M + H] calcd for C10H19NO3S: 234.12; found 234.0.

Step 3: Synthesis of (2S,3S)-1-((S)-fe/t-butylsulfinyl)-3-methylaziridine-2-carbo xylic acid To a solution of ethyl (2S,3S)-1-((S)-tert-butylsulfinyl)-3-methylaziridine-2-carbo xylate (80.0 mg, 0.34 mmol) in THF (1.0 mL) and H 2 O (0.2 mL) at 0 °C was added LiOHeH 2 O (32.9 mg, 1.37 mmol). The resulting mixture was warmed to room temperature and stirred for 4 h and was then acidified to pH 3 with HCI (aq.). The aqueous layer was extracted with EtOAc (3 x 10 mL) and the combined organic layers were washed with brine, dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired crude product (70 mg, 99% yield). LCMS (ESI) m/z: [M + H] calcd for CeHisNOaS: 206.09; found 206.0.

Intermediate A-21 and A-22. Synthesis of AFmethyl-N-((S)-1-(((fl)-1-methylazlrldln-2- yl)sulfonyl)pyrrolldlne-3-carbonyl)-L-vallne and AAmethyFAA((S)-1-(((S)-1-methylazlrldln-2- yl)sulfonyl)pyrrolldlne-3-carbonyl)-L-vallne

Step 1: Synthesis of methyl N-methyl-N-((S)-1-(vinylsulfonyl)pyrrolidine-3-carbonyl)-L-v alinate To a mixture of methyl A/-methyl-A/-((S)-pyrrolidine-3-carbonyl)-L-valinate (7.0 g, 28.89 mmol) in MeCN (200 mL) at -20 °C was added DIPEA (10.0 mL, 57.78 mmol) followed by ethenesulfonyl chloride (4.0 g, 31.78 mmol). The resulting solution was stirred for 2 h at -20 °C and was then diluted with EtOAc

(800 mL). The resulting solution was washed with brine (3 x 100 mL) and concentrated under reduced pressure. Purification by normal phase chromatography (50% EtOAc/pet. ether) afforded the desired product (4.8 g, 49.9%, yield). LCMS (ESI) m/z: [M + H] calcd for C14H24N 2 OSS: 333.15; found 333.1 . Step 2. Synthesis of methyl AA((3S)-1 -((1 ,2-dibromoethyl)sulfonyl)pyrrolidine-3-carbonyl)-N- methyl-L-valinate

To a solution of methyl N-methyl-N-((S)-1-(vinylsulfonyl)pyrrolidine-3-carbonyl)-L-v alinate (4.5 g, 13.54 mmol) in CCU (100 mL) at 0 °C was added Bra (2.77 mL, 54.15 mmol). The resulting solution was stirred for overnight and was then quenched by the addition of sat. NaHCOa (100 mL). The aqueous layer was extracted with EtOAc (3 x 200 mL) and the combined organic layers were washed with brine, dried with N32S04, filtered, and concentrated under reduced pressure. Purification by normal phase chromatography (25% EtOAc/ pet. ether) afforded the desired product (2.6 g, 39.0% yield). LCMS (ESI) m/z: [M + H] calcd for CuhtaBrzNpOsS: 492.99; found 493.0.

Step 3: Synthesis of methyl N-methyl-N-((S)-1-(((fi)-1-methylaziridin-2-yl)sulfonyl)pyrr olidine-3- carbonyl)-L-valinate and methyl N- methyl-N-((S)-1-(((S)-1-methylaziridin-2-yl)sulfonyl)pyrroli dine-3- carbonyl)-L-valinate

To a solution of methyl N-((3S)-1-((1 ,2-dibromoethyl)sulfonyl)pyrrolidine-3-carbonyl)-N-methyl-L- valinate (2.6 g, 5.28 mmol) in DMSO (250 mL) was added methanamine hydrochloride (1.07 g, 15.85 mmol) and EtaN (7.37 mL, 52.82 mmol). The reaction mixture was heated to 75 °C and stirred overnight. The mixture was then cooled to room temperature and diluted with EtOAc (1.5 L). The resulting mixture was washed with sat. NhLCI (2 x 200 mL) and brine (2 x 200 mL) and the organic layer was then concentrated under reduced pressure. Purification by reverse phase chromatography (40→60% MeCN/HzO) afforded a mixture of the desired products. The diastereomers were separated by prep-SFC (28% MeOH/COz) to afford methyl W-methyl-N-((S)-1-(((/7)-1-methylaziridin-2-yl)sulfonyl)pyrr olidine-3- carbonyl)-L-valinate (0.46 g, 24% yield) and methyl N-methyl-N-((S)-1 -(((S)-1 -methylaziridin-2- yl)sulfonyl)pyrrolidine-3-carbonyl)-L-valinate (0.35 g, 18.3% yield). LCMS (ESI) m/z: [M + H] calcd for C15H27N3O5S: 362.17; found 362.1.

Step 4: Synthesis of N-methyl-N-((S)-1-(((fl)-1-methylaziridin-2-yl)sulfonyl)pyrr olidine-3-carbonyl)-

L-valine

To a solution of methyl N-methyl-N-((S)-1-(((fl)-1-methylaziridin-2-yl)sulfonyl)pyrr olidine-3- carbonyl)-L-valinate (200.0 mg, 0.55 mmol) in THF (2.0 mL) and H 2 O (2.0 mL) at 0 °C was added LIOH (53.0 mg, 2.21 mmol). The resulting solution was stirred for 2 h at 0 °C and then the reaction mixture was acidified to pH 6 with 1 M HCI. The aqueous layer was extracted with EtOAc and the combined organic layers were washed with brine, dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by reverse phase chromatography (5→55% MeCN/H 2 O) afforded the desired product (110 mg, 57.2%, yield). LCMS (ESI) m/z : [M + H] calcd for CuHasNsOsS: 348.16; found 348.1.

Step 5: Synthesis of A/-methyl-W-((S)-1-(((S)-1-methylaziridin-2-yl)sulfonyl)pyrr olidine-3-carbonyl)-

L-valine

To a solution of methyl N-methyl-N-((S)-1-(((S)-1-methylaziridin-2-yl)sulfonyl)pyrro lidine-3- carbonyl)-L-valinate (200.0 mg, 0.55 mmol) in THF (2.0 mL) and H 2 O (2.0 mL) at 0 °C was added LiOH (53.0 mg, 2.21 mmol). The resulting solution was stirred for 2 h at 0 °C and then the reaction mixture was acidified to pH 6 with 1 M HCI. The aqueous layer was extracted with EtOAc and the combined organic layers were washed with brine, dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by reverse phase chromatography (5→55 MeCN/H 2 O) afforded the desired product (121 mg, 62.9%, yield). LCMS (ESI) m/z : [M + H] calcd for CuHasNsOsS: 348.16; found 348.1.

Intermediate A-23. Synthesis of (2S)-3-methyl-2-(1-oxo-7-((S)-1-tritylaziridine-2-carbonyl)- 2,7-dlazaspiro[4.4]nonan-2-yl)butanolc acid

Step 1: Synthesis of 1 -(tert-butyl) 3-methyl 3-allylpyrrolidine-1 ,3-dicarboxylate To a mixture of 1 -(tert-butyl) 3-methyl pyrrolidine-1 ,3-dicarboxylate (10 g, 43.616 mmol) in THF (100 mL) at -78 °C was added 1M LIHMDS (65.42 mL, 65.424 mmol) dropwise. The resulting mixture was stirred at -78 °C for 1 h and then a solution of allyl bromide (7.91 g, 65.423 mmol) in THF was added dropwise over 10 min. The resulting mixture was stirred at -78 °C for an additional 2 h and was then quenched by the addition of sat. NH4CI at 0 °C. The resulting mixture was extracted with EtOAc (3 x 100 mL) and the combined organic layers were washed with brine (2 x 80 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by silica gel column chromatography (20% EtOAc/pet. ether) afforded the desired product (10 g, 76% yield).

Step 2. Synthesis of 1 -(tert-butyl) 3-methyl 3-(2-oxoethyl)pyrrolidine-1 ,3-dicarboxylate To a mixture of 1 -(tert-butyl) 3-methyl 3-allylpyrrolidine-1 ,3-dicarboxylate (11.0 g, 40.84 mmol) and 2,6-lutidine (8.75 g, 81.68 mmol) in dioxane (190 mL) and H 2 O (19 mL) at 0 °C was added K20S04*2HZ0 (0.75 g, 2.04 mmol). The resulting mixture was stirred at 0 °C for 15 min and then NalO* (34.94 g, 163.36 mmol) was added in portions. The mixture was warmed to room temperature and stirred for an additional 3 h, then was quenched by the addition of sat. Na2S20aat 0 °C. The resulting mixture was extracted with EtOAc (3 x 300 mL) and the combined organic layers were washed with brine (200 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by reverse phase chromatography (0→40% MeCN/H 2 O, 0.1% HCOaH) afforded the desired product (6.4 g, 51% yield).

Step 3: Synthesis of 1 -(tert-butyl) 3-methyl 3-(2-(((S)-1-(benzyloxy)-3-methyl-1-oxobutan-2- yl)amino)ethyl)pyrrolidine-1 ,3-dicarboxylate

To a mixture of 1 -(tert-butyl) 3-methyl 3-(2-oxoethyl)pyrrolidine-1 ,3-dicarboxylate (6.30 g, 23.220 mmol) and benzyl L-valinate (7.22 g, 34.831 mmol) in MeOH (70 mL) at 0 °C was added ZnCIa (4.75 g, 34.831 mmol). The resulting mixture was warmed to room temperature and stirred for 30 min, then cooled to 0 °C. NaBHaCN (2.92 g, 46.441 mmol) was added in portions then the mixture was warmed to room temperature and stirred for 2 h. The reaction was quenched by the addition of sat. NhUCI at 0 °C and the resulting mixture was then extracted with EtOAc (3 x 200 mL). The combined organic layers were washed with brine (150 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by silica gel column chromatography (33% EtOAc/pet. ether) afforded the desired product (6.4 g, 53% yield). LCMS (ESI) m/z. [M + H] calcd for CasHaeNaOe: 463.28; found 463.3.

Step 4: Synthesis of tert-butyl 7-((S)-1-(benzyloxy)-3-methyl-1-oxobutan-2-yl)-6-oxo-2,7- diazaspiro[4.4]nonane-2-carboxylate

To a mixture of 1 -(tert-butyl) 3-methyl 3-(2-(((S)-1-(benzyloxy)-3-methyl-1-oxobutan-2- yl)amino)ethyl)pyrrolidine-1 ,3-dicarboxylate (4.50 g, 9.728 mmol) and DIPEA (16.6 mL, 97.28 mmol) in toluene (50 mL) was added DMAP (1.19 g, 9.728 mmol) and then mixture was heated to 80 °C. After 24 h the reaction was cooled to room temperature and concentrated under reduced pressure. Purification by reverse phase chromatography (15→60% MeCN/HzO, 0.1% HCOaH) afforded the desired product (3 g, 64% yield). LCMS (ESI) m/z. [M + H] calcd for C24H34N 2 O5: 431.26; found 431 .2.

Step 5: Synthesis of benzyl (2S)-3-methyl-2-(1-oxo-2,7-diazaspiro[4.4]nonan-2-yl)butanoa te

To a solution of tert-butyl 7-((S)-1-(benzyloxy)-3-methyl-1-oxobutan-2-yl)-6-oxo-2,7- diazaspiro[4.4]nonane-2-carboxylate (400.0 mg, 0.929 mmol) in DCM (3.0 mL) at 0 °C was added TFA (1.50 mL, 20.195 mmol) dropwise. The resulting mixture was stirred at 0 °C for 1 h and was then concentrated under reduced pressure. The TFA residue was further removed by azeotropic distillation with toluene three times to afford the desired product (400 mg, crude). LCMS (ESI) m/z. [M + H] calcd for C19H26N 2 O3: 331.20; found 331.1.

Step 6: Synthesis of benzyl (2S)-3-methyl-2-(1-oxo-7-((S)-1-tritylaziridine-2-carbonyl)- 2,7- diazaspiro[4.4]nonan-2-yl)butanoate

To a solution of benzyl (2S)-3-methyl-2-(1-oxo-2,7-diazaspiro[4.4]nonan-2-yl)butanoa te (400.0 mg, 1 .21 mmol) and DIPEA (2.06 mL, 12.11 mmol) in DMF (5 mL) at 0 °C was added (S)-1-tritylaziridine- 2-carboxylic acid (558.26 mg, 1.695 mmol) followed by COMU (673.55 mg, 1.574 mmol) in portions. The resulting mixture was stirred at 0 °C for 1 h and was then diluted with H 2 O (50 mL). The aqueous layer was extracted with EtOAc (3 x 20 mL) and the combined organic layers were washed with brine (20 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by prep-TLC (33% EtOAc/pet. ether) afforded the desired product (510 mg, 59% yield). LCMS (ESI) m/z. [M + H] calcd for C41H43N3O4: 642.34; found 642.3. Step 7: Synthesis of (2S)-3-methyl-2-(1 -oxo-7-((S)-1 -tritylaziridine-2-carbonyl)-2,7- diazaspiro[4.4]nonan-2-yl)butanoic acid

To a mixture of benzyl (2S)-3-methyl-2-(1 -oxo-7-((S)-1 -tritylaziridine-2-carbonyl)-2,7- diazaspiro[4.4]nonan-2-yl)butanoate (480.0 mg, 0.748 mmol) in toluene (35.0 mL) was added Pd/C 200.0 mg, 1.879 mmol). The resulting mixture was placed under an atmosphere of hfe (1 atm), heated to 50 °C and stirred for 3 h. The mixture was cooled to room temperature, filtered, the filter cake was washed with MeOH (3 x 10 mL), and the filtrate was concentrated under reduced pressure to afford the desired product (310 mg, 67% yield). LCMS (ESI) m/z. [M - H] calcd for C34H37N3O4: 550.27; found 550.3.

Intermediate A-24. Synthesis of (2S)-3-methyl-2-(1-oxo-7-((fl)-1-trltylazlrkilne-2-carbonyl& gt;- 2,7-dlazaspiro[4.4]nonan-2-yl)butanolcacld o

O o o

COMU, DIPEA N Pd/C, H,

BnO NH BnO N <¾ N

NTrt HO' N <¾

NTrt

DMF toluene

Step 1: Synthesis of benzyl (2S)-3-methyl-2-(1 -oxo-7-((fl)-1 -tritylaziridine-2-carbonyl)-2,7- diazaspiro[4.4]nonan-2-yl)butanoate

To a solution of benzyl (2S)-3-methyl-2-(1-oxo-2,7-diazaspiro[4.4]nonan-2-yl)butanoa te (400.0 mg, 1.21 mmol) and (fl)-1 -tritylaziridine-2-carboxylic acid (518.4 mg, 1.57 mmol) in DMF (4.0 mL) at 0 °C was added DIPEA (1.0 mL, 6.05 mmol) followed by COMU (621.7 mg, 1.45 mmol). The resulting mixture was stirred for 1 h and was then diluted with H 2 O (40 mL). The aqueous layer was extracted with EtOAc (3 x 15 mL) and the combined organic layers were washed with brine (2 x 10 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue was purified by prep-TLC (33% EtOAc/pet. ether) to afford the desired product (540 mg, 62% yield). LCMS (ESI) m/z. [M + H] calcd for C41H43N3O4: 642.33; found 642.4.

Step 2. Synthesis of (2S)-3-methyl-2-(1 -oxo-7-((fl)-1 -tritylaziridine-2-carbonyl)-2,7- diazaspiro[4.4]nonan-2-yl)butanoic acid

To a solution of benzyl (2S)-3-methyl-2-(1 -oxo-7-((fl)-1 -tritylaziridine-2-carbonyl)-2,7- diazaspiro[4.4]nonan-2-yl)butanoate (510.0 mg, 0.80 mmol) in toluene (30 mL) was added Pd/C (250.0 mg, 2.35 mmol). The resulting mixture was placed under a hydrogen atmosphere (1 atm), heated to 50 °C, and stirred for 3 h. The reaction was then cooled to room temperature, filtered, the filter cake was washed with MeOH (3 x 10 mL), and the filtrate was concentrated under reduced pressure to afford the desired crude product (330 mg). LCMS (ESI) m/z. [M + H] calcd for C34H37N3O4: 552.29; found 552.3.

Intermediate A-25 and A-26. Synthesis of benzyl (S)-3-methyl-2-((S)-1 -oxo-2,7- diazasplro[4.4]nonan-2-yl)butanoate and benzyl (S)-3-methyl-2-((fl)-1 -oxo-2,7- dlazasplro[4.4]nonan-2-yl)butanoate

Step 1: Synthesis of tert-butyl (fl)-7-((S)-1 -(benzyloxy)-3-methyl-1 -oxobutan-2-yl)-6-oxo-2,7- diazaspiro[4.4]nonane-2-carboxylate and tert-butyl (S)-7-((S)-1 -(benzyloxy)-3-methyl-1 -oxobutan-2-yl)-6- oxo-2, 7-diazaspiro[4.4]nonane-2-carboxylate

To a mixture of 1 -(tert-butyl) 3-methyl 3-(2-(((S)-1 -(benzyloxy)-3-methyl-1 -oxobutan-2- yl)amino)ethyl)pyrrolidine-1 ,3-dicarboxylate (4.50 g, 9.728 mmol) and DIPEA (16.6 mL, 97.28 mmol) in toluene (50 mL) was added DMAP (1.19 g, 9.728 mmol) and then mixture was heated to 80 °C. After 24 h the reaction was cooled to room temperature and concentrated under reduced pressure. Purification by reverse phase chromatography (10→50% MeCN/H 2 O, 0.1% HCOaH). The diastereomers were then separated by chiral prep-SFC (30% EtOH/COa) to afford tert-butyl (fl)-7-((S)-1 -(benzyloxy)-3-methyl-1 - oxobutan-2-yl)-6-oxo-2,7-diazaspiro[4.4]nonane-2-carboxylate (1.0 g, 32% yield, LCMS (ESI) m/z : [M +

H] calcd for C24H34N 2 O5: 431.26; found 431.2) and tert-butyl (S)-7-((S)-1 -(benzyloxy)-3-methyl-1 - oxobutan-2-yl)-6-oxo-2,7-diazaspiro[4.4]nonane-2-carboxylate carboxylate (1.0 g, 32% yield, LCMS (ESI) m/z: [M + H] calcd for C24H34N 2 O5: 431.26; found 431.2).

Step 2. Synthesis of benzyl (S)-3-methyl-2-((S)-1-oxo-2,7-diazaspiro[4.4]nonan-2-yl)buta noate To a solution of tert-butyl (5fl)-7-[(2S)-1 -(benzyloxy)-3-methyl-1 -oxobutan-2-yl]-6-oxo-2,7- diazaspiro[4.4] nonane-2-carboxylate (1.40 g, 3.25 mmol) in DCM (14 mL) at 0 °C was added TFA (5.0 mL, 67.3 mmol). The resulting mixture was stirred at 0 °C for 1 h and was then concentrated under reduced pressure. The mixture was diluted with H 2 O (20 mL) and was basified to pH 8 with sat. NaHCOa (aq.) at 0 °C. The resulting mixture was extracted with EtOAc (3 x 50 mL) and combined organic layers were washed with brine (40 mL), dried over NaaSCk filtered, and concentrated under reduced pressure to afford the desired product (1.4 g, crude). LCMS (ESI) m/z: [M + H] calcd for CigHaeNaOa: 331.20; found 331.2).

Step 3: Synthesis of benzyl (S)-3-methyl-2-((fl)-1-oxo-2,7-diazaspiro[4.4]nonan-2-yl)but anoate To a solution of tert-butyl (5S)-7-[(2S)-1 -(benzyloxy)-3-methyl-1 -oxobutan-2-yl]-6-oxo-2,7- diazaspiro[4.4] nonane-2-carboxylate (1.0 g, 2.3 mmol) in DCM (10 mL) at 0 °C was added TFA (4.0 mL, 53.9 mmol). The resulting mixture was stirred at 0 °C for 1 h and was then concentrated under reduced pressure. The mixture was diluted with H 2 O (10 mL) and was basified to pH 8 with sat. NaHCOa (aq.) at 0 °C. The resulting mixture was extracted with EtOAc (3 x 20 mL) and combined organic layers were washed with brine (20 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (1.0 g, crude). LCMS (ESI) m/z·. [M + H] calcd for CigHaeNaOa: 331.20; found 331.1). Intermediate A-27. Synthesis of (2S)-3-methyl-2-(1-oxo-7-((fl)-1-tritylaziridine-2-carbonyl) - 2,7-diazaspiro[4.4]nonan-2-yl)butanoic acid o

Step 1: Synthesis of benzyl (S)-3-methyl-2-((S)-1 -oxo-7-((fl)-1 -tritylaziridine-2-carbonyl)-2,7- diazaspiro[4.4]nonan-2-yl)butanoate

To a solution of benzyl (S)-3-methyl-2-((S)-1-oxo-2,7-diazaspiro[4.4]nonan-2-yl)buta noate (400 mg, 1.2 mmol) and DIPEA (1.1 mL, 6.1 mmol) in DMF (5.0 mL) at 0 °C was added (A)-1-tritylaziridine-2- carboxylic acid (480 mg, 1.5 mmol) and HATU (550 mg, 1.5 mmol). The resulting mixture was stirred for 1 h then purified by reverse phase chromatography (15→80% MeCN/HzO, 0.5% NH4HCO3) to afford the desired product (500 mg, 57% yield). LCMS (ESI) m/z. [M + H] calcd for C41H43N3O4: 642.34; found 642.3.

Step 2. Synthesis of (2S)-3-methyl-2-(1 -oxo-7-((f7)-1 -tritylaziridine-2-carbonyl)-2,7- diazaspiro[4.4]nonan-2-yl)butanoic acid

A solution of benzyl (S)-3-methyl-2-((S)-1-oxo-7-((fl)-1-tritylaziridine-2-carbon yl)-2,7- diazaspiro[4.4]nonan-2-yl)butanoate (450 mg, 0.70 mmol) and Pd/C (120 mg, 1.13 mmol) in toluene (30 mL) at 50 °C was stirred under a hydrogen atmosphere (1 atm). The mixture was stirred for 3 h and then was filtered, and the filter cake was washed with MeOH (3 x 30 mL). The filtrate was concentrated under reduced pressure to afford the desired product (430 mg, crude). LCMS (ESI) m/z. [M + H] calcd for C34H37N3O4: 552.29; found 552.3.

Intermediate 28. Synthesis of (S)-3-methyl-2-((fl)-1-oxo-7-((fl)-1-tritylaziridine-2-carbo nyl)- 2,7-diazaspiro[4.4]nonan-2-yl)butanoic ackJ

Step 1: Synthesis of benzyl (S)-3-methyl-2-((fl)-1-oxo-7-((fl)-1-tritylaziridine-2-carbo nyl)-2,7- diazaspiro[4.4]nonan-2-yl)butanoate

To a solution of benzyl (S)-3-methyl-2-((fl)-1-oxo-2,7-diazaspiro[4.4]nonan-2-yl)but anoate (500 mg, 1.5 mmol) and DIPEA (1.3 mL, 7.6 mmol) in DMF (7.0 mL) at 0 °C was added (/¾-1-tritylaziridine-2- carboxylic acid (550 mg, 1.7 mmol) and HATU (630 mg, 1.7 mmol). The resulting mixture was stirred for 1 h then purification by reverse phase chromatography (10→80% MeCN/H 2 O, 0.5% NH4HCO3) afforded desired product (700 mg, 64% yield) as an off-white solid. LCMS (ESI) m/z. [M + H] calcd for C41H43N3O4: 642.34; found 642.3.

Step 2. Synthesis of (S)-3-methyl-2-((fl)-1 -oxo-7-((fl)-1 -tritylaziridine-2-carbonyl)-2,7- diazaspiro[4.4]nonan-2-yl)butanoic acid A solution of benzyl (S)-3-methyl-2-((fl)-1 -oxo-7-((fl)-1 -tritylaziridine-2-carbonyl)-2,7- diazaspiro[4.4]nonan-2-yl)butanoate (650 mg, 0.70 mmol) and Pd/C (140 mg, 1.3 mmol) in toluene (30 mL) at 50 °C was stirred under a hydrogen atmosphere (1 atm). The mixture was stirred for 3 h and then was filtered, and the filter cake was washed with MeOH (3 x 30 mL). The filtrate was concentrated under reduced pressure to afford the desired product (550 mg, crude). LCMS (ESI) m/z. [M + H] calcd for C34H37N3O4: 552.29; found 552.3.

Intermediate A-29. Synthesis of (S)-2-((fl)-7-(ferf-butoxycarbonyl)-1-oxo-2,7- diazaspiro[4.4]nonan-2-yl)-3-methylbutanoic acid assumed assumed

To a solution of tert-butyl (fl)-7-((S)-1 -(benzyloxy)-3-methyl-1 -oxobutan-2-yl)-6-oxo-2,7- diazaspiro[4.4]nonane-2-carboxylate (600 mg, 1.4 mmol) in toluene (20 mL) was added Pd/C (120 mg,

1.1 mmol). The reaction mixture was heated at 50 °C and stirred under a hydrogen atmosphere (1 atm) for 3 h. The mixture was filtered, and the filter cake was washed with MeOH (3 x 20 mL). The filtrate was concentrated under reduced pressure to afford the desired product (550 mg, crude). LCMS (ESI) m/z. [M - H] calcd for C17H28N 2 O5: 339.19; found 339.1.

Intermediate A-30. Synthesis of (S)-2-((S)-7-(tert-butoxycarbonyl}-1-oxo-2,7- diazaspiro[4.4]nonan-2-yl)-3-methylbutanoic acid

Bn Pd/C, H 2

"Boc toluene 'BOC assumed assumed

To a solution of tert-butyl (S)-7-((S)-1 -(benzyloxy)-3-methyl-1 -oxobutan-2-yl)-6-oxo-2,7- diazaspiro[4.4]nonane-2-carboxylate (550 mg, 1.3 mmol) in toluene (30 mL) was added Pd/C (120 mg,

1.1 mmol). The reaction mixture was heated at 50 °C and stirred under a hydrogen atmosphere (1 atm) for 3 h. The mixture was filtered, and the filter cake was washed with MeOH (3 x 20 mL). The filtrate was concentrated under reduced pressure to afford the desired product (550 mg, crude). LCMS (ESI) m/z. [M - H] calcd for C17H28N 2 O5: 339.19; found 339.2.

Intermediate A-31. Synthesis of (Α)-3-ιηβΙΙιγΙ-2-(((5)-Λί-ιιιβΙΙιγΙ- Ι ΜΓηγΐ8ζΐΓΜΙηβ-2- carboxamido)methyl)butanoic acid

O O

NaH, Mel LIOH

THF THF Step 1: Synthesis of (fl)-3-methyl-2-(((S)-1-tritylaziridine-2-carboxamido)methyl )butanoic acid

To a solution of (S)-1 -tritylaziridine-2-carboxylic acid (1 g, 2.9 mmol) in DMF (10 mL) at 0 °C was added DIPEA (2.5 mL, 14.55 mmol) followed by COMU (1.12 g, 2.62 mmol). The resulting mixture was stirred for 20 min and (fl)-2-(aminomethyl)-3-methylbutanoic acid (382.0 mg, 2.91 mmol) was added. The resulting mixture was warmed to room temperature and stirred for an additional 2 h. The reaction mixture was then quenched with H 2 O and the aqueous layer was extracted with EtOAc. The combined organic layers were washed with brine, dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The crude residue was purified by reverse phase chromatography (30→70% MeCN/H20 + 0.1% NH4HCO3) to afford the desired product (850 mg, 63% yield). LCMS (ESI) m/z. [M - H] calcd for C28H30N 2 O3: 441.22; found 441.2.

Step 2. Synthesis of methyl (fl)-3-methyl-2-(((S)-1-tritylaziridine-2-carboxamido)methyl )butanoate

To a solution of (fl)-3-methyl-2-(((S)-N-methyl-1-tritylaziridine-2-carboxami do)methyl)butanoic acid (840.0 mg, 1.90 mmol) in MeOH (5.0 mL) at 0 °C was added TMSCHN 2 (10.0 mL, 0.45 mmol). The resulting mixture was warmed to room temperature and stirred for 2 h, at which point the reaction mixture was concentrated under reduced pressure. The residue was purified by reverse phase chromatography (30→80% MeCN/H20 + 0.1% NH4HCO3) to afford the desired product (450 mg, 52% yield). LCMS (ESI) m/z. [M - H] calcd for C29H32N 2 O3: 455.23; found 455.1.

Step 3: Synthesis of methyl (fl)-3-methyl-2-(((S)-N-methyl-1-tritylaziridine-2- carboxamido)methyl)butanoate

To a solution of methyl (fl)-3-methyl-2-(((S)-1-tritylaziridine-2-carboxamido)methyl )butanoate (440.0 mg, 0.96 mmol) in THF (5.0 mL) at 0 °C was added NaH (46.25 mg, 1.93 mmol). The resulting mixture was stirred for 30 min and then Mel (1.37 g, 9.65 mmol) was added. The resulting mixture was warmed to room temperature and stirred for an additional 4 h. The reaction mixture was then quenched with H 2 O and the aqueous layer was extracted with EtOAc (3 x 300 mL). The combined organic layers were washed with brine (3 x 200 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by reverse phase chromatography (10→90% MeCN/H 2 O + 0.1% NH4HCO3) to afford the desired product (340 mg, 75% yield). LCMS (ESI) m/z. [M + H] calcd for C30H34N 2 O3: 471.26; found 471.3.

Step 4: Synthesis of (fl)-3-methyl-2-(((S)-N-methyl-1 -tritylaziridine-2- carboxamido)methyl)butanoic acid

To a solution of methyl (fl)-3-methyl-2-(((S)-N-methyl-1-tritylaziridine-2- carboxamido)methyl)butanoate (340.0 mg, 0.72 mmol) in MeOH (3.0 mL) and H 2 O (3.0 mL) was added LiOH*H20 (242.5 mg, 5.78 mmol). The resulting mixture was stirred for 16 h at room temperature and was then acidified to pH 4 with KHSO4 (1 N). The resulting mixture was extracted with EtOAc (3 x 300 mL) and the combined organic layers were washed with brine (3 x 300 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by reverse phase chromatography (10→80% MeCN/H20 + 0.1% NH4HCO3) to afford the desired product (260 mg). LCMS (ESI) m/z. [M + H] calcd for C29H32N 2 O3: 455.23; found 455.1.

Intermediate A-32. Synthesis of N-methyl-W-(1-((fl)-1-tritylaziridine-2-carbonyl)piperidine- 4- carbonyl)-L-valine

Step 1: Synthesis of methyl W-methyl-N-(1 -((fl)-1 -tritylaziridine-2-carbonyl)piperidine-4-carbonyl)-

L-valinate

To a mixture of methyl N-methyl-N-(piperidine-4-carbonyl)-L-valinate (750 mg, 2.93 mmol) and (fl)-1 -tritylaziridine-2-carboxylic acid (1.13 g, 3.43 mmol) in DMF (7 mL) at 0 °C was added DIPEA (2.50 mL, 14.62 mmol) followed by HATU (2.20 g, 5.79 mmol) in portions. The resulting mixture was warmed to room temperature and stirred for 3 h. The reaction mixture was diluted with EtOAc (300 mL) and the mixture was washed with brine (2 x 150 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by normal phase chromatography (50% EtOAc/hexanes) afforded the desired product (1.5 g, 90.3% yield). LCMS (ESI) m/z: [M + H] calcd for C35H41N3O4: 568.32; found 568.3.

Step 2. Synthesis of N-methyl-AF(1-((fl)-1-tritylaziridine-2-carbonyl)piperidine- 4-carbonyl)-L-valine To a solution of methyl N-methyl-AF(1-((fl)-1-tritylaziridine-2-carbonyl)piperidine- 4-carbonyl)-L- valinate (500 mg, 0.881 mmol) in THF (5 mL) at 0 °C was added a solution of LiOH (111 mg, 2.64 mmol) in H 2 O (2.6 mL). The resulting mixture was warmed to room temperature and stirred for 4 h. The reaction mixture was diluted with H 2 O (300 mL) and acidified to pH 5 with 1 M HCI. The resulting mixture was extracted with DCM (3 x 100 mL) and the combined organic layers were washed with brine (2 x 150 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (600 mg, crude) which was used without further purification. LCMS (ESI) m/z: [M - H] calcd for C34H39N3O4: 552.29; found 552.3.

Intermediate A-33. Synthesis of A#-methyl-AF(1 -((S)-1 -tritylazlridlne-2-carbonyl)plperidlne-4- carbonyl)-L-vallne

Step 1: Synthesis of methyl N-methyl-N-(1 -((S)-1 -tritylaziridine-2-carbonyl)piperidine-4-carbonyl)- L-valinate

To a mixture of methyl N-methyl-N-(piperidine-4-carbonyl)-L-valinate (0.90 g, 3.511 mmol) and (S)-1 -tritylaziridine-2-carboxylic acid (2.31 g, 7.022 mmol) in DMF (10 mL) at 0 °C was added DIPEA (3.06 mL, 17.57 mmol) and HATU (2.67 g, 7.022 mmol). The resulting mixture was warmed to room temperature and stirred for 2 h. The reaction mixture was diluted with EtOAc (50 mL) and the mixture was washed with H 2 O, brine (100 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by normal phase chromatography (100% EtOAc) afforded the desired product (1.47 g, 73.7% yield). LCMS (ESI) m/z: [M + H] calcd for C35H41N3O4: 568.32; found 568.3.

Step 2. Synthesis of AFmethyl-W-(1-((S)-1-tritylaziridine-2-carbonyl)piperidine-4 -carbonyl)-L-valine To a solution of methyl AFmethyl-AF(1-((S)-1-thtylazihdine-2-carbonyl)pipehdine-4-ca rbonyl)-L- valinate (1.0 g, 1.76 mmol) in THF (15 mL) at 0 °C was added a solution of LiOH (370 mg, 8.80 mmol) in H 2 O (15 mL). The resulting mixture was warmed to room temperature and stirred for 3 h. The reaction mixture was acidified to pH 6 with 1 M HCI. The aqueous layer was extracted with EtOAc (2 x 50 mL) and the combined organic layers were dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure to adfford the desired product (1.33 g, crude) which was used without further purification. LCMS (ESI) m/z. [M + H] calcd for C34H39N3O4: 554.30; found 554.3.

Intermediate A-34. Synthesis of sodium (fl)-1-methyl-5-(1-tritylaziridine-2-carboxamldo)- 1 f#-imidazole-2-cartx)xylate

Step 1: Synthesis of methyl 5-amino-1 -methyl-1 /^-imidazole-2-carboxylate To a mixture of methyl 1 -methyl-5-nitro-1 H-imidazole-2-carboxylate (1.0 g, 5.401 mmol) in MeOH (15 mL) was added Pd/C (500 mg). The resulting mixture was placed under an atmosphere of h½ (1 atm) and stirred for 3 h. The mixture was filtered, the filter cake was washed with MeOH (3 x 20 mL), and the filtrate was concentrated under reduced pressure to afford the desired product (1.0 g, crude). LCMS (ESI) m/z: [M + H] calcd for CeHgNsOa: 156.08; found 156.1.

Step 2. Synthesis of methyl (R)- 1 -methyl-5-(1 -tritylaziridine-2-carboxamido)-1 /-/-imidazole-2- carboxylate

To a mixture of (fl)-1-tritylaziridine-2-carboxylic acid (2.55 g, 7.741 mmol) in DCM (12.0 mL) at 0 °C was added a solution of isobutyl chloroformate (845.06 mg, 6.187 mmol) and N-methylmorpholine (1.04 g, 10.282 mmol) in DCM in portions over 30 min. To the resulting mixture was added methyl 5- amino-1 -methyl-1 H- imidazole-2-carboxylate (800.0 mg, 5.156 mmol). The mixture was stirred at room temperature overnight then diluted with DCM (300 mL) and washed with H 2 O (3 x 100 mL). The organic layer was washed with brine (2 x 150 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by silica gel column chromatography (25% EtOAc/hexanes) afforded the desired product (1.2 g, 49.9% yield). LCMS (ESI) m/z : [M + H] calcd for CaeHaeN^s: 467.21 ; found 467.2.

Step 3: Synthesis of sodium (fl)-1-methyl-5-(1-tritylaziridine-2-carboxamido)-1 /-/-imidazole-2- carboxylate

To a mixture of methyl (fl)-1-methyl-5-(1-tritylaziridine-2-carboxamido)-1 H- imidazole-2- carboxylate (300 mg, 0.643 mmol) in THF (3 mL) was added a solution of NaOH (38.58 mg, 0.965 mmol) in H 2 O. The resulting mixture was stirred for 2 h and then concentrated under reduced pressure to afford the desired product (400 mg, crude). LCMS (ESI) m/z: [M + H] calcd for C27H24N4O3: 453.19; found 453.2.

Intermediate A-35. Synthesis of (S)-1-methyl-5-(1-tritylaziridine-2-carboxamido)-1H- imidazole-2-carboxylic acid o ,

M Trt

HO Jr Trt sobutyl chloroformate q t

I q

N N' W-m ethyl morpholine NaOH

MeO V I - NH ® "" 1 MeO. o ’ DCM n ΝΗ » HO.

MeOH, H 2 0

O ' O * Step 1: Synthesis of methyl (S)-1-methyl-5-(1-tritylaziridine-2-carboxamido)-1 H-imidazole-2- carboxylate

To a mixture of (S)-1 -tritylaziridine-2-carboxylic acid (1.18 g, 3.577 mmol) in DCM (15 mL) at 0 °C was added isobutyl chloroformate (423.41 mg, 3.100 mmol) and N-methylmorpholine (0.39 mL, 3.862 mmol) dropwise. The resulting mixture was stirred at 0 °C for 1 h then methyl 5-amino-1 -methyl-1 H- imidazole-2-carboxylate (370.0 mg, 2.385 mmol) was added. The mixture was warmed to room temperature and stirred overnight. The reaction was quenched with sat. NaHCOs at 0 °C and the resulting mixture was extracted with DCM (2 x 100 mL). The combined organic layers were washed with brine (150 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by silica gel column chromatography (100% EtOAc) afforded the desired product (380 mg, 34.2% yield). LCMS (ESI) m/z·. [M + H] calcd for C28H26N403: 467.21 ; found 467.3.

Step 2. Synthesis of (S)-1 -methyl-5-(1 -tritylaziridine-2-carboxamido)-1 H- imidazole-2-carboxylic acid

To a mixture of methyl (S)-1 -methyl-5-(1 -tritylaziridine-2-carboxamido)-1 W-imidazole-2- carboxylate (380.0 mg, 0.815 mmol) in MeOH (5 mL) at 0 °C was added NaOH (146.60 mg, 3.665 mmol) in H 2 O (3.6 mL) dropwise. The resulting mixture was warmed to room temperature and stirred for 6 h then was acidified to pH 6 with 1 M HCI. The resulting mixture was extracted with DCM (2 x 100 mL), and the combined organic layers were washed with brine (150 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (350 mg, crude). LCMS (ESI) m/z: [M - H] calcd for C27H24N4O3: 451.17; found 451.1.

Intermediate A-36 and A-37. Synthesis of (fl)-N- methyl-N- ((1 -methylaziridin-2- yl)sulfonyl)glycine and (S)-AAmethyl-AA((1-methylazlrldln-2-yl)sulfonyl)glycine

BnBr, KJCOJ TFA S0 2 CI 2 , Et j N

ΗΟ γ'γ β ~ Boc Br,

BnO acetone rr DCM rr MeCN rr * DCM

Step 1: Synthesis of benzyl N-(tert-butoxycarbonyl)-N- methylglycinate

To a stirred mixture of [(tert-butoxycarbonyl)(methyl)amino]acetic acid (15. g, 79.28 mmol) in acetone (150 mL) was added BnBr (14.14 mL, 82.70 mmol) and K2CO3 (21.91 g, 158.55 mmol) in portions at 0 °C . The resulting mixture was stirred for 4 h at room temperature. The resulting mixture was filtered, the filter cake was washed with acetone (3 x 100 mL), and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (33% EtOAc/pet. ether) to afford the desired product (15.2 g, 68.6% yield). LCMS (ESI) m/z: [M + Na] calcd for C15H21NO4: 302.14; found 302.0.

Step 2: Synthesis of benzyl methylglycinate To a stirred solution of benzyl N-(tert-butoxycarbonyl)-N-methylglycinate (10.0 g, 35.80 mmol) in DCM (100 mL) was added TFA (50 mL) dropwise at 0 °C. The resulting mixture was stirred for 1 h at 0 °C and then the resulting mixture was concentrated under reduced pressure to afford the desired product (7.80 g, crude). LCMS (ESI) m/z. [M + H] calcd for C10H13NO2: 180.10; found 179.1.

Step 3: Synthesis of benzyl N-methyl-N-(vinylsulfonyl)glycinate

To a solution of benzyl methylglycinate (15.60 g, 87.04 mmol) and EfaN (36.4 mL, 261.1 mmol) in MeCN (300 mL) at -70 °C was added a solution of 2-chloroethanesulfonyl chloride (17.03 g, 104.47 mmol) in MeCN (150 mL). The resulting mixture was warmed to room temperature and stirred for 20 min. The reaction mixture was cooled -50 °C and additional EtsN (36.4 mL, 261.1 mmol) was added to reaction mixture. The reaction mixture was warmed to room temperature and stirred for 1 h. The reaction was then quenched with H 2 O at 0 °C. The mixture was acidified to pH 6 with 1 M HCI aq and was then extracted with DCM (800 mL), dried over Na2SO<, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (33% EtO Ac/pet. ether) to afford the desired product (7.53 g, 32.1% yield). LCMS (ESI) m/z. [M + H 2 O] calcd for C12H15NO4S: 287.08; found 287.2.

Step 4: Synthesis of benzyl N-((1 ,2-dibromoethyl)sulfonyl)-N-methylglycinate To a solution of benzyl N-methyl-N-(vinylsulfonyl)glycinate (5.58 g, 20.7 mmol) in DCM (50 mL) at -20 °C was added a solution of BT2 (1.06 mL, 6.64 mmol) in DCM (10 mL). The resulting mixture was warmed to room temperature and stirred overnight. The reaction mixture was then cooled to 0 °C and quenched with sat. aq. Na2S20a (30 mL). The resulting mixture was washed with sat. aq. Na2HCOs and then extracted with DCM (2 x 200 mL), the combined organic layers dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (33% EtOAc/pet. ether) to afford the desired product (5.1 g, 57.4% yield). LCMS (ESI) m/z. [M + H 2 O] calcd for Ci2HisBr2N04S: 444.92; found 444.9.

Step 5: Synthesis of benzyl (fl)-N-methyl-N- ((1-methylaziridin-2-yl)sulfonyl)glycinate and benzyl (S)-N-methyl-AA((1 -methylaziridin-2-yl)sulfonyl)glycinate

To a stirred solution of benzyl ΛΑ((1 ,2-dibromoethyl)sulfonyl)-N-methylglycinate (7.20 g, 16.78 mmol) and methylamine hydrochloride (3.39 g, 50.2 mmol) in DMSO (750 mL) was added EtsN (23.32 mL, 230.47 mmol). The resulting mixture was stirred for 2 h at room temperature then heated to 75 °C and stirred overnight. The reaction mixture was cooled to room temperature and extracted with EtO Ac (2 x 1000 mL). The combined organic layers were washed with H 2 O (1500 mL) and brine (1500 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (EtOAc) to afford a mixture of diastereomers. The diastereomers were separated by prep-SFC (10% EtOH/Hex) to afford benzyl (fl)-N-methyl-N-((1-methylaziridin-2-yl)sulfonyl)glycinate (500 mg, 31.3% yield) and benzyl (S)-N-methyl-N-((1-methylaziridin-2-yl)sulfonyl)glycinate (600 mg, 37.5% yield). LCMS (ESI) m/z. [M + H] calcd for CisHieN 2 04S: 299.11 ; found 299.0.

Step 6: Synthesis of (fl)-N- methyl-N-((1-methylaziridin-2-yl)sulfonyl)glycine A suspension of benzyl (fl)-N-methyl-N-((1-methylaziridin-2-yl)sulfonyl)glycinate (300.0 mg) and Pd(OH)2/C (150.0 mg) in THF at room temperature was stirred under an atmosphere of hydrogen (1 atm) for 3 h. The mixture was filtered, the filter cake was washed with MeOH (3 x 20 mL), and the filtrate was concentrated under reduced pressure to afford the desired product (206 mg, crude). LCMS (ESI) m/z. [M + H] calcd for C6H12N 2 O4S: 209.06; found 209.0.

Step 7: Synthesis of (S)-N-methyl-/\A((1-methylaziridin-2-yl)sulfonyl)glycine A suspension of benzyl (fl)-AAmethyl-N-((1-methylaziridin-2-yl)sulfonyl)glycinate (300.0 mg, 1.01 mmol) and Pd(OH)a/C (150.0 mg) in THF at room temperature was stirred under an atmosphere of hydrogen (1 atm) for 3 h. The mixture was filtered, the filter cake was washed with MeOH (3 x 20 mL), and the filtrate was concentrated under reduced pressure to afford the desired product (216 mg, crude). LCMS (ESI) m/z: [M + H] calcd for CeHiaNa04S: 209.06; found 209.1.

Intermediate A-38. Synthesis of (2¾3S)-1-(ferH»utylsulfinyl)-3-cyclopropylaziridine-2- carboxylic acid

Step 7: Synthesis of (£)-N-(cydopropylmethylene)-2-methylpropane-2-sulfinamide To a suspension of (S)-2-methylpropane-2-sulfinamide (4.0 g, 33.0 mmol) and CuSO* n (15.80 g, 99.01 mmol) in DCM (200.0 mL) was added cyclopropanecarbaldehyde (4.63 g, 66.0 mmol). The resulting mixture was stirred overnight and was then filtered, the filter cake was washed with DCM (3 x 100 mL), and the filtrate was concentrated under reduced pressure to afford the desired product (3.5 g,

61.2% yield). LCMS (ESI) m/z : [M + H] calcd for CeHisNOS: 174.10; found 174.1.

Step 2: Synthesis of ethyl (2S,3S)-1 -(tert-butylsulfinyl)-3-cyclopropylaziridine-2-carboxylate To a solution of ethyl bromoacetate (481.91 mg, 2.886 mmol) in THF (5.0 mL) at -78 °C was added LiHMDS (2.90 mL, 2.90 mmol). The resulting mixture was stirred for 2 h at -78 °C and then a solution of (E)-N-(cyclopropylmethylene)-2-methylpropane-2-sulfinamide (250.0 mg, 1.443 mmol) was added. The resulting mixture was stirred for 2 h at -78 °C and was then was then quenched with H 2 O at 0 °C. The aqueous layer was extracted with EtOAc (3 x 50 mL), and the combined organic layers were dried over Na 2 SO 4 . filtered, and concentrated under reduced pressure. The residue was purified by prep- TLC (17% EtOAc/pet. ether) to afford the desired product (250 mg, 66.8% yield). LCMS (ESI) m/z: [M +

H] calcd for CiaHaiNOaS: 260.13; found 260.1.

Step 3: Synthesis of (2S,3S)-1 -(fe/1-butylsulfinyl)-3-cyclopropylaziridine-2-carboxylic acid A solution of ethyl (2S,3S)-1-(tert-butylsulfinyl)-3-cyclopropylaziridine-2-carb oxylate (500.0 mg,

1.928 mmol) in THF (2.0 mL) and H 2 O (2.0 mL) at 0 °C was added LiOHeH 2 O (121.34 mg, 2.89 mmol). The reaction mixture was stirred for 1 h and was then acidified to pH 6 with 1 M HCI (aq.). The resulting mixture was extracted with EtOAc (2 x 10 mL) and the combined organic layers were washed with brine (10 mL), dried over Na 2 SO 4 , filtered, and the filtrate was concentrated under reduced pressure to afford the desired product (400 mg, 89.7% yield). LCMS (ESI) m/z: [M + H] calcd for C10H17NO3S: 232.10; found 232.0. Intermediate A-39. Synthesis of (2/?,3fl)-3-(methoxymethyl)-1-tritylaziridlne-2-carboxylic acid

MSNH 2 EtO ΟΜβ

PPh*. AD -mix-beta ΟΜο SOCI,

EtOOC EKXXr^V^OMe

HOAc, MeOH t-BuOH/Η,Ο f ¾ Η DCM κ toL

Step 1: Synthesis of ethyl ( E)-4-methoxybut-2-enoate

To a solution of ethyl but-2-ynoate (10.0 g, 89.18 mmol) in MeOH (8.80 mL, 118.594 mmol) and HOAc (1.05 mL, 18.3 mmol) was added a solution of PPha (1.20 g, 4.58 mmol) in toluene (60.0 mL). The resulting solution heated to 110 °C and stirred overnight. The reaction mixture was cooled to room temperature and was then diluted with HzO (60 mL). The resulting solution was extracted with EtO Ac (2 x 60), and the combined organic layers were washed with brine (2 x 20 mL), dried over NazSO*, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (9% EtOAc/pet. ether) to afford the desired product (4.9 g, 38.1% yield). LCMS (ESI) m/z. [M + H] calcd for C7H12O3: 145.09; found 144.9.

Step 2: Synthesis of ethyl (2S,3fl)-2,3-dihydroxy-4-methoxybutanoate To a solution of ethyl (£)-4-methoxybut-2-enoate (5.0 g, 34.68mmol), and methanesulfonamide (3.30 g, 34.68 mmol) in f-BuOH (150.0 mL) and H 2 O (100.0 mL) was added AD-mix-β (48.63 g, 62.43 mmol). The resulting solution was heated to 30 °C and stirred overnight. The solution was then cooled to room temperature and adjusted to pH 2 with KHSO4. The resulting solution was extracted with EtO Ac (2 x 100 mL) and the combined organic layers were dried over NazS04, filtered, and concentrated under reduced pressure to afford the desired product (1.28 g, crude). LCMS (ESI) m/z. [M + H] calcd for C7H14O5: 179.09; found 179.0.

Step 3: Synthesis of ethyl (4S,5fl)-5-(methoxymethyl)-1 ,3,2-dioxathiolane-4-carboxylate 2-oxide To a solution of ethyl (2S,3fl)-2,3-dihydroxy-4-methoxybutanoate (4.10 g, 23.01 mmol) in DCM (20.0 mL) at 0 °C was added SOCIz (5.47 g, 45.9 mmol). The resulting mixture was heated to 50 °C and stirred for 3 h. The reaction mixture was then cooled to room temperature and concentrated under reduced pressure to afford the desired product (4.0 g, crude).

Step 4: Synthesis of ethyl (2/?,3S)-2-azido-3-hydroxy-4-methoxybutanoate To a solution of ethyl (4S,5fl)-5-(methoxymethyl)-1 ,3,2-dioxathiolane-4-carboxylate 2-oxide (4.0 g crude, 17.84 mmol) in DMF (20.0 mL) at 0 °C was added NaNa (5.80 g, 89.22 mmol). The resulting mixture was heated to 35 °C and stirred overnight. The reaction mixture was then diluted with HzO (200 mL) and was extracted with EtO Ac (3 x 50 mL). The combined organic layers were washed with brine (3 x 50 mL), dried over NazS04, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (17% EtOAc/pet. ether) to afford the desired product (1 .0 g, 27.6% yield). LCMS (ESI) m/z. [M + H] calcd for C7H13N3O4: 204.10; found 204.0.

Step 5: Synthesis of ethyl (2/?,3fl)-3-(methoxymethyl)aziridine-2-carboxylate To a solution of ethyl (2f?,3S)-2-azido-3-hydroxy-4-methoxybutanoate(1.0 g, 4.92 mmol) in DMF (10 mL) at 0 °C was added PPha (1.29 g, 4.92 mmol) in portions over 30 min. The reaction solution was then warmed to room temperature and stirred for 30 min. The reaction mixture was then heated to 85 °C and stirred until the reaction was complete. The reaction mixture was then concentrated under reduced pressure and purified by prep-TLC (33% EtOAc/pet. ether) to afford the desired product (480 mg, 61.3% yield). LCMS (ESI) m/z: [M + H] calcd for C7H13NO3: 160.10; found 160.1.

Step 6: Synthesis of ethyl (2R,3fl)-3-(methoxymethyl)-1 -tritylaziridine-2-carboxylate To a solution of ethyl (2fl,3fl)-3-(methoxymethyl)aziridine-2-carboxylate (480.0 mg, 3.02 mmol) and EtsN (2.1 mL, 15.0 mmol) in DCM (10 mL) at 0 °C was added Trt-CI (1.681 g, 6.031 mmol). The resulting mixture was warmed to room temperature and stirred for 2 h. The mixture was concentrated then concentrated under reduced pressure and the residue was purified by prep-TLC (5% EtOAc/pet. ether) to afford the desired product (700 mg, crude).

Step 7: Synthesis of (2fl,3fl)-3-(methoxymethyl)-1-tritylaziridine-2-carboxylic acid To a solution of ethyl (2 R, 3f?)-3-(methoxymethyl)-1-(triphenylmethyl)aziridine-2-carbox ylate (200.0 mg, 0.498 mmol) in THF (5.0 mL) and H 2 O (5 mL) was added LiOHeH 2 O (41.81 mg, 0.996 mmol). The resulting solution was stirred at room temperature for 24 h. The mixture was then diluted with H 2 O (10 mL) and extracted with EtOAc (20 mL). The aqueous layer was then acidified to pH 7 with sat. aq. NH4CI and extracted with EtOAc (2 x 10 mL). The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (60 mg, 32.3% yield). LCMS (ESI) m/z: [M - H] calcd for C24H23NO3: 372.16; found 372.1.

Intermediate A-40. Synthesis of (2¾3S)-1-(tert-butylsulflnyl)-3-(4-methoxyphenyl)azlrldlne- 2-carboxylic acid

Step 1: (£)-N-(4-methoxybenzylidene)-2-methylpropane-2-sulfinamide A solution of (S)-2-methylpropane-2-sulfinamide (2.50 g) and anisaldehyde (2.81 g) in Ti(OEt)4 (20.0 mL) was stirred at 70 °C for 1 h. The resulting mixture was cooled to room temperature, diluted with

EtOAc (60 mL), and then poured into H 2 O. The mixture was filtered, and the filter cake was washed with EtOAc (3 x 50 mL). The resulting mixture was extracted with EtOAc (3 x 50 mL) and the combined organic layers were washed with brine (50 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (25% EtOAc/pet. ether) to afford the desired product (4 g, 81.0% yield). LCMS (ESI) m/z: [M + H] calcd for C12H17NO2S: 240.11 ; found 240.1.

Step 2: Synthesis of ethyl (2S,3S)-1-(tert-butylsulfinyl)-3-(4-methoxyphenyl)aziridine- 2- carboxylate

To a solution of ethyl 2-bromoacetate (5.60 g, 33.5 mmol) in THF (100 mL) at -78 °C was added LiHMDS (1 M in THF, 34 mL, 33.473 mmol). After 30 min a solution of (E)-N-(4-methoxybenzylidene)-2- methylpropane-2-sulfinamide (4 g, 16.74 mmol) in THF (20 mL) was added. The resulting mixture was stirred at -78 °C for additional 3 h. The reaction was then quenched with sat. aq. NhLCI. The mixture was extracted with EtOAc (3 x 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. The residue was purified by silica gel chromatography (25% EtOAc/pet. ether) to afford the desired product (2.7 g, 49.6% yield). LCMS (ESI) m/z: [M + H] calcd for CieHasNO+S: 326.14; found 326.1.

Step 3: Synthesis of (2S,3S)-1-(tert-butylsulfinyl)-3-(4-methoxyphenyl)aziridine- 2-carboxylic acid To a solution of ethyl (2S,3S)-1-(tert-butylsulfinyl)-3-(4-methoxyphenyl)aziridine- 2-carboxylate (80 0.0 mg, 2.68 mmol) in THF (2.0 mL) at 0 °C was added a solution of LiOHeH 2 O (309.46 mg, 7.38 mmol) i n H 2 O (3.0 mL). The resulting mixture was warmed to room temperature and stirred for 4 h. The mixture w as then acidified to pH 6 with sat. aq. NH4CI and then extracted with EtOAc (3 x 50 mL). The combined or ganic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desi red product (690 mg, 94.4% yield). LCMS (ESI) m/z: [M - H] calcd for C14H19NO4S: 296.10; found 296.2.

Intermediate A-41. Synthesis of (2S,3fl)-3-(4-methoxyphenyl)azlrldine-2-carboxyllc acid

OMe OMe

MsNH 2

AD-mix-alpha // \ NeCI, E¾N (/ \ TMSNs, TBAF

EtOOC EtOOC

Etooc'' 's%%/ ^^ t-BuOH/H 2 0 DCM THF

HO OH NeO ¾>H

OMe

EtOOC

Nj 'OH

Step i: Synthesis of ethyl (2/?,3S)-2,3-dihydroxy-3-(4-methoxyphenyl)propanoate

To a solution of ethyl p-methoxycinnamate (5.0 g, 24.24 mmol) in IBuOH (70.0 mL) and H 2 O (70.0 mL) at 0 °C was added AD-mix-a (33.80 g, 43.39 mmol) and methanesulfonamide (2.31 mg, 0.024 mmol). The resulting mixture was warmed to room temperature and stirred overnight. The reaction was then cooled to 0 °C and quenched with KHSOA (aq.). The mixture was extracted with EtOAc (3 x 100 mL) and the combined organic layers were washed with brine (2 x 90 mL), dried over Na 2 SO 4 . filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (50% EtOAc/pet. ether) to afford the desired product (5.7 g, 88.1% yield).

Step 2: Synthesis of ethyl (2/?,3S)-3-hydroxy-3-(4-methoxyphenyl)-2-(((4- nitrophenyl)sulfonyl)oxy)propanoate

To a solution of ethyl (2f?,3S)-2,3-dihydroxy-3-(4-methoxyphenyl)propanoate (3.0 g, 12.49 mmol) and EtaN (0.174 mL, 1.249 mmol) in DCM (30.0 mL) at 0 °C was added 4-nitrobenzenesulfonyl chloride (2.76 g, 12.49 mmol). The resulting mixture was stirred for 1 h and was then diluted with H 2 O. The mixture was extracted with DCM (3 x 100 mL) and the combined organic layers were washed with brine (2 x 100 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by prep-TLC (50% EtOAc/pet. ether) to afford the desired product (3.8 g, 68.0% yield). LCMS (ESI) m/z: [M + Na] calcd for CieHigNOgS: 448.07; found 448.2.

Step 3: Synthesis of ethyl (2S,3S)-2-azido-3-hydroxy-3-(4-methoxyphenyl)propanoate

To a solution of ethyl (2fl,3S)-3-hydroxy-3-(4-methoxyphenyl)-2-(((4- nitrophenyl)sulfonyl)oxy)propanoate (1.20 g, 2.82 mmol) in THE at 0 °C was added TBAF (1 M in THF,

5.64 mL, 5.64 mmol) and TMSNs (648.79 mg, 5.64 mmol). The resulting mixture was heated to at 60 °C and stirred for 16 h. The reaction was then cooled to at 0 °C and quenched with sat. aq. NhUCI. The mixture was extracted with EtOAc (3 x 100 mL) and the combined organic layers were washed with H 2 O (2 x 100 mL), dried over Na 2 SO 4 . filtered, and concentrated under reduced pressure. The residue was purified by prep-TLC (33% EtOAc/pet. ether) to afford the desired product (540 mg, 70.7% yield).

Step 4: Synthesis of ethyl (2S,3fl)-3-(4-methoxyphenyl)aziridine-2-carboxylate

To a solution of ethyl (2S,3S)-2-azido-3-hydroxy-3-(4-methoxyphenyl)propanoate (440.0 mg,

1.659 mmol) in DMF was added PPhs (522.06 mg, 1.99 mmol). The resulting mixture was stirred at room temperature for 30 min and was then heated to 80 °C and stirred overnight. The mixture was then extracted with EtOAc (3 x 100 mL), and the combined organic layers were washed with H 2 O (2 x 100 mL), dried over anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by prep-TLC (25% EtOAc/pet. ether) to afford the desired product (200 mg, 51.8% yield). LCMS (ESI) m/z : [M + H] calcd for C12H15NO3: 222.12; found 222.1.

Step 5: Synthesis of (2S,3fl)-3-(4-methoxyphenyl)aziridine-2-carboxylic acid To a solution of ethyl (2S,3fl)-3-(4-methoxyphenyl)aziridine-2-carboxylate (200.0 mg, 0.904 mmol) in MeOH and H 2 O at 0 °C was added LiOHeH 2 O (86.6 mg, 3.62 mmol). The resulting mixture was stirred for 1 h and was then neutralized to pH 7 with HCI (aq.). The mixture was extracted with EtOAc (3 x 100 mL) and the combined organic layers were washed with H 2 O (2 x 100 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (180 mg, 97.9% yield). LCMS (ESI) m/z: [M - H] calcd for C10H11NO3: 192.07; found 192.0.

Intermediate A-42. Synthesis of (2/?,3S)-3-(4-methoxyphenyl)azlridine-2-carboxylic acid

OMe OMe

OMe WSNH 2 // \ NsCI, // \

-JJ AD-mlx-beta Et j N TMSNg, TBAF

EtOOC EtOOC

EtOOC t-BuOH/H 2 0 DCM THF

Hd OH NsO OH

Step /. Synthesis of ethyl (2S,3fl)-2,3-dihydroxy-3-(4-methoxyphenyl)propanoate

To a solution of ethyl p-methoxycinnamate (5.0 g, 24.24 mmol) in /BuOH (70.0 mL) and H 2 O (70.0 mL) at 0 °C was added AD-mix-B (33.80 g, 43.39 mmol) and methanesulfonamide (2.31 mg, 0.024 mmol). The resulting mixture was warmed to room temperature and stirred overnight. The reaction was then cooled to 0 °C and quenched with KHSO* (aq.). The mixture was extracted with EtOAc (3 x 100 mL) and the combined organic layers were washed with brine (2 x 90 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (50% EtOAc/pet. ether) to afford the desired product (5.7 g, 88.1 % yield).

Step 2: Synthesis of ethyl (2S,3fl)-3-hydroxy-3-(4-methoxyphenyl)-2-(((4- nitrophenyl)sulfonyl)oxy)propanoate

To a solution of ethyl (2S,3fl)-2,3-dihydroxy-3-(4-methoxyphenyl)propanoate (5.80 g, 24.14 mmol) and EtaN (10.1 mL, 72.42 mmol) in DCM (30.0 mL) at 0 °C was added 4-nitrobenzenesulfonyl chloride (5.34 g, 24.1 mmol). The resulting mixture was stirred for 1 h and was then diluted with H 2 O. The mixture was extracted with DCM (3 x 100 mL) and the combined organic layers were washed with brine (2 x 100 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by prep-TLC (50% EtOAc/pet. ether) to afford the desired product (7.2 g, 67.0% yield). LCMS (ESI) m/z: [M + H] calcd for CieHigNOgS: 426.09; found 426.2.

Step 3: Synthesis of ethyl (2/?,3fl)-2-azido-3-hydroxy-3-(4-methoxyphenyl)propanoate To a solution of ethyl (2S,3fl)-3-hydroxy-3-(4-methoxyphenyl)-2-(((4- nitrophenyl)sulfonyl)oxy)propanoate (5.0 g, 11.75 mmol) in THE at 0 °C was added TBAF (1 M in THE, 23.5 mL, 23.51 mmol) and TMSNs (2.7 g, 23.5 mmol). The resulting mixture was heated to at 60 °C and stirred for 16 h. The reaction was then cooled to at 0 °C and quenched with sat. aq. NhLCI. The mixture was extracted with EtOAc (3 x 100 mL) and the combined organic layers were washed with H 2 O (2 x 100 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by prep-TLC (33% EtOAc/pet. ether) to afford the desired product (2.3 g, 70.1% yield).

Step 4: Synthesis of ethyl (2/?,3S)-3-(4-methoxyphenyl)aziridine-2-carboxylate To a solution of ethyl (2f?,3fl)-2-azido-3-hydroxy-3-(4-methoxyphenyl)propanoate (2.30 g, 8.67 mmol) in DMF was added PPha (2.73 g, 10.4 mmol). The resulting mixture was stirred at room temperature for 30 min and was then heated to 80 °C and stirred overnight. The mixture was then extracted with EtOAc (3 x 100 mL), and the combined organic layers were washed with H 2 O (2 x 100 mL), dried over anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by prep-TLC (25% EtOAc/pet. ether) to afford the desired product (1.6 g, 79.2% yield). LCMS (ESI) m/z·. [M + H] calcd for C12H15NO3: 222.12; found 222.1.

Step 5: Synthesis of (2/?,3S)-3-(4-methoxyphenyl)aziridine-2-carboxylic acid To a solution of ethyl (2S,3fl)-3-(4-methoxyphenyl)aziridine-2-carboxylate (200.0 mg, 0.904 mmol) in MeOH and H 2 O at 0 °C was added LiOHeH 2 O (86.6 mg, 3.62 mmol). The resulting mixture was stirred for 1 h and was then neutralized to pH 7 with HCI (aq.). The mixture was extracted with EtOAc (3 x 100 mL) and the combined organic layers were washed with H 2 O (2 x 100 mL), dried over Na 2 SO 4 . filtered, and concentrated under reduced pressure to afford the desired product (180 mg, 97.9% yield). LCMS (ESI) m/z: [M - H] calcd for C10H11NO3: 192.07; found 192.0.

Intermediate A-43. Synthesis of (2S,3S)-1-((S)-fert-butylsulfinyl)-3-phenylaziridine-2- carboxylic acid

Step 1: Synthesis of (S,E)-N-benzylidene-2-methylpropane-2-sulfinamide A solution of (S)-2-methylpropane-2-sulfinamide (2.50 g, 20.6 mmol), titanium ethoxide (9.41 g,

41.25 mmol) and benzaldehyde (2.19 g, 20.7 mmol) was heated at 70 °C for 1 h, cooled, and diluted with H 2 O (250 mL). The aqueous layer was extracted with EtOAc (3 x 80 mL) and the combined organic layers were washed with brine (2 x 100 mL), dried with NaaSO^ filtered and concentrated under reduced pressure to afford the desired product (4.3 g, crude) which was used without further purification. LCMS (ESI) m/z: [M + H] calcd for C11H15NOS: 210.10; found 210.2. Step 2. Synthesis of ethyl (2S,3S)-1-((S)-tert-butylsulfinyl)-3-phenylaziridine-2-carbo xylate To a solution of ethyl bromoacetate (798 mg, 4.78 mmol) in THE (15 mL) at -78 °C was added LiHMDS (1M in THE, 4.78 mL, 4.78 mmol). After 1 h, (S,E)-N-benzylidene-2-methylpropane-2-sulfinamide (500 mg, 2.39 mmol) in THE (5 mL) was added in portions over 20 min. The reaction mixture was stirred at -78 °C for 2 h and then quenched by the addition of sat. NH4CI. The aqueous layer was extracted with EtOAc (3 x 40 mL) and the combined organic layers were washed with brine (2 x 30 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by reverse phase chromatography (30→60% MeCN/H20, 0.1% HCO2H) afforded the desired product (480 mg, 61% yield). LCMS (ESI) m/z. [M + H] calcd for C15H21NO3S: 296.13; found 296.2.

Step 2. Synthesis (2S,3S)-1 -((S)-tert-butylsulfinyl)-3-phenylaziridine-2-carboxylic acid To a solution of ethyl (2S,3S)-1-((S)-tert-butylsulfinyl)-3-phenylaziridine-2-carbo xylate (600 mg, 2.03 mmol) in THE (4.0 mL) at 0 °C was added a solution of LiOH (97.2 mg, 4.06 mmol) in H 2 O (4.0 mL). The resulting mixture was stirred for 2 h at 0 °C and then acidified to pH 5 with 1 M HCI. The aqueous layer was extracted with EtOAc (3 x 40 mL) and the combined organic layers were washed with brine (2 x 20 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired compound (450mg, crude) which was used without further purification. LCMS (ESI) m/z. [M + H] calcd for C13H17NO3S: 268.10; found 268.1.

Intermediate A-44. Synthesis of (2fl,3fl)-1-((fl)-tert-butylsulfinyl)-3-phenylazirldlne-2- carboxylic acid

Step 1: Synthesis (/?,E)-N-benzylidene-2-methylpropane-2-sulf inamide

A solution (fl)-2-methylpropane-2-sulfinamide (2.50 g, 20.6 mmol), titanium tetraethoxide (9.41 g, 41.3 mmol) and benzaldehyde (2.19 g, 20.6 mmol) was heated 70 °C for 1 h, cooled, and diluted with H 2 O (250 mL). The aqueous layer was extracted with EtOAc (3 x 90 mL) and the combined organic layers were washed with brine (2 x 100 mL), dried with Na 2 SO 4 , filtered and concentrated under reduced pressure to afford the desired product (4.2 g, crude) which was used without further purification. LCMS (ESI) m/z: [M + H] calcd for C11H15NOS: 210.10; found 210.1.

Step 2. Synthesis of ethyl (2f?,3f?)-1-((fl)-tert-butylsulfinyl)-3-phenylaziridine-2-ca rboxylate

To a solution of ethyl bromoacetate (6.38 g, 38.2 mmol) in THE (150 mL) at -78 °C was added LiHMDS (1 M in THE, 7.19 mL, 42.9 mmol). After 1 h, (f?,E)-N-benzylidene-2-methylpropane-2- sulfinamide (4.0 g, 19.1 mmol) in THE (50 mL) was added in portions over 20 min. The reaction mixture was stirred at -78 °C for 2 h and then quenched by the addition of sat. NHtCI. The aqueous layer was extracted with EtOAc (3 x 80 mL) and the combined organic layers were washed with brine (2 x 60 mL), dried with Na 2 SO 4 , filtered and concentrated under reduced pressure. Purification by reverse phase chromatography (30→60% MeCN/H 2 O, 0.1% HCOaH) afforded the desired product (3.9 g, 62% yield). LCMS (ESI) m/z. [M + H] calcd for C15H21NO3S: 296.13; found 296.2. Step 3: Synthesis (2/?,3fl)-1-((fl)-terf-butylsulfinyl)-3-phenylaziridine-2-ca rboxylic acid To a solution of ethyl (2/?,3/3)-1-((fl)-tert4>utylsulfinyl)-3-phenylaziridine-2 -carboxylate (200 mg, 0.677 mmol) in THF (1.5 mL) at 0 °C was added a solution of LiOH (32.4 mg, 1.35 mmol) in H 2 O (1.3 mL). The resulting mixture was stirred for 2 h at 0 °C and then acidified to pH 5 with 1 M HCI. The aqueous layer was extracted with EtOAc (3 x 20 mL) and the combined organic layers were washed with brine (2 x 10 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired compound (220 mg, crude) which was used without further purification. LCMS (ESI) m/z. [M + H] calcd for C13H17NO3S: 268.10; found 268.4.

Intermediate A-45 and A-46. Synthesis of and (S)-N- (1-(2-methoxyethyl)azlridlne-2- carbonyl)-A#-methylglyclne and (fl)-A/-(1-(2-methoxyethyl)azlrldlne-2-carbonyl)-N-methylgly cine

Step 1: Synthesis of tert-butyl W-acryloyl-W-methylglycinate

To a mixture of tert-butyl methylglycinate hydrochloride (1.0 g, 5.5 mmol) and NaHCOs (1.39 g, 16.5 mmol) in THF (10 mL) and H 2 O (5.0 mL) at 0 °C was added acryloyl chloride (750 mg, 8.26 mmol). The resulting solution was stirred for 2 h at room temperature and the reaction was then quenched by the addition H 2 O (50 mL). The aqueous layer was extracted with EtOAc (2 x 50 mL) and the combined organic layers were washed with brine, dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by normal phase chromatography (10→33% EtO Ac/pet. ether) afforded the desired product (900 mg, 73.8% yield). LCMS (ESI) m/z [M + H] calcd for C10H17NO3: 200.13; found 200.2.

Step 2. Synthesis of tert-butyl N-(2,3-dibromopropanoyl)-N-methylglycinate To a solution of tert-butyl A/-acryloyl-N-methylglycinate (2.0 g, 10.1 mmol) in DCM (40 mL) at -20 °C was added Bra (3.21 g, 20.1 mmol). The resulting mixture was stirred for 2 h at -20 °C and then quenched by the addition of NaaSaOa (100 mL). The aqueous layer was extracted with DCM (2 x 100 mL) and the combined organic layers were washed with brine, dried with Na 2 SO 4 , and concentrated under reduced pressure to afford the desired product (2.4 g, crude) which was used without further purification. LCMS (ESI) m/z. [M + Na] calcd for CioHvBraNOs: 381.96; found 381.8.

Step 3: Synthesis of tert-butyl (S)-N-(1-(2-methoxyethyl)aziridine-2-carbonyl)-N-methylglyci nate and tert-butyl (R)-N-( 1 -(2-methoxyethyl)aziridine-2-carbonyl)-W-methylglycinate

To a solution of tert-butyl N-(2,3-dibromopropanoyl)-N-methylglycinate (4.0 g, 11.1 mmol) and 2- methoxyethan-1 -amine (4.18 g, 55.7 mmol) in THF (40 mL) was added EtaN (4.66 mL, 33.4 mmol). The resulting solution was stirred at 35 °C overnight was then quenched by the addition of H 2 O. The aqueous layer was extracted with DCM (2 x 100 mL) and the combined organic layers were washed with brine, dried with Na 2 SO 4 . filtered, and concentrated under reduced pressure. Purification by reverse phase chromatography (30→50% MeCN/H 2 O) afforded a mixture of the desired products. The enantiomers were separated by chiral preparative normal phase chromatography (hexane, 10 mM NHa-MeOH/EtOH) to afford tert-butyl (S)-N-(1-(2-methoxyethyl)aziridine-2-carbonyl)-N-methylglyci nate (400 mg, 33.3% yield) and tert-butyl (fl)-W-(1-(2-methoxyethyl)aziridine-2-carbonyl)-N-methylglyc inate (360 mg, 30% yield). LCMS (ESI) m/z. [M + H] calcd for Ci 3 Ha 4 Na04: 273.18; found 273.0.

Step 4: Synthesis of (S)-N-(1-(2-methoxyethyl)aziridine-2-carbonyl)-AAmethylglyci ne To a solution of tert-butyl (S)-N-(1-(2-methoxyethyl)aziridine-2-carbonyl)-W-methylglyci nate (250 mg, 0.918 mmol) in DCM (6.0 mL) at 0 °C was added TEA (3.0 mL). The resulting mixture was stirred at 2 h at 0 °C and then concentrated under reduced pressure to afford the desired product (250 mg, crude) which was used without further purification. LCMS (ESI) m/z [M + H] calcd for CgHieNaO*: 217.12; found 217.1.

Step 5: Synthesis of (fl)-N-(1-(2-methoxyethyl)aziridine-2-carbonyl)-N-methylglyc ine To a solution tert-butyl (f7)-AA(1-(2-methoxyethyl)aziridine-2-carbonyl)-N-methylglyc inate (180 mg, 0.661 mmol) in DCM (6.0 mL) at 0 °C was added TEA (3.0 mL). The resulting mixture was stirred at 2 h at 0 °C and then concentrated under reduced pressure to afford the desired product (150mg, crude) which was used without further purification. LCMS (ESI) m/z [M + H] calcd for CgHieNaO*: 217.12; found 217.1.

Intermediate A-47 and A-48. Synthesis of A#-methyl-AH1 -(((fl)-1 -methylazlrldln-2- yl)sulfonyl)piperidine-4-carbonyl)-L-valine and N-methyl- Λ#·(1 -(((S)-1 -methylazlrldin-2- yl)sulfonyl)piperidine-4-carbonyl)-L-valine

Step 1: Synthesis of methyl N-methyl-N-(1-(vinylsulfonyl)piperidine-4-carbonyl)-L-valina te To a solution of 2-chloroethanesulfonyl chloride (1.91 g, 11.7 mmol) in THF (20 mL) at -70 °C was added methyl N-methyl-N-(piperidine-4-carbonyl)-i-valinate (2.0 g, 7.8 mmol) followed by EtaN (790 L, 780 mol). After warming to -50 °C additional EtsN (790 L, 780 mol) was added and the reaction mixture warmed to room temperature. After 1 h the reaction was quenched at 0 °C by the addition of h½0 (30 mL), acidified to pH 6 with 1 M HCI, and extracted with CHCla (3 x 30 mL). The combined organic layers were dried with MgS04, filtered, and concentrated under reduced pressure. Purification by normal phase chromatography (50% EtOAc/pet. ether) afforded the desired product (560 mg, 20.7% yield). LCMS (ESI) m/zr. [M + H] calcd forCi5H2eN 2 0sS: 347.17; found 347.2.

Step 2. Synthesis of methyl N-( 1 -((1 ,2-dibromoethyl)sulfonyl)piperidine-4-carbonyl)-W-methyl-L- valinate

To a solution of methyl N-methyl-N-(1-(vinylsulfonyl)piperidine-4-carbonyl)-L-valina te (580 mg,

1.67 mmol) in CCU (28 mL) at room temperature was added Bra (580 mg, 1.67 mmol) The resulting mixture was stirred overnight at room temperature and then quenched by the addition of sat. NaHCOs (30 mL). The aqueous layer was extracted with DCM (3 x 30 mL) and the combined organic layers were dried with N 32804, filtered and concentrated under reduced pressure to afford the desired product which was used without further purification. LCMS (ESI) m/z: [M + H] calcd for CisHaeBraNaOsS: 506.99; found 506.9.

Step 3: Synthesis of methyl W-methyl-N-(1-(((fl)-1-methylaziridin-2-yl)sulfonyl)piperidi ne-4- carbonyl)-L-valinate and methyl N- methyl-N-(1-(((S)-1-methylaziridin-2-yl)sulfonyl)piperidine- 4-carbonyl)- L-valinate

To a solution of methyl N-(1-((1 ,2-dibromoethyl)sulfonyl)piperidine-4-carbonyl)-N- methyl-L- valinate (4.80 g, 9.481 mmol) in DMSO (48 mL) was added methanamine hydrochloride (1 .92 g, 28.436 mmol) and EtsN (13.2 mL, 94.8 mmol). The reaction mixture was heated to 75 °C and stirred overnight. The mixture was then cooled to 0 °C, diluted NhLCI, and extracted with EtOAc (600 mL). The organic layer was washed with brine, dried with Na 2 SO 4 , filtered, concentrated under reduced pressure. Purification with normal phase chromatography (86% EtOAc/hexane) afforded a mixture of the desired products. The diastereomers were separated by prep-SFC chromatography (20% IPA/COa) to afford methyl N-methyl-AA(1-(((/?)-1-methylaziridin-2-yl)sulfonyl)piperidi ne-4-carbonyl)-L-valinate (700 mg, 38.9% yield) and methyl N-methyl-N-(1-(((S)-1-methylaziridin-2-yl)sulfonyl)piperidin e-4-carbonyl)-L· valinate (790 mg, 43.9% yield). LCMS (ESI) m/z: [M + H] calcd for CieHagNsOsS: 376.19; found 376.1.

Step 4: Synthesis of N-methyl-N-(1-(((fl)-1-methylaziridin-2-yl)sulfonyl)piperidi ne-4-carbonyl)-L· valine

To a solution of methyl W-methyl-A/-(1-(((fl)-1-methylaziridin-2-yl)sulfonyl)piperid ine-4-carbonyl)- L-valinate (200 mg, 0.533 mmol) in THF (2.0 mL) at 0 °C was added 1 M LiOH (1 mL) The resulting mixture was stirred for 3 h at room temperature and then acidified to pH 6 with 1 M HCI. The aqueous layer was extracted with EtOAc (3 x 10 mL) and the combined organic layers were dried with NazS04, filtered, and concentrated under reduced pressure to afford the desired product which was used without further purification. LCMS (ESI) m/z: [M + H] calcd for C15H27N3O5S: 362.18; found 362.2.

Step 5: Synthesis of N-methyl-N-(1-(((S)-1-methylaziridin-2-yl)sulfonyl)piperidin e-4-carbonyl)-L- valine

To a solution methyl N-methyl-N-(1-(((S)-1-methylaziridin-2-yl)sulfonyl)piperidin e-4-carbonyl)-L- valinate (300 mg, 0.799 mmol) in THF (3.0 mL) at 0 °C was added 1 M LiOH (3.0 mL). The resulting mixture was stirred for 3 h at room temperature and then acidified to pH 6 with 1 M HCI. The aqueous layer was extracted with EtOAc (3 x 10 mL) and the combined organic layers dried with Na 2 SO 4 , filtered and concentrated under reduced pressure to afford the desired product which was used without further purification. LCMS (ESI) m/z: [M + H] calcd for C15H27N3O5S: 362.18; found 362.2. Intermediate A-49 and A-50. Synthesis of W-methyl-W-(1-(((fl)-1-methylaziridin-2- yl)sulfonyl)azetidine-3-cait>onyl)-i.-valine and N-methyl-N-(1 -(((S)-1 -methylaziridin-2- yl)sulfonyl)azetidine-3-cait>onyl)-i.-valine

ΥΥ

Step 1: Synthesis of methyl N-methyl-N-(1-(vinylsulfonyl)azetidine-3-carbonyl)-L-valinat e To a solution of 2-ch loroethanesu Ifony I chloride (357 mg, 2.19 mmol) in EtaO (4.0 mL) at -70 °C was added an EtaO (4.0 mL) solution of methyl N-(azetidine-3-carbonyl)-N-methyl-Z.-valinate (500 mg, 2.19 mmol) followed by EtsN (0.304 mL, 2.19 mmol). The resulting mixture was stirred for 30 min at -50 °C at which time EfeN (0.304 mL, 2.19 mmol) was added. The resulting mixture was stirred for 1 h at room temperature and then quenched with H 2 O at 0 °C. The mixture was acidified to pH 6 with 1 M HCI and extracted with CHCIs (3 x 10 mL). The combined organic layers were washed with brine, dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by normal phase chromatography (50% EtOAc/pet. ether) afforded the desired product (180 mg, 25.8% yield). LCMS (ESI) m/z. [M + H] calcd for C13H22N 2 O5S: 319.13; found 319.1.

Step 2. Synthesis of methyl ΛΑ(1 -((1 ,2-dibromoethyl)sulfonyl)azetidine-3-carbonyl)-N-methyl-L- valinate

To a solution of methyl N-methyl-AA(1-(vinylsulfonyl)azetidine-3-carbonyl)-L-valinat e (460 mg, 1.45 mmol) in CCI4 (6.0 mL) at room temperature was added a CCL (2.0 mL) solution of Bra (346 mg, 2.17 mmol). The resulting mixture was stirred overnight and then quenched at 0 °C by the addition of sat. NaHCOs and NaaSaOa. The aqueous layer was extracted with DCM (3 x 10 mL) and the combined organic layers were washed with brine, dried with Na 2 SO 4 , and concentrated under reduced pressure to afford the desired product (500 mg) which was used without further purification. LCMS (ESI) m/z. [M + H] calcd for CisHaaBraNaOsS: 478.97; found 478.0.

Step 3: Synthesis of methyl W-methyl-N-(1 -(((fl)-1 -methylaziridin-2-yl)sulfonyl)azetidine-3- carbonyl)-L-valinate and methyl N- methyl-N-(1 -(((S)-1 -methylaziridin-2-yl)sulfonyl)azetidine-3-carbonyl)-L- valinate

To a solution of methyl N-(1-((1 ,2-dibromoethyl)sulfonyl)azetidine-3-carbonyl)-N- methyl-L-valinate (260 mg, 0.54 mmol) in DMSO (4.0 mL) was added methanamine hydrochloride (110.0 mg, 1.63 mmol) and EtaN (0.758 mL, 5.44 mmol). The resulting mixture was heated to 75 °C and stirred overnight. The mixture was then cooled to room temperature, diluted with H 2 O (10 mL), and extracted with EtOAc (3 x 5 mL). The combined organic layers were washed with brine, dried with Na 2 SO 4 , filtered and concentrated under reduced pressure. Purification by normal phase chromatography (50% EtOAc/pet. ether) afforded a mixture of the desired products. The diastereomers were separated by chiral prep normal phase chromatography (hexane, 10 mM NHa-MeOH /IPA) to afford methyl N-methyl-W-(1 -(((fl)-1 -methylaziridin- 2-yl)sulfonyl)azetidine-3-carbonyl)-L-valinate (0.59 g, 35% yield) and methyl W-methyl-N-(1 -(((S)-1 - methylaziridin-2-yl)sulfonyl)azetidine-3-carbonyl)-L-valinat e (0.56 g, 33% yield). LCMS (ESI) m/z. [M + H] calcd for C14H25N3O5S: 348.16; found 348.2.

Step 4: Synthesis of N-methyl-N-(1-(((fl)-1-methylaziridin-2-yl)sulfonyl)azetidin e-3-carbonyl)-L- valine

To a solution of methyl N-methyl-N-(1-(((fl)-1-methylaziridin-2-yl)sulfonyl)azetidin e-3-carbonyl)-L· valinate (225.0 mg, 0.65 mmol) in THF (1.5 mL) at 0 °C was added LIOH (77.0mg, 3.23 mmol) dissolved in H 2 O (1.5 mL). The resulting mixture was stirred for 2 h at room temperature and then acidified to pH 6 with 1 M HCI. The aqueous layer was extracted with EtOAc and the combined organic layers were washed with brine, dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (270 mg) which was used without further purification. LCMS (ESI) m/z. [M + H] calcd for C13H23N3O5S: 334.15; found 334.0.

Step 5: Synthesis of N-methyl-N-(1-(((S)-1-methylaziridin-2-yl)sulfonyl)azetidine -3-carbonyl)-L· valine

To a solution of methyl N-methyl-N-(1-(((S)-1-methylaziridin-2-yl)sulfonyl)azetidine -3-carbonyl)-L· valinate (365.0 mg, 1.05 mmol) in THF (2.0 mL) and H 2 O (2.0 mL) at 0 °C was added LiOH hydrate (132.0 mg, 3.15 mmol). The resulting mixture was stirred for 2 h at room temperature then acidified to pH 6 with 1 M HCI and diluted with H 2 O (20 mL). The aqueous layer was extracted with EtOAc and the combined organic layers were washed with brine, dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (257 mg, crude) which was used without further purification. LCMS (ESI) m/z. [M + H] calcd for C13H23N3O5S: 334.15; found 334.3.

Intermediate A-51. Synthesis of 2-((1 /?,5S)-2,4-dioxo-6-trityl-3,6-diazabicyclo[3.1.0]hexan-3- yl)acetic acid

Step 1: Synthesis of benzyl 2-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetate To a solution of 2-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetic acid (5.0 g, 32.2 mmol) and EfaN (13.5 mL, 96.7 mmol) in THF (80 mL) at 0 °C was added benzyl bromide (11.03 g, 64.5 mmol). The resulting mixture was stirred overnight at room temperature and then filtered. The filter cake was washed with THF (3 x 40 mL) and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography (16% EtOAc/hexanes) afforded the desired product (4.4 g, 55.7% yield) LCMS (ESI) m/z. [2M + Na] calcd for C13H11NO4: 513.14; found 513.2.

Step 2. Synthesis of benzyl 2-((1 /?,5S)-2,4-dioxo-6-trityl-3,6-diazabicyclo[3.1.0]hexan-3-yl) acetate To a solution of benzyl 2-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)acetate of (1.0 g, 4.0 mmol) in toluene (10 mL) was added trityl azide (1.36 g, 4.89 mmol). The resulting mixture was stirred overnight at 120 °C and then concentrated under reduced pressure. Purification by reverse flash chromatography (50→80% MeCN/HzO) afforded the desired product (400 mg, 19.5% yield). LCMS (ESI) m/z [M + Na] calcd for C32H26N 2 O4: 525.19; found 525.2.

Step 3: Synthesis of 2-((1 fif,5S)-2,4-dioxo-6-trityl-3,6-diazabicyclo[3.1.0]hexan-3-yl )acetic acid To a solution of benzyl 2-((1 f?,5S)-2,4-dioxo-6-trityl-3,6-diazabicyclo[3.1.0]hexan-3-yl) acetate (220 mg, 0.438 mmol) in THF (8.0 mL) was added Pd(OH)z/C (60 mg). The resulting solution was placed under a hydrogen atmosphere for 3 h using a Ha balloon, filtered through Celite, and concentrated under reduced pressure to afford the desired product (160 mg, crude) which was used without further purification. LCMS (ESI) m/z [M - H] calcd for C25H20N 2 O4: 411.13; found 411.2.

Intermediate A-52. Synthesis of (Ο-Β-ηΐθΙΙ^Ζ-ίδ-οχο-Ζ-ΜδΜ-ΙΠΙγΙβζΐ ΜΙηβ^-οβΑοηγΙ)- 2,6-dlazaspiro[3.4]octan-6-yl)butanoic acid

Step 1: Synthesis of 1 -(tert-butyl) 3-methyl 3-allylazetidine-1 ,3-dicarboxylate To a solution of 1 -(tert-butyl) 3-methyl azetidine-1 ,3-dicarboxylate (20.0 g, 92.9 mmol) and LiHMDS (140 mL, 1M in THF, 139 mmol) in THF (200 mL) at -78 °C was added ally! bromide (16.9 g, 139 mmol). The resulting solution was stirred overnight at room temperature and then quenched with the addition of sat. NH4CI (100 mL) and diluted with EtOAc (800 mL). The organic layer was washed with brine (3 x 300 mL), dried with NaaSCX, filtered and concentrated under reduced pressure. Purification by silica gel column chromatography (17% EtO Ac/pet. ether) afforded the desired product (15.0 g, 63.2% yield). LCMS (ESI) m/z [M + H - ®u] calcd for C13H21NO4: 200.10; found 200.0

Step 2. Synthesis of 1 -(tert-butyl) 3-methyl 3-(2-oxoethyl)azetidine-1 ,3-dicarboxylate To a solution of 1 -(fart-butyl) 3-methyl 3-allylazetidine-1 ,3-dicarboxylate (6.0 g, 23 mmol) and 2,6- lutidine (504 mg, 47.0 mmol) in dioxane (60 mL) and H 2 O (60 mL) at 0 °C was added Kz0s04*2H20 (433 mg, 1 .18 mmol). The resulting mixture was stirred at room temperature for 15 min then Nal04 (20.1 g, 94.0 mmol) was added at 0 °C. The reaction was stirred for 3 h at room temperature and then quenched with sat. NazSaOs at 0 °C. The aqueous layer was extracted with EtOAc (2 x 400 mL) and the combined organic layers were washed with 1 M HCI (2 x 80 mL), brine (2 x 100 mL), dried with Na 2 SO 4 , filtered and concentrated under reduced pressure to afford the desired product (2.84 g, crude) which was used without further purification. LCMS (ESI) m/z [M - H] calcd for C12H19NO5: 256.12; found 256.0 Step 3: Synthesis of 1 -(tert-butyl) 3-methyl (S)-3-(2-((1 -methoxy-3-methyl-1 -oxobutan-2- yl)amino)ethyl)azetidine-1 ,3-dicarboxylate

To a solution of 1 -(tert-butyl) 3-methyl 3-(2-oxoethyl)azetidine-1 ,3-dicarboxylate (13.0 g, 50.5 mmol) and methyl L-valinate hydrochloride (7.29 g, 55.6 mmol) in MeOH (130 mL) at 0 °C were added ZnCl 2 (7.57 g, 55.6 mmol) and NaBHaCN (6.35 g, 101 mmol).The resulting mixture was stirred at room temperature overnight, partially concentrated under reduced pressure and diluted with EtOAc (500 mL). The resulting solution was washed with brine (3 x 200 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by normal phase chromatography (10→66% EtOAc/pet. ether) afforded the desired product (7.72 g, 41.0% yield). LCMS (ESI) m/z. [M + H] calcd for C18H32N 2 O6:

373.24; found 373.1.

Step 4: Synthesis of tert-butyl (S)-6-(1 -methoxy-3-methyl-1 -oxobutan-2-yl)-5-oxo-2,6- diazaspiro[3.4]octane-2-carboxylate

To a solution of 1 -(tert-butyl) 3-methyl (S)-3-(2-((1 -methoxy-3-methyl-1 -oxobutan-2- yl)amino)ethyl)azetidine-1 ,3-dicarboxylate (6.0 g, 16 mmol) and DIPEA (28.0 mL, 161 mmol) in toluene (60 mL) at room temperature was added DMAP (197 mg, 1.61 mmol). The resulting mixture was stirred at 80 °C overnight, diluted with EtOAc (50 mL), washed with H 2 O (50 mL), brine (3 x 50 mL) dried with Na 2 SO 4 , and filtered, and concentrated under reduced pressure. Purification by reverse phase chromatography 45→80% MeCN/H 2 O) afforded the desired product (4.3 g, 78.4% yield). LCMS (ESI) m/z. [M + H - IBu] calcd for CvHaeNaOs: 285.15; found 285.0

Step 5: Synthesis of methyl (S)-3-methyl-2-(5-oxo-2,6-diazaspiro[3.4]octan-6-yl)butanoat e

To a solution of tert-butyl (S)-6-(1-methoxy-3-methyl-1-oxobutan-2-yl)-5-oxo-2,6- diazaspiro[3.4]octane-2-carboxylate (2.7 g, 7.9 mmol) in DCM (27 mL) at room temperature was added TFA (8.10 mL, 71 .0 mmol).The resulting mixture was stirred for 1 h and then concentrated under reduced pressure to afford the desired product, (1.70 g, crude) which was used without further purification. LCMS (ESI) m/z : [M + H] calcd for C12H20N 2 O3: 241.16; found 240.1.

Step 6: Synthesis of methyl (S)-3-methyl-2-(5-oxo-2-((S)-1-tritylaziridine-2-carbonyl)-2 ,6- diazaspiro[3.4]octan-6-yl)butanoate

To a solution methyl (S)-3-methyl-2-(5-oxo-2,6-diazaspiro[3.4]octan-6-yl)butanoat e (700 mg, 2.91 mmol) and (S)-1-tritylaziridine-2-carboxylic acid (1.15 g, 3.50 mmol) in DMF (7.0 mL) at 0 °C was added DIPEA (2.5 mL,14.6 mmol). After 30 min HATU (1.66 g, 4.37 mmol) was added and the resulting mixture was stirred at room temperature for 1 h. The reaction was then diluted with EtOAc (20 mL) and the organic layer was washed with sat. NhUCI (50 mL), brine (3 x 50 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by normal phase chromatography (0→80% EtOAc/pet. ether) afforded the desired product (300 mg, 18.7% yield). LCMS (ESI) m/z. [M + H] calcd for C34H37N3O4: 552.29; found 552.2.

Step 7: Synthesis of (S)-3-methyl-2-(5-oxo-2-((S)-1-tritylaziridine-2-carbonyl)-2 ,6- diazaspiro[3.4]octan-6-yl)butanoic acid

To a solution of methyl (S)-3-methyl-2-(5-oxo-2-((S)-1-tritylaziridine-2-carbonyl)-2 ,6- diazaspiro[3.4]octan-6-yl)butanoate (700 mg, 1.27 mmol) in THF (10 mL) and H 2 O (2.0 mL) at 0 °C was added LiOH (152 mg, 6.34 mmol). After 30 min the reaction mixture was warmed to room temperature for 1 h and then acidified to pH 6 with 1 M HCI. The aqueous layer was extracted with EtOAc (3 x 50 mL) and the combined organic layers were washed with brine, dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (300 mg, 18.7% yield) which was used without further purification. LCMS (ESI) m/z. [M + H] calcd for C33H35N3O4: 538.27; found 538.2. Intermediate A-53. Synthesis of (S)-3-methyl-2-(5-oxo-2-((fl)-1-trltylazlridlne-2-carbonyl)- 2,6-diazaspiro[3.4]octan-6-yl)butanoic acid Synthesis of methyl (S)-3-methyl-2-(5-oxo-2-((fl)-1-tritylaziridine-2-carbonyl)- 2,6-diazaspiro[3.4]octan-6-yl)butanoate

To a solution ffl)-1 -tritylaziridine-2-carboxylic acid (1.0 g, 3.0 mmol) and methyl (S)-3-methyl-2- (5-oxo-2,6-diazaspiro[3.4]octan-6-yl)butanoate (875 mg, 3.64 mmol) in DMF (10 mL) at 0 °C was added DIPEA (2.64 mL, 15.2 mmol). After 30 min HATU (1.73 g, 4.554 mmol) was added and the resulting mixture was stirred for 1 h at room temperature. The reaction was then diluted with EtOAc (20 mL) and the organic layer was washed with sat. NFUCI (50 mL), brine (3 x 50 mL), dried with NaaSOA, filtered, and concentrated under reduced pressure. Purification by silica gel chromatography (0→80% EtOAc/pet. ether) afforded the desired product (789 mg, 47% yield). LCMS (ESI) m/z: [M + H] calcd for C34H37N3O4: 552.29; found 552.3.

Step 2. Synthesis of (S)-3-methyl-2-(5-oxo-2-((fl)-1-tritylaziridine-2-carbonyl)- 2,6- diazaspiro[3.4]octan-6-yl)butanoic acid

To a stirred solution of methyl (S)-3-methyl-2-(5-oxo-2-((fl)-1-tritylaziridine-2-carbonyl)- 2,6- diazaspiro[3.4]octan-6-yl)butanoate (900 mg, 1.63 mmol) in THF (10 mL) and H 2 O (2.5 mL) at 0 °C was added LiOH (156 mg, 6.53 mmol). After 30 min the reaction mixture was warmed to room temperature for 1 h and then acidified to pH 6 with 1 M HCI. The aqueous layer was extracted with EtOAc (3 x 50 mL) and the combined organic layers were washed with brine, dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (240 mg, 27.4% yield). LCMS (ESI) m/z: [M + H] calcd for C33H35N3O4: 538.27; found 538.2.

Intermediate A-54. Synthesis of (S)-1-((fl)-2-(methoxycarbonyl)azlrldlne-1- carbonyl)pyrroMdine-3-carboxyMc acid

Step 1: Synthesis of methyl (fl)-aziridine-2-carboxylate

A suspension of 1 -benzyl 2-methyl (fl)-aziridine-1 ,2-dicarboxylate (1.50 g, 6.4 mmol) and Pd/C (300 mg, 2.8 mmol) in THF (15 mL) under an atmosphere of hydrogen (1 atm) was stirred for 3 h before the solids were removed by filtration. The crude solution was concentrated under reduced pressure which afforded desired product (600 mg, crude). LCMS (ESI) m/z: [M + H] calcd for C4H7NO2: 102.06; found 102.3.

Step 2. Synthesis of benzyl (S)-1 -((fl)-2-(methoxycarbonyl)aziridine-1 -carbonyl)pyrrolidine-3- carboxylate

To solution of methyl (fl)-aziridine-2-carboxylate (1.0 g, 9.90 mmol) and benzyl (S)-pyrrolidine-3- carboxylate (2.63 g, 10.9 mmol, HCI salt) in DCM (30.0 mL) at -10 °C was added DIPEA (10.3 mL, 59.3 mmol) followed by triphosgene (880 mg, 2.97 mmol). The resulting solution was stirred for 30 min and was then quenched by the addition of H 2 O (50 mL). The aqueous layer was extracted with DCM (2 x 100 mL), washed with brine (2 x 50 mL), dried over Na 2 SO 4 , and concentrated under reduced pressure. Purification by prep-TLC (50% EtOAc/pet. ether) afforded desired product (1.30 g, 28% yield). LCMS (ESI) m/z. [M + H] calcd for C17H20N 2 O5: 333.15; found 333.2.

Step 3: Synthesis of (S)-1-((fl)-2-(methoxycarbonyl)aziridine-1-carbonyl)pyrrolid ine-3-carboxylic acid

To a solution of benzyl (S)-1-((fl)-2-(methoxycarbonyl)aziridine-1-carbonyl)pyrrolid ine-3- carboxylate (200 mg, 600 pmol) in MeOH (5 mL) and DCM (5 mL) under Ha was added Pd(OH)a/C (130 mg, 90 pmol). The resulting mixture was stirred for 30 min at room temperature and then the mixture was filtered. The filter cake was washed with MeOH (2 x 10 mL) and the filtrate was concentrated under reduced pressure which afforded desired product (140 mg, 96% yield) as an off-white solid. LCMS (ESI) m/z. [M + H] calcd for C10H14N 2 O5: 243.10; found 243.3.

Intermediate A-55. Synthesis of (S)-1-((S)-2-(methoxycarbonyl)azirldlne-1- carbonyl)pyrrolldlne-3-carboxyllc ackf o b"'(A NH o cbz-

Bn' H vI O

Step 1: Synthesis of methyl (S)-aziridine-2-carboxylate

A suspension of 1 -benzyl 2-methyl (S)-aziridine-1 ,2-dicarboxylate (200 mg, 850 pmol) and Pd/C (20 mg, 38 pmol) in THF (4.0 mL) under an atmosphere of hydrogen (1 atm) was stirred for 2 h before the solids were removed by filtration. The crude solution was concentrated under reduced pressure which afforded desired product (92 mg, crude). LCMS (ESI) m/z. [M + H] calcd for C4H7NO2: 102.06; found 102.3.

Step 2. Synthesis of benzyl (S)-1-((S)-2-(methoxycarbonyl)aziridine-1-carbonyl)pyrrolidi ne-3- carboxylate

To a solution of methyl (S)-aziridine-2-carboxylate (900 mg, 8.9 mmol) and benzyl (S)-pyrrolidine- 3-carboxylate (2.37 g, 9.80 mmol, HCI salt) in DCM (30 mL) at -10 °C was added DIPEA (9.30 mL, 53.4 mmol) followed by triphosgene (790 mg, 2.67 mmol). The resulting solution was stirred for 30 min and was then quenched by the addition of H 2 O (50 mL). The aqueous layer was extracted with DCM (2 x 100 mL), washed with brine (2 x 50 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by prep-TLC (50% EtOAc/pet. ether) afforded desired product (360 mg, 8.5% yield) as an off-white oil. LCMS (ESI) m/z. [M + H] calcd for C17H20N 2 O5: 333.15; found 333.2.

Step 3: Synthesis of (S)-1-((S)-2-(methoxycarbonyl)aziridine-1-carbonyl)pyrrolidi ne-3-carboxylic acid

To a solution of benzyl (S)-1-((S)-2-(methoxycarbonyl)aziridine-1-carbonyl)pyrrolidi ne-3- carboxylate (130 mg, 390 pmol) in MeOH (3 mL) and DCM (3 mL) under Hawas added Pd(OH)z/C (55 mg, 39 pmol). The resulting solution was stirred for 30 min at room temperature and then the reaction mixture was filtered. The filter cake was washed with MeOH (2 x 10 mL) and the filtrate was concentrated under reduced pressure which afforded desired product (90 mg, 95% yield) as an off-white solid. LCMS (ESI) m/z. [M + H] calcd for C10H14N 2 O5: 243.10; found 243.3.

Intermediate A-56. Synthesis of (2/?,3S)-3-cyclopropylaziridine-2-carboxylic acid

Step 1: Synthesis of ethyl (2S,3fl)-3-cyclopropyl-2,3-dihydroxypropanoate

A solution of ethyl (E)-3-cyclopropylacrylate (10.4 mL, 71 mmol) in tert-BuOH (270 mL) and H 2 O (270 mL) was stirred at 0 °C. After 5 min MsNHa (6.8 g, 71 mmol) and (DHQD)2PHAL (100 g, 130 mmol) were added and the reaction mixture was warmed to room temperature. After stirring overnight, sat. NaaSOa was added and the mixture was stirred for 30 min. The mixture was acidified to pH 6 with KH2PO4. Purification by silica gel column chromatography (33% EtO Ac/pet. ether) afforded desired product (5.5 g, 44% yield).

Step 2. Synthesis of ethyl (2S,3fl)-3-cyclopropyl-3-hydroxy-2-(((4- nitrophenyl)sulfonyl)oxy)propanoate

A solution of ethyl (2S,3fl)-3-cyclopropyl-2,3-dihydroxypropanoate (5.40 g, 31.0 mmol) and EtaN (13.0 mL, 93.0 mmol) in DCM (20 mL) was stirred at 0 °C and a solution of 4-n itrobenzenesu Ifonyl chloride (6.53 g, 29.5 mmol) in DCM (10 mL) was added. The reaction mixture was stirred for 1.5 h and was then extracted with DCM (3 x 200 mL). The combined organic layers were washed with brine (100 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by silica gel column chromatography (33% EtO Ac/pet. ether) afforded desired product (6.9 g, 62% yield).

Step 3: Synthesis of ethyl (2fl,3fl)-2-azido-3-cyclopropyl-3-hydroxypropanoate

A mixture of ethyl (2S,3fl)-3-cyclopropyl-3-hydroxy-2-(((4-nitrophenyl)sulfonyl )oxy)propanoate (6.90 g, 19.2 mmol) and NaNs (6.24 g, 96.0 mmol) in DMF (70.0 mL) was heated to 50 °C. The reaction mixture was stirred for 5 h and then extracted with EtOAc (3 x 200 mL). The combined organic layers were washed with brine (100 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by silica gel column chromatography (20% EtOAc/pet. ether) afforded desired product (2.8 g, 73% yield).

Step 4: Synthesis of ethyl (2/?,3S)-3-cyclopropylaziridine-2-carboxylate A mixture of triphenylphosphine (1.84 g, 7.02 mmol) in DMF (5 mL) was stirred at 0 °C. After 5 min ethyl (2/?,3/ : ?)-2-azido-3-cyclopropyl-3-hydroxypropanoate (1.40 g, 7.03 mmol) was added and the reaction was warmed to room temperature. The reaction mixture was heated to 80 °C and stirred for 1 h. The mixture was then cooled to room temperature and extracted with EtO Ac (3 x 50 mL). The combined organic layers were washed with brine (50 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by silica gel column chromatography (20% EtOAc/pet. ether) afforded the desired product (230 mg, 46% yield). LCMS (ESI) m/z. [M + H] calcd for CBH13NO2: 156.10; found 156.2. Step 5: Synthesis of lithium (2fl,3S)-3-cyclopropylaziridine-2-carboxylate To a mixture of ethyl (2/?,3S)-3-cyclopropylaziridine-2-carboxylate (230 mg, 1.5 mmol) in MeOH (3.0 mL) was added LiOH*H20 (125 mg, 3.0 mmol). The reaction was stirred for 3 h and then filtered. The filtrate was concentrated under reduced pressure which afforded the desired product (150 mg, crude). LCMS (ESI) m/z. [M + H] calcd for C6H9NO2: 128.07; found 128.2.

Intermediate A-57. Synthesis of (2fl,3S)-3-cyclopropylazlrldine-2-carboxyllc acid

Step 1: Synthesis of ethyl (2S,3 fi)-3-cyclopropyl-2 ,3-dihyd roxypropanoate

A solution of ethyl (£)-3-cyclopropylacrylate (10.4 mL, 71 mmol) in tert-BuOH (270 mL) and H 2 O (270 mL) was stirred at 0 °C. After 5 min MSNH2 (6.8 g, 71 mmol) and (DHQD)2PHAL (100 g, 130 mmol) were added and the reaction mixture was warmed to room temperature. After stirring overnight, sat. Na2S03 was added and the mixture was stirred for 30 min. The mixture was acidified to pH 6 with KH2PO4. Purification by silica gel column chromatography (33% EtOAC/pet. ether) afforded desired product (5.5 g, 44% yield).

Step 2. Synthesis of ethyl (2S,3/3)-3-cyclopropyl-3-hydroxy-2-(((4- nitrophenyl)sulfonyl)oxy)propanoate

A solution of ethyl (2S,3fl)-3-cyclopropyl-2,3-dihydroxypropanoate (5.40 g, 31.0 mmol) and EfeN (13.0 mL, 93.0 mmol) in DCM (20 mL) was stirred at 0 °C and a solution of 4-n itrobenzenesu Ifonyl chloride (6.53 g, 29.5 mmol) in DCM (10 mL) was added. The reaction mixture was stirred for 1.5 h and was then extracted with DCM (3 x 200 mL). The combined organic layers were washed with brine (100 mL), dried with Na2SC>4, filtered, and concentrated under reduced pressure. Purification by silica gel column chromatography (33% EtOAc/pet. ether) afforded desired product (6.9 g, 62% yield).

Step 3: Synthesis of ethyl (2/?,3fl)-2-azido-3-cyclopropyl-3-hydroxypropanoate

A mixture of ethyl (2S,3fl)-3-cyclopropyl-3-hydroxy-2-(((4-nitrophenyl)sulfonyl )oxy)propanoate (6.90 g, 19.2 mmol) and NaNa (6.24 g, 96.0 mmol) in DMF (70.0 mL) was heated to 50 °C. The reaction mixture was stirred for 5 h and then extracted with EtOAc (3 x 200 mL). The combined organic layers were washed with brine (100 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by silica gel column chromatography (20% EtOAc/pet. ether) afforded desired product (2.8 g, 73% yield).

Step 4: Synthesis of ethyl (2/?,3S)-3-cyclopropylaziridine-2-carboxylate A mixture of triphenylphosphine (1.84 g, 7.02 mmol) in DMF (5 mL) was stirred at 0 °C. After 5 min ethyl (2f?,3fl)-2-azido-3-cyclopropyl-3-hydroxypropanoate (1.40 g, 7.03 mmol) was added and the reaction was warmed to room temperature. The reaction mixture was heated to 80 °C and stirred for 1 h. The mixture was then cooled to room temperature and extracted with EtO Ac (3 x 50 mL). The combined organic layers were washed with brine (50 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by silica gel column chromatography (20% EtOAc/pet. ether) afforded the desired product (230 mg, 46% yield). LCMS (ESI) m/z. [M + H] calcd for CeHisNOa: 156.10; found 156.2.

Step 5: Synthesis of lithium (2fl,3S)-3-cyclopropylaziridine-2-carboxylate To a mixture of ethyl (2/?,3S)-3-cyclopropylaziridine-2-carboxylate (230 mg, 1.5 mmol) in MeOH (3.0 mL) was added LiOH-H 2 O (125 mg, 3.0 mmol). The reaction was stirred for 3 h and then filtered. The filtrate was concentrated under reduced pressure which afforded the desired product (150 mg, crude). LCMS (ESI) m/z. [M + H] calcd for CeHgNOa: 128.07; found 128.2.

Intermediate A-58. Synthesis of (2S,3fl)-3-cyclopropylazlrldlne-2-carboxyllc acid

Step 1: Synthesis of ethyl (2S,3fl)-3-cyclopropylaziridine-2-carboxylate A mixture of PPhs (1.4 g, 5.4 mmol) in DMF (15.0 mL) was stirred at 0 °C. After 30 min, ethyl (2S,3S)-2-azido-3-cyclopropyl-3-hydroxypropanoate (980 mg, 4.92 mmol) was added. The reaction mixture was heated to 80 °C. After 2 h the reaction was quenched by the addition of H 2 O (20 mL) and was extracted with EtO Ac (3 x 30 mL). Purification by silica gel column chromatography (17% EtO Ac/pet. ether) afforded desired product (500 mg, 65% yield).

Step 2. Synthesis of lithium (2S,3/3)-3-cyclopropylaziridine-2-carboxylate To a solution of ethyl (2S,3fl)-3-cyclopropylaziridine-2-carboxylate (450 mg, 2.9 mmol) in THF (6.0 mL) and H 2 O (2.0 mL) was added LiOH (90 mg, 3.8 mmol). The reaction was stirred for 2 h and then filtered. The filtrate was concentrated under reduced pressure which afforded the desired product (300 mg, crude).

Intermediate A-59. Synthesis of (fl)-3-methyl-2-(((fl)-N-methyl-1-trltylazlrldlne-2- carboxamido)methyl)butanoic acid

Step 1: Synthesis of methyl (fl)-2-(((iert-butoxycarbonyl)(methyl)amino)methyl)-3- methylbutanoate

To a solution of (/3)-2-(((tert-butoxycarbonyl)amino)methyl)-3-methylbutanoic acid (500 mg, 2.16 mmol) in DMF (10.0 mL) at 0 °C was added NaH (130 mg, 5.40 mmol). After 30 min, Mel (540 μΙ_, 8.65 mmol) was added and the reaction was warmed to room temperature. After 2 h the reaction was cooled to 0 °C and quenched by the addition of sat. aq. NH4CI (10 mL). The resulting mixture was extracted with EtOAc (3 x 20 mL) and the combined organic layers were washed with brine (40 mL), dried with Na 2 SO 4 , and concentrated under reduced pressure. Purification by prep-TLC (9% EtOAc/pet. ether) afforded the desired product (500 mg, 89.2% yield). LCMS (ESI) m/z. [M + H] calcd for C13H25NO4: 260.19; found 260.2.

Step 2. Synthesis of methyl (/3)-3-methyl-2-((methylamino)methyl)butanoate

To a solution of methyl (fl)-2-(((tert-butoxycarbonyl)(methyl)amino)methyl)-3-methyl butanoate (500 mg, 1.93 mmol) in DCM (5.0 mL) at 0 °C was added TFA (2.50 mL) dropwise. The resulting mixture was warmed to room temperature. After 2 h the reaction mixture was concentrated under reduced pressure to afford desired product (600 mg, crude) as a yellow solid.

Step 3: Synthesis of methyl (fl)-3-methyl-2-(((fl)-N-methyl-1-tritylaziridine-2- carboxamido)methyl)butanoate

To a solution of methyl (fl)-3-methyl-2-((methylamino)methyl)butanoate (550 mg, 3.45 mmol) and (fl)-1 -tritylaziridine-2-carboxylic acid (1.25 g, 3.80 mmol) in DCM (5.0 mL) at 0 °C was added DIPEA (1.81 mL, 10.4 mmol) followed by HATU (1.58 g, 4.15 mmol). The resulting mixture was warmed to room temperature. After 2 h the reaction was quenched with H 2 O (30 mL) and extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (50 mL), dried with NaaSO^ and concentrated under reduced pressure. Purification by silica gel column chromatography (9% EtOAc/pet. ether) afforded the desired product (300 mg, 19% yield) as a yellow oil. LCMS (ESI) m/z : [M + H] calcd for C30H34N 2 O3: 471.27; found 471.3.

Step 4: Synthesis of (fl)-3-methyl-2-(((fl)-AFmethyl-1-tritylaziridine-2- carboxamido)methyl)butanoic acid

To a solution of methyl (fl)-3-methyl-2-(((fl)-N-methyl-1-tritylaziridine-2- carboxamido)methyl)butanoate (200 mg, 0.425 mmol) in THF (2.0 mL) at 0 °C was added LiOH-H 2 O (89 mg, 2.13 mmol) in H 2 O (2.0 mL) dropwise. The resulting mixture was warmed to room temperature. After 5 h the mixture was neutralized to pH 7 with 1 M HCI. The reaction was extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (30 mL), dried with Na 2 SO 4 , and concentrated under reduced pressure to afford product (200 mg, crude) as an off-white solid. The crude product was used in the next step directly without further purification. LCMS (ESI) m/z. [M + H] calcd for C29H32N 2 O3: 457.25; found 457.2.

Intermediate A-60. Synthesis of sodium (fl)-1-methyl-5-(1-tritylaziridine-2-carboxamido)- 1 H- imidazole-2-carboxylate

Step 1: Synthesis of methyl 5-amino-1 -methyl-1 H-\ midazole-2-carboxylate A mixture of methyl 1 -methyl-5-nitro-1 H-imidazole-2-carboxylate (1.0 g, 5.401 mmol) and Pd/C (500.0 mg) in MeOH (15 mL) at room temperature was stirred under an atmosphere of hydrogen (1 atm) for 3 h. The mixture was filtered and the filter cake was washed with MeOH (3 x 20 mL). The filtrate was concentrated under reduced pressure to afford the desired product (1 .0 g, crude). LCMS (ESI) m/z. [M + Na] calcd for C6H9N3O2: 156.08; found 156.1.

Step 2. Synthesis of methyl (/3)-1 -methyl-5-(1 -tritylaziridine-2-carboxamido)-1 -imidazole-2- carboxylate

A solution of (/?)-1 -tritylaziridine-2-carboxylic acid (2.55 g, 7.741 mmol) in DCM (12.0 mL) at 0 °C was added in portions over 30 min to a solution of isobutyl chloroformate (845.1 mg, 6.187 mmol) and N- methylmorpholine (1.04 g, 10.282 mmol) in DCM. To the mixture was then added methyl 5-amino-1- methyl-1 H-imidazole-2-carboxylate (800.0 mg, 5.156 mmol) at 0 °C. The resulting mixture was warmed to room temperature and stirred overnight. The mixture was diluted with DCM (300 mL) and washed with H 2 O (3 x 100 mL), washed with brine (2 x 150 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (25% EtOAc/hexanes) to afford the final product (1.2 g, 49.9% yield). LCMS (ESI) m/z. [M + H] calcd for C^HalSUOs: 467.21 ; found 467.2.

Step 3: Synthesis of sodium -methyl-5-(1 -tritylaziridine-2-carboxamido)-1 H- imidazole-2- carboxylate

To a solution of methyl (fl)-1-methyl-5-(1-tritylaziridine-2-carboxamido)-1 -imidazole-2- carboxylate (300.0 mg, 0.643 mmol) in THF (3 mL) at room temperature was added NaOH-hfeO (38.6 mg, 0.965 mmol). The resulting solution was warmed to room temperature and stirred for 2 h. The solution was concentrated under reduced pressure to afford the desired product (400 mg, crude). LCMS (ESI) m/z. [M + H] calcd for C27H24N4O3: 453.19; found 453.2.

Intermediate A-61. Synthesis of (S)-1-methyl-5-(1-trltylazlrldlne-2-carboxamldo)-1H- imidazole-2-carboxylic acid

Step 1: Synthesis of methyl (S)-1 -methyl-5-(1 -tritylaziridine-2-carboxamido)-1 H- imidazole-2- carboxylate

To a solution of (S)-1 -tritylaziridine-2-carboxylic acid (1.18 g, 3.577 mmol) in DCM (15 mL) at 0 °C was added isobutyl chloroformate (423.4 mg, 3.100 mmol) and N-methyl morpholine (0.39 mL) 3.862 mmol). The resulting mixture was stirred for 1 h at 0 °C and then methyl 5-amino-1 -methyl-1 H-imidazole- 2-carboxylate (370.0 mg, 2.385 mmol) was added and the resulting mixture was warmed to at room temperature and stirred overnight. The reaction mixture was quenched with sat. aq. NaHCOs at 0 °C before being extracted with DCM (2 x 100 mL). The combined organic layers were washed with brine (150 mL), dried over NazSCX filtered, and concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography (100% EtOAc) to afford the final product (380 mg, 34.2% yield) as a yellow solid. LCMS (ESI) m/z. [M + Na] calcd for CaeHzelNUOs: 467.21 ; found 467.3.

Step 2. Synthesis of (S)-1 -methyl-5-(1 -tritylaziridine-2-carboxamido)-1 H- imidazole-2-carboxylic To a solution of NaOH (146.6 mg, 3.665 mmol) in H 2 O (3.6 mL) at 0 °C was added a solution of methyl (S)-1 -methyl-5-(1 -tritylaziridine-2-carboxamido)-1 H- imidazole-2-carboxylate (380.0 mg, 0.815 mmol) in MeOH (5 mL). The resulting solution was warmed to room temperature and stirred for 6 h. The mixture was acidified to pH 6 with aq. 1 M HCI before being extracted with DCM (2 x 100 mL). The combined organic layers were washed with brine (150 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (350 mg, crude). LCMS (ESI) m/z: [M + H] calcd for Ca7Ha4N 4 03: 451.18; found 451.1.

Intermediate A-62. Synthesis of 4-((2S)-1-(tert-butylsulflnyl)azirldin-2-yl)benzolc acid

Step 1: Synthesis of methyl (£)-4-(((tert-butylsulfinyl)imino)methyl)benzoate To a solution of methyl 4-formylbenzoate (100.0 mg, 0.609 mmol) and 2-methylpropane-2- sulfinamide (76.0 mg, 0.627 mmol) in DCM (2.0 mL) was added CuSO* (291.7 mg, 1.827 mmol). The resulting solution was stirred overnight at room temperature and was then filtered. The filter cake was washed with EtOAc (3 x 200 mL) and the filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (67% EtOAc/pet. ether) to afford the desired product (2 g, 61.4% yield). LCMS (ESI) m/z: [M + H] calcd for C13H17NO3S: 268.10; found 268.0.

Step 2. Synthesis of methyl 4-((2S)-1-(tert-butylsulfinyl)aziridin-2-yl)benzoate Methyl (E)-4-(((terf-butylsulfinyl)imino)methyl)benzoate (500.0 mg, 1.863 mmol), MeaS*l- (1.14 g, 5.590 mmol), and 60% NaH (134.15 mg, 5.590 mmol) were dissolved in DMSO (10.0 mL) at room temperature. The resulting mixture was stirred for 2 h and then the reaction was quenched by the addition of sat. aq. NhUCI (10 mL). The resulting mixture was extracted with EtOAc (3 x 50 mL) and the combined organic layers were washed with brine (2 x 100 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography (10% EtOAc/pet. ether) to afford the desired product (300 mg, 57.0% yield). LCMS (ESI) m/z: [M + H] calcd for C14H19NO3S: 282.12; found 282.1.

Step 3: Synthesis of 4-((2S)-1-(tert-butylsulfinyl)aziridin-2-yl)benzoic acid To a solution of methyl 4-((2S)-1-(tert-butylsulfinyl)aziridin-2-yl)benzoate (400.0 mg, 1.422 mmol) in THE (5.0 mL) and H 2 O (1.0 mL) was added LiOH (103.0 mg, 4.301 mmol). The resulting mixture was stirred overnight at room temperature and was then acidified to pH ~3 with 1 M HCI. The mixture was extracted with EtOAc (3 x 200 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The resulting residue was purified by prep-TLC (10% MeOH/DCM) to afford the desired product (130 mg, 91.2% yield). LCMS (ESI) m/z : [M - H] calcd for C13H17NO3S: 266.09; found 266.0. Intermediate A-63. Synthesis of (S)-3-methyl-2-((fl)-2-oxo-3-((fl)-1-tritylaziridine-2- carboxamido)pyrrolidin-1-yl)butanoic acid

Step 1: Synthesis of benzyl (S)-2-((fl)-3-amino-2-oxopyrrolidin-1-yl)-3-methylbutanoate

To a solution of benzyl (S)-2-((fl)-3-((tert-butoxycarbonyl)amino)-2-oxopyrrolidin-1 -yl)-3- methylbutanoate (1.0 g, 2.561 mmol) in DCM (10.0 mL) was added 4M HCI in 1 ,4-dioxane (5.0 mL) at 0 °C. The resulting mixture was stirred for 2 h at room temperature under an argon atmosphere. The resulting mixture was concentrated under reduced pressure to afford the desired crude product (890 mg, crude) as a yellow solid. LCMS (ESI) m/z: [M + H] calcd for C16H22N 2 O3: 291.17; found 291.1.

Step 2. Synthesis of benzyl (S)-3-methyl-2-((fl)-2-oxo-3-((fl)-1-tritylaziridine-2- carboxamido)pyrrolidin-1-yl)butanoate

To a solution of benzyl (S)-2-((fl)-3-amino-2-oxopyrrolidin-1-yl)-3-methylbutanoate (450.0 mg,

1.550 mmol) and (fl)-1-tritylaziridine-2-carboxylic acid (765.8 mg, 2.325 mmol) in DMF were added HATU (1.179 g, 3.100 mmol) and DIPEA (1.35 mL, 7.75 mmol) dropwise at 0 °C. The resulting mixture was stirred for 2 h at room temperature and was then extracted with EtOAc (2 x 100 mL). The combined organic layers were washed with ΗΣΟ, brine (20 mL), dried over Na 2 SO 4 . filtered, and concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography (50% EtOAc/pet. ether) to afford the desired product (470 mg, 50.4% yield). LCMS (ESI) m/z: [M + H] calcd for CMHMNSO*: 602.31 ; found 602.3.

Step 2. Synthesis of (S)-3-methyl-2-((fl)-2-oxo-3-((fl)-1-tritylaziridine-2-carbo xamido)pyrrolidin-1- yl)butanoic acid

A suspension of benzyl (S)-3-methyl-2-((fl)-2-oxo-3-((fl)-1-tritylaziridine-2- carboxamido)pyrrolidin-1-yl)butanoate (430.0 mg, 0.715 mmol) and Pd(OH)2/C (230.0 mg, 1.638 mmol) were in THF (5 mL) was stirred for 3 h under and atmosphere of hydrogen (1 atm). The resulting mixture was filtered and the filter cake was washed with MeOH (2 x 50 mL). The filtrate was concentrated under reduced pressure to afford the crude final product (16 mg, crude) as a white solid. LCMS (ESI) m/z. [M + H] calcd for C31H33N3O4: 510.24; found 510.1.

Intermediate A-64. Synthesis of potassium (S)-1-lsopropylazlrldlne-2-cartx>xylate

Step 1: Synthesis benzyl isopropyl-L-serinate To a solution of benzyl L-serinate (3.65 g, 18.69 mmol), KOAc (1.83 g, 18.69 mmol), and acetone (2.5 mL, 33.66 mmol) in DCM (60.0 mL) was added NaBH(AcO)a (4.76 g, 22.436 mmol) in portions at 0 °C. The resulting mixture was stirred overnight at room temperature. The reaction was quenched by the addition of sat. aq. NaHCOa (50 mL) at room temperature. The resulting mixture was extracted with DCM (3 x 80 mL). The combined organic layers were washed with brine (50 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (67% EtOAc/hexanes) to afford the desired product (2.7 g, 60.9% yield) as an off-white solid. LCMS (ESI) m/z: [M + H] calcd for C13H19NO3: 238.14; found 238.2.

Step 2. Synthesis of benzyl (S)-1-isopropylaziridine-2-carboxylate

To a solution of benzyl isopropyl-L-serinate (2.70 g, 11.378 mmol), EtsN (4.75 mL, 34.134 mmol) and DMAP (2.57 mg, 0.021 mmol) in DCM (50.0 mL) was added a solution of TsCI (2.60 g, 13.65 mmol) in DCM dropwise at 0 °C. The resulting mixture was stirred overnight at room temperature and was then stirred for 4 h at 40 °C. The reaction mixture was diluted with H 2 O (80 mL) and was then extracted with DCM (2 x 50 mL). The combined organic layers were washed with brine (30 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (20% EtOAc/hexanes) to afford the desired product (2.3 g, 93.2% yield). LCMS (ESI) m/z. [M + H] calcd for C13H17NO2: 220.13; found 220.1.

Step 3: Synthesis of potassium (S)-1-isopropylaziridine-2-carboxylate

To a solution of benzyl (S)-1-isopropylaziridine-2-carboxylate (800.0 mg, 3.65 mmol) and H 2 O (6.0 mL) and THF (8.0 mL) was added a solution of KOH (245.62 mg, 4.378 mmol) in H 2 O (2.0 mL) dropwise at 0 °C. The resulting mixture was stirred for 2 h at room temperature. The mixture was diluted with H 2 O (10 mL) and the aqueous layer was washed with MTBE (3 x 8 mL). The aqueous layer was dried by lyophilization to afford the desired product (400 mg, crude). LCMS (ESI) m/z. [M + H] calcd for CeHuNOz: 130.09; found 130.0.

Intermediate A-65. Synthesis of potassium (fl)-1-isopropylaziridine-2-carboxylate

Step 1: Synthesis benzyl isopropyl-D-serinate

To a solution of benzyl D-serinate (2.10 g, 10.757 mmol), KOAc (1.06 g, 10.757 mmol), and acetone (1.2 mL, 16.136 mmol) in DCM (40.0 mL) was added a solution of NaBH(AcO)s (2.96 g, 13.984 mmol) in portions at 0 °C. The resulting mixture was stirred overnight at room temperature. The reaction was quenched by the addition of sat. aq. NaHCOs (50 mL) and the mixture was extracted with DCM (3 x 50 mL). The combined organic layers were washed with brine (50 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (67% EtOAc/hexanes) to afford the desired product (1.7 g, 66.6% yield). LCMS (ESI) m/z. [M + H] calcd for C13H19NO3: 238.14; found 238.0.

Step 2. Synthesis of benzyl (fl)-1-isopropylaziridine-2-carboxylate

To a solution of benzyl isopropyl-D-serinate (1.75 g, 7.375 mmol), EtsN (2.58 mL, 18.437 mmol) and DMAP (90.09 mg, 0.737 mmol) in DCM (30.0 mL) was added a solution of TsCI (1.69 g, 8.850 mmol) in DCM dropwise at 0 °C. The resulting mixture was stirred overnight at room temperature before being stirred for 4 h at 40 °C. The mixture was diluted with H 2 O (80 mL) and then extracted with DCM (3 x 50 mL). The combined organic layers were washed with brine (50 mL), dried over Na 2 SO 4 . filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (20% EtOAc/hexanes) to afford the desired product (1.4 g, 86.6% yield). LCMS (ESI) m/z: [M + H] calcd for C13H17NO2: 220.13; found 219.9.

Step 3: Synthesis of potassium (fl)-1-isopropylaziridine-2-carboxylate

To a solution of benzyl (/3)-1-isopropylaziridine-2-carboxylate (600.0 mg, 2.736 mmol) in H 2 O (3.0 mL) and THF (5.0 mL) was added a solution of KOH (184.22 mg, 3.283 mmol) in H 2 O (2.0 mL) dropwise at 0 °C. The resulting mixture was stirred for 2 h at room temperature. The mixture was then diluted with H 2 O (10 mL) and the aqueous layer was washed with MTBE (3 x 8 mL). The aqueous layer was then dried by lyophilization to afford the desired product (260 mg, crude). LCMS (ESI) m/z: [M + H] calcd for CeHnNOa: 130.09; found 130.1.

Intermediate A-66. Synthesis of AFmethyl-N-(3-oxo-4-((fl)-1-trltylazlridlne-2- carbonyl)piperazine-1 -carbonyl)-i.-valine

Step 1: Synthesis of benzyl N-(chlorocarbonyl)-N-methyl-L-valinate

To a solution of benzyl methyl-L-valinate (2.0 g, 9.038 mmol) in DCM (20.0 mL) was added a solution of triphosgene (800 mg, 2.711 mmol) and pyridine (2.14 g, 27.113 mmol) in DCM (20.0 mL) dropwise at 0 °C. The resulting mixture was stirred for 2 h at room temperature before being diluted with EtOAc. The solution was stirred for 30 min at room temperature and was then filtered. The filtrate was concentrated under reduced pressure to afford the crude product which was used in the next step directly without further purification. LCMS (ESI) m/z. [M + H] calcd for CuHieCINOa: 284.11 ; found 283.1.

Step 2. Synthesis of benzyl N-methyl-N-(3-oxopiperazine-1-carbonyl)-L-valinate To a solution of piperazin-2-one (100.0 mg, 0.999 mmol) and EtaN (0.487 mL, 3.496 mmol) in DCM (5.0 mL) was added a solution of benzyl N-(chlorocarbonyl)-N- methyl-L-valinate (311.75 mg, 1.099 mmol) in DCM (5 mL) dropwise at 0 °C. The resulting mixture was stirred for 4 h at room temperature.

The mixture was then diluted with H 2 O (5 mL), the aqueous layer was extracted with DCM (3 x 5 mL), and the combined organic layers were concentrated under reduced pressure. The residue was purified by prep-TLC (100% EtOAc) to afford the desired product (200 mg, 57.6% yield). LCMS (ESI) m/z: [M + H] calcd for CieHasNaO^ 348.19; found 348.1.

Step 3: Synthesis of benzyl N-methyl-N-(3-oxo-4-((fl)-1 -tritylaziridine-2-carbonyl)piperazine-1 - carbonyl)-L-valinate

To a solution of (ft)-1 -tritylaziridine-2-carboxylic acid (711.11 mg, 2.159 mmol) in THF was added EtaN (0.40 mL, 2.878 mmol) and isobutyl chlorocarbonate (255.53 mg, 1.871 mmol) dropwise at 0 °C under a nitrogen atmosphere. The resulting mixture was stirred for 1 h at room temperature and then benzyl N-methyl-N-(3-oxopiperazine-1-carbonyl)-L-valinate (500.0 mg, 1.439 mmol) was added at room temperature. The resulting mixture was warmed to 70 °C and stirred overnight. The reaction was then cooled to room temperature and concentrated under reduced pressure. The residue was purified by prep- TLC (EtOAc/50% pet. ether) to afford the desired product (200 mg, 21.1% yield). LCMS (ESI) m/z. [M +

H] calcd for C40H42N4O5: 659.32; found 677.4.

Step 4: Synthesis of N-methyl-N-(3-oxo-4-((fl)-1 -tritylaziridine-2-carbonyl)piperazine-1 -carbonyl)-

L-valine

A suspension of benzyl N- methyl-N-(3-oxo-4-((fl)-1 -tritylaziridine-2-carbonyl)piperazine-1 - carbonyl)-L-valinate (140.0 mg, 0.212 mmol) and Pd/C (50.0 mg) in THF (3 mL) was stirred for 2 h under a hydrogen atmosphere (1 atm). The mixture was then filtered and the filter cake was washed with MeOH (3 x 15 mL). The filtrate was concentrated under reduced pressure to afford the desired product (100 mg, crude). LCMS (ESI) m/z. [M - H] calcd for C33H36N4OS: 567.26; found 567.1.

Intermediate A-67. Synthesis of (S)-1-(2-((tert-butyldiphenylsilyl)oxy)ethyl)azirldine-2- carboxylic acid

|/X/ OTBDPS

BnOH

DIPEA, HATU O Et j SIH, TEA o Kzco,

V BnO

MeCN V" DCM Bntr^S^NH DMSO

UOH O

OTBDPS

BnO THF, H j O

Step 1: Synthesis of benzyl (S)-1 -tritylaziridine-2-carboxylate

To a solution of (S)-1 -tritylaziridine-2-carboxylic acid (500.0 mg, 1.518 mmol), benzyl alcohol (246.2 mg, 2.277 mmol) and DIPEA (0.793 mL, 4.554 mmol) in MeCN (10.0 mL) was added HATU (1 .73 mg, 4.554 mmol). The resulting solution was stirred for 3 h at room temperature and was then concentrated under reduced pressure. The crude residue was purified by prep-TLC (50% EtOAc/pet. ether) to afford the desired product (300 mg, 47.1% yield) as an off-white slid. LCMS (ESI) m/z [M + Na] calcd for C29H25NO2: 442.18; found 442.3.

Step 2. Synthesis of benzyl (S)-aziridine-2-carboxylate

To a solution of benzyl (S)-1 -tritylaziridine-2-carboxylate (300.0 mg, 0.715 mmol) in DCM (5.0 mL) at 0 °C was added TEA (326.2 mg, 2.860 mmol) and EtaSIH (332.6 mg, 2.860 mmol). The resulting mixture was stirred at 0 °C for 3 h and was then concentrated under reduced pressure. The residue was purified by prep-TLC (10% MeOH/DCM) to afford the desired product (130 mg, 82.1% yield). LCMS (ESI) m/z. [M + H] calcd for C10H11NO2: 178.09; found 178.2.

Step 3: Synthesis of benzyl (S)-1-(2-((tert-butyldiphenylsilyl)oxy)ethyl)aziridine-2-car boxylate

To a solution of benzyl (S)-aziridine-2-carboxylate (400.0 mg, 2.257 mmol) and tert-butyl(2- iodoethoxy)diphenylsilane (1.85 g, 4.52 mmol) in DMSO (10.0 mL) was added K2CO3 (935.9 mg, 6.772 mmol) at room temperature. The mixture was stirred at 60 °C for 5 h. The mixture was diluted with H 2 O (30.0 mL) and was extracted with EtOAc (2 x 30 mL). The combined organic layers were washed with brine (2 x 50 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The resulting residue was purified by prep-TLC (20% EtOAc/pet. ether) to afford the desired product (200 mg, 15.4% yield). LCMS (ESI) m/z. [M + H] calcd for CasHssNOaSi: 460.23; found 460.0. Step 4: Synthesis of lithium (S)-1-(2-((tert-butyldiphenylsilyl)oxy)ethyl)aziridine-2-car boxylate To a solution of benzyl (S)-1-(2-((tert-butyldiphenylsilyl)oxy)ethyl)aziridine-2-car boxylate (200.0 mg, 0.435 mmol) in MeOH (2.0 mL) was added LiOH'H 2 O (36.5 mg, 0.870 mmol). The resulting mixture was stirred overnight and was then concentrated under reduced pressure to afford the desired product (200 mg, crude). LCMS (ESI) m/z: [M + H] calcd for C21H27NO3S1: 370.18; found 370.1.

Intermediate A-68. Synthesis of (fl)-1-(2-((tert-butyldiphenylsilyl)oxy)ethyl)aziridine-2- carboxylic acid

Step 1: Synthesis of methyl benzyl (fl)-1-(2-((tert-butyldiphenylsilyl)oxy)ethyl)aziridine-2- carboxylate

To a solution of benzyl (fl)-aziridine-2-carboxylate (600.0 mg, 3.386 mmol) and K2CO3 (1.87 g, 13.544 mmol) in DMSO (8.0 mL) was added tert-butyl(2-iodoethoxy)diphenylsilane (1.39 g, 3.386 mmol) in portions at room temperature. The resulting mixture was stirred at 80 °C for 16 h. The reaction mixture was then cooled to room temperature and concentrated under reduced pressure. The residue was purified by reverse phase chromatography (60→90% MeCN/H 2 O) to afford the desired product (150 mg, 9.6% yield) as a colorless solid. LCMS (ESI) m/z: [M + Na] calcd for CaeHaaNOaSi: 482.21 ; found 482.3. Step 2. Synthesis of (fl)-1-(2-((te/f-butyldiphenylsilyl)oxy)ethyl)aziridine-2-ca rboxylic acid To a solution of methyl benzyl (R)- 1 -(2-((tert-butyldiphenylsilyl)oxy)ethyl)aziridine-2-carboxyl ate (180.0 mg, 0.392 mmol) in H 2 O (2.0 mL) and THE (3.0 mL) at 0 °C was added a solution of LiOH-H 2 O (32.87 mg, 0.392 mmol) in H 2 O (1.0 mL). The resulting mixture was diluted with HzO (6.0 mL) and the aqueous layer was washed with MTBE (3 x 4 mL). The aqueous layer was dried by lyophilization which afforded the desired product (140 mg, crude). LCMS (ESI) m/z: [M + H] calcd for C21H27NO3S1: 370.18; found 370.0.

Intermediate A-69. Synthesis of 6-((2S)-1-(tert-butylsulfinyl)aziridin-2-yl)nicotinic acid

Step 1: Synthesis of methyl (E)-6-(((tert-butylsulfinyl)imino)methyl)nicotinate To a mixture of methyl 6-formylnicotinate (2.0 g, 12.11 mmol) and 2-methylpropane-2-sulfinamide (2.94 g, 24.26 mmol) in DCM (60 mL) was added CuSO* (5.80 g, 36.34 mmol). The resulting mixture was stirred at room temperature for 18 h. The reaction mixture was filtered, the filter cake was washed with DCM (3 x 30 mL), and the filtrate was concentrated under reduced pressure. Purification by normal phase chromatography (66% EtOAc/pet. ether) afforded the desired product (2.581 g, 80% yield). LCMS (ESI) m/z: [M + H] calcd for C12H16N 2 O3S: 269.10; found 269.1.

Step 2. Synthesis of 6-((2S)-1-(tert-butylsulfinyl)aziridin-2-yl)nicotinic acid To a suspension of NaH (60%, 179.76 mg, 7.491 mmol) in DMSO (20 mL) at 0 °C was added MeaS + l- (1.53 g, 7.491 mmol) and the resulting mixture was warmed to room temperature and stirred for 1 h. To the reaction mixture was added a solution of methyl (E)-6-(((tert-butylsulfinyl)imino)methyl)nicotinate (670.0 mg, 2.497 mmol) in DMSO (20 mL) in portions. The mixture was stirred at room temperature for 3 h and was then diluted with EtOAc. The mixture was acidified to pH 4 with 1 M HCI and then the aqueous layer was extracted with EtOAc. The combined organic layers were concentrated under reduced pressure. Purification by reverse phase chromatography (10→15% MeCN/H 2 O) afforded the desired product (313 mg, 45% yield). LCMS (ESI) m/z. [M + H] calcd for C12H16N 2 O3S: 269.10; found 269.1 .

Intermediate A-70. Synthesis of W-methyl-N-(N-methyl-N-((fi)-1-trltylazlrkllne-2-carbonyl)-D - alanyl)-L-vallne

Boc

ΊΠΓ

O

HATU, DIPEA UOH

DMF TOF/H j O

Step 1: Synthesis of methyl N-(N-(iert-butoxycarbonyl)-N- methyl-D-alanyl)-N-methyl-L-valinate

To a solution of methyl-L-valinate hydrochloride (1.0 g, 5.51 mmol) and N-( tert-butoxycarbonyl)- N-methyl-D-alanine (1.34 g, 6.59 mmo) in DCM (20.0 mL) at 0 °C was added EtaN (2.3 mL, 16.51 mmol) and HATU (2.72 g, 7.16 mmol). The mixture was warmed to room temperature and stirred for 4 h. The reaction mixture was then diluted with DCM (20 mL) and washed with sat. aq. NI-LCI (2 x 40 mL) and brine (40 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (20% EtOAc/pet. ether) to afford the desired product (1.5 g, 82.5% yield). LCMS (ESI) m/z: [M + H] calcd for CieHsoNaOs: 331.23; found 331.1 .

Step 2. Synthesis of methyl N-methyl-N-(methyl-D-alanyl)-L-valinate

To a solution of methyl N-(N-(tert-butoxycarbonyl)-N-methyl-D-alanyl)-N-methyl-Z_-va linate (1.50 g, 4.54 mmol) in DCM (9.0 mL) at 0 °C was added TFA (4.5 mL). The resulting mixture was warmed to room temperature and stirred for 1 h. The reaction mixture was concentrated under reduced pressure to afford the desired product (1 g, crude). LCMS (ESI) m/z. [M + H] calcd for C11H22N 2 O3: 231 .17; found 231.2.

Step 3: Synthesis of methyl W-methyl-N-(AAmethyl-N- ((fl)-1-tritylaziridine-2-carbonyl)-D-alanyl)-L- valinate

To a solution of methyl N-methyl-AA(methyl-D-alanyl)-L-valinate (900.0 mg, 3.91 mmol) and (R)- 1 -tritylaziridine-2-carboxylic acid (1.544 g, 4.689 mmol) in DMF (20.0 mL) at 0 °C was added DIPEA (3.4 mL, 19.54 mmol) and HATU (2.228 g, 5.86 mmol). The mixture was warmed to room temperature and stirred for 1 h. The reaction mixture was diluted with H 2 O (50 mL) and extracted with EtOAc (3 x 50 mL). The combined organic layers were dried over NaaSOA, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (33% EtOAc/pet. ether) to afford the desired product (1.2g, 56.7% yield). LCMS (ESI) m/z: [M + H] calcd for C33H39N3O4: 542.30; found 542.3. Step 4: Synthesis of N-methyl-N-(N-methyl-W-((fl)-1-tritylaziridine-2-carbonyl)-D -alanyl)-L-valine To a solution of methyl N-methyl-N-(N-methyl-N-((fl)-1-tritylaziridine-2-carbonyl)-0 -alanyl)-L- valinate (200.0 mg, 0.369 mmol) in THF (2.0 mL) at 0 °C was added a solution of LiOHeH 2 O (77 mg, 1.84 mmol) in H 2 O (1.85 mL). The resulting mixture was warmed to room temperature and stirred overnight. The mixture was adjusted to pH 9 with 1 M HCI and then adjusted to pH 7 with aq. NFLCI. The aqueous layer was extracted with EtOAc (3 x 20 mL). The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (200mg, crude. LCMS (ESI) m/z : [M + H] calcd for C32H37N3O4: 528.29; found 528.3.

Intermediate A-71 and A-72. Synthesis of (2/7,3S)-1 -(4-methoxybenzyl)-3- (trifluoromethyl)azlridine-2-carboxylic acid and (2S,3fi)-1-(4-methoxybenzyl)-3- (trifluoromethyl)aziridine-2-carboxylic acid

Step 1: Synthesis of ethyl 1-(4-methoxybenzyl)-3-(trifluoromethyl)aziridine-2-carboxyla te A solution of 1 -ethoxy-2, 2, 2-trifluoroethan-1 -ol (2.17 mL, 18.37 mmol) and p- methoxybenzy lami ne (1.89 mL, 14.58 mmol) in toluene (46 mL) was refluxed for 16 h under Dean-Stark conditions. The reaction was concentrated under reduced pressure and the resulting residue was dissolved in THF (80 mL) and cooled to -78 °C. BFs*EtaO (0.360 mL, 2.92 mmol) was added to the solution, followed by dropwise addition of ethyl diazoacetate (1.83 mL, 17.50 mmol). The reaction was stirred for 4 h at room temperature. The reaction mixture was quenched by addition of aq. sat. NaHCOs (5 mL), and the resulting solution was extracted with DCM (3 x 50 mL). The combined organic layers were washed with H 2 O (20 mL) and brine (10 mL). The organic phase was dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (1 →10% EtOAc/pet. ether) afford the desired product (2 g, 45.2 yield).

Step 2. Synthesis of ethyl (2f?,3S)-1 -(4-methoxybenzyl)-3-(trifluoromethyl)aziridine-2-carboxylat e and ethyl (2S,3fl)-1 -(4-methoxybenzyl)-3-(trifluoromethyl)aziridine-2-carboxylat e

Ethyl 1-(4-methoxybenzyl)-3-(trifluoromethyl)aziridine-2-carboxyla te (1 g) was purified by SFC separation (column: REGIS(S,S)WHELK-01 (250 mm * 25 mm, 10 urn); mobile phase: [Neu- IRA]; B%: 13% - 13%, min) to afford ethyl (2/?,3S)-1-(4-methoxybenzyl)-3-(trifluoromethyl)aziridine-2- carboxylate (530 mg) and ethyl (2S,3fl)-1-(4-methoxybenzyl)-3-(trifluoromethyl)aziridine-2- carboxylate (470 mg).

Step 3: Synthesis of (2/?,3S)-1 -(4-methoxybenzyl)-3-(trifluoromethyl)aziridine-2-carboxylic acid To a solution of ethyl (2f?,3S)-1-(4-methoxybenzyl)-3-(trifluoromethyl)aziridine-2- carboxylate (430 mg, 1 .42 mmol) in EtOH (4 mL) and H 2 O (6 mL) was added NaOH (113.42 mg, 2.84 mmol). The mixture was stirred at room temperature for 5 h. The mixture was acidified with aq. HCI (2M) to pH = 1 - 2. The reaction mixture was poured into H 2 O (3 mL) and the aqueous phase was extracted with EtOAc (3 x 3 mL). The combined organic phase was washed with brine (5 mL), dried over NazSO^ filtered, and concentrated under reduced pressure to afford the desired product (350 mg, 89.1% yield). LCMS (ESI) m/z. [M + H] calcd for Ci2HnFNO3:274.08; found 274.1

Step 4: Synthesis of (2S,3/3)-1-(4-methoxybenzyl)-3-(trifluoromethyl)aziridine-2- carboxylic acid To a solution of ethyl (2S,3fl)-1-(4-methoxybenzyl)-3-(trifluoromethyl)aziridine-2- carboxylate (370 mg, 1.22 mmol) in H 2 O (2 mL) and EtOH (4 mL) was added NaOH (97.59 mg, 2.44 mmol). The mixture was stirred at room temperature for 5 h. The mixture was brought to pH = 1 - 2 with the addition of aq. HCI (2 M). The reaction mixture was poured into H 2 O (3 mL) and the aqueous phase was extracted with EtOAc (3 x 3 mL). The combined organic phase was washed with brine (5 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (300 mg, 89.0% yield). LCMS (ESI) m/z. [M + H] calcd for Ci2HnFNO3:234.08; found 234.2

Intermediate A-73 and A-74. Synthesis of (2S,3S)-1-benzyl-3-(trlfluoiOmethyl)azirldlne-2- carboxylic acid and (2fl,3fl)-1-benzyl-3-(trlfluoromethyl)azirldlne-2-carboxyllc acid

Step 1: Synthesis of ethyl (2S,3/3)-2,3-dibromo-4,4,4-trifluorobutanoate To a solution of ethyl (£)-4,4,4-trifluorobut-2-enoate (5 g, 29.74 mmol, 4.42 mL) in CCU (90 mL) was added Bra (1.69 mL, 32.72 mmol) and the solution was stirred at 75 °C for 5 h. The reaction mixture was concentrated under reduced pressure to give the desired product (10.72 g, crude).

Step 2. Synthesis of ethyl (2S,3S)-1-benzyl-3-(trifluoromethyl)aziridine-2-carboxylate To a solution of ethyl (2S,3fl)-2,3-dibromo-4,4,4-trifluorobutanoate (10.72 g, 32.69 mmol) in EtOH (30 mL) was slowly added the solution of BnNHa (12.47 mL, 114.42 mmol) in EtOH (120 mL) at -5 °C under Na. The mixture was warmed to room temperature and stirred for 15 h. The mixture was concentrated under reduced pressure and EtO Ac (120 mL) was added to the residue. The precipitate was filtered off and the filtrate was washed with aqueous HCI (3%, 180 mL) and H 2 O (100 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (20% EtO Ac/pet. ether) to afford the desired product (6.02 g,67.4% yield).

Step 3: Synthesis of ethyl (2fl,3fl)-1-benzyl-3-(trifluoromethyl)aziridine-2-carboxylat e and (2S,3S)- 1 -benzyl-3-(trifluoromethyl)aziridine-2-carboxylic acid

Ethyl (2/?,3fl)-1-benzyl-3-(trifluoromethyl)aziridine-2-carboxylat e and (2S,3S)-1-benzyl-3- (trifluoromethyl)aziridine-2-carboxylic acid were synthesized in Enzyme Screening Platform, based on the procedure in Tetrahedron Asymmetry 1999, 10, 2361.

Step 5: Synthesis of (2f?,3fl)-1-benzyl-3-(trifluoromethyl)aziridine-2-carboxylic acid To a solution of ethyl (2fl,3/3)-1-benzyl-3-(trifluoromethyl)aziridine-2-carboxylat e (200 mg, 731.93 pmol) in EtOH (5 mL) was added NaOH (2 M, 548.95 pL) and the mixture was stirred at room temperature for 1 h. The reaction mixture was concentrated under reduced pressure to remove EtOH. Then to the mixture was added HCI (1 M) to adjust pH to 1 , and extracted with EtOAc (3 x 5 mL). The combined organic layers were washed with brine (2 x 10 mL), dried over Na 2 SO 4 . filtered and concentrated under reduced pressure to afford the desired product (138 mg, 76.9% yield). LCMS (ESI) m/z. [M + H] calcd for C11H10F3NO2: 246.07; found 245.9. Intermediate A-75. Synthesis of 1 -(oxetan-3-yl)aziridine-2-carboxylic acid

Step 1: Synthesis of methyl 1-(oxetan-3-yl)aziridine-2-carboxylate

To a solution of methyl 2,3-dibromopropanoate (515.46 pL, 4.07 mmol) in MeOH (15 mL) was added DIPEA (3.54 mL, 20.33 mmol). After addition, the mixture was stirred for 15 min, and then oxetan- 3-amine (297.25 mg, 4.07 mmol) was added dropwise. The resulting mixture was stirred at room temperature for 12 h. The reaction mixture was poured into H 2 O (20 mL), the aqueous phase was extracted with DCM (2 x 25mL ). The combined organic phase was washed with brine (20 mL), dried with Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (10%→30% EtOAc/pet. ether) to afford the desired product (380 mg, 59.5% yield).

Step 2. Synthesis of 1 -(oxetan-3-yl)aziridine-2-carboxylic acid

To a solution of methyl 1-(oxetan-3-yl)aziridine-2-carboxylate (280 mg, 1.78 mmol) in EtOH (3 mL) was added NaOH (2 M, 1.34 mL) at room temperature and the resulting mixture was stirred for 3 h. The reaction mixture was adjusted to pH 8 by the addition of HCI (1 M), and lyophilized to afford the desired product (200 mg, 78.4% yield).

Intermediate A-76. Synthesis of (2S,3S)-1-((S)-tert-butylsulflnyl)-3-cyclobutylazlrldlne-2- carboxylic acid

H o *N¾< o o o O

,?

·Π(Οβ) < UHMDS N •J NaOH

N' a >J< " HO' a** THF roll THF MeCNfH dr* j O

Step 1: Synthesis of (S,E)-N-(cyclobutylmethylene)-2-methylpropane-2-sulfinamide To a solution of cyclobutanecarbaldehyde (0.5 g, 5.94 mmol) in THF (10 mL) was added (S)-2- methylpropane-2-sulfinamide (792.48 mg, 6.54 mmol) and Ti(OEt)4 (2.47 mL, 11.89 mmol). The mixture was stirred at 75 °C for 3 h. The reaction mixture was cooled to room temperature and quenched by addition brine (30 mL), and filtered to remove solids. The mixture was extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (2 x 10 mL), dried over NazSO^ filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel chromatography (2%→10% EtOAc/pet. ether) to afford the desired product (907.3 mg, 39.9% yield). LCMS (ESI) m/z·. [M + H] calcd for C9H17NOS: 188.1 ; found 188.3.

Step 2. Synthesis of ethyl (2S,3S)-1-((S)-tert-butylsulfinyl)-3-cyclobutylaziridine-2-c arboxylate To a solution of ethyl 2-bromoacetate (1.60 g, 9.61 mmol, 1.06 mL) in THF (9 mL) was added LiHMDS (1 M, 9.61 mL) at -78 °C, after 2 min, (S,E)-N-(cyclobutylmethylene)-2-methylpropane-2- sulfinamide (0.9 g, 4.81 mmol) was added. The mixture was stirred at -78 °C for 2 h. The reaction mixture was quenched by addition HzO (25 mL) at -78 °C and warmed to room temperature, then the mixture extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (2 x 5 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue, which was purified by silica gel chromatography (10%→20% EtOAc/pet. ether) to afford the desired product (426 mg, crude). LCMS (ESI) m/z. [M + H] calcd for C13H23NO3S: 274.14; found 274.3.

Step 3: Synthesis of (2S,3S)-1-((S)-tert-butylsulfinyl)-3-cyclobutylaziridine-2-c arboxylic acid To a solution of (2S,3S)-1-((S)-tert-butylsulfinyl)-3-cyclobutylaziridine-2-c arboxylate (100 mg, 365.78 pmol) in MeCN (0.5 mL) and H 2 O (0.5 mL) was added NaOH (21.95 mg, 548.67 pmol) at 0 °C, the mixture was warmed to room temperature and stirred for 2 h. The reaction mixture was adjusted to pH 5 by addition aq. 10% citric acid (~10 mL) and was then extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (2 x 5 mL), dried over Na 2 SO 4 . filtered and concentrated under reduced pressure to afford the desired product (92.6 mg, crude). LCMS (ESI) m/z: [M + H] calcd for C11H19NO3S: 246.11 ; found 246.3.

Intermediate A-77. Synthesis of (2/?,3fl)-1-((fl)-tert-butylsulfinyl)-3-cyclobutylazlrldine- 2- carboxylic acid

U

Step 1: Synthesis of (f?,£)-N-(cyclobutylmethylene)-2-methylpropane-2-sulfinamid e

To a solution of cyclobutanecarbaldehyde (0.25 g, 2.97 mmol) in THE (5 mL) was added (fl)-2- methylpropane-2-sulfinamide (396.24 mg, 3.27 mmol) and Ti(OEt)4 (1.36 g, 5.94 mmol, 1.23 mL). The mixture was stirred at 75 °C for 3 h in two batches. The two batches were combined and the reaction mixture was quenched by the addition of brine (15 mL). The solution was extracted with EtOAc (3 x 20 mL) and the combined organic layers were washed with brine (2 x 5 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue, which was purified by silica gel chromatography (10%— >20% EtOAc/pet. ether) to afford the desired product (786.7 mg, 70.7% yield). LCMS (ESI) m/z. [M + H] calcd for C9H17NOS: 188.1 ; found 188.3.

Step 2. Synthesis of ethyl (2fl,3fl)-1-((fl)-tert-butylsulfinyl)-3-cydobutylaziridine-2 -carboxylate

To a solution of ethyl 2-bromoacetate (236.19 pL , 2.14 mmol) in THE (2 mL) was added UHMDS (1 M, 2.14 mL) at -78 °C, after 30 min, (/?,E)-N-(cyclobutylmethylene)-2-methylpropane-2-sulfinamide (0.2 g, 1.07 mmol) was added. The mixture was warmed to -40 °C and stirred for 4 h. The reaction mixture was quenched by addition H 2 O (18 mL) at -40 °C and warmed to room temperature The mixture was extracted with EtOAc (3 x 15 mL) and the combined organic layers were washed with brine (2 x 5 mL), dried over NazS04, filtered and concentrated under reduced pressure to give a residue, which was purified by prep-TLC (20% EtOAc/pet. ether) to afford the desired product (0.1 g, crude). LCMS (ESI) m/z: [M + H] calcd for C13H23NO3S: 274.14; found 274.3.

Step 3: Synthesis of (2f?,3f?)-1-((fl)-tert-butylsulfinyl)-3-cyclobutylaziridine- 2-carboxylic acid

In two batches, to a solution ethyl (2/7,3fl)-1 -((fl)-tert-butylsulfinyl)-3-cyclobutylaziridine-2- carboxylate (25 mg, 91.44 pmol) in MeCN (0.25 mL) and H 2 O (0.25 mL) was added NaOH (5.49 mg, 137.17 pmol) at 0 °C, the mixture was warmed to room temperature and stirred for 5 h. The reaction mixtures were combined, and adjust to pH to 5 with aq. 10% citric acid (10 mL), then extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (2 x 5 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to afford the desired product (53 mg, crude). LCMS (ESI) m/z: [M + H] calcd for C11H19NO3S: 246.11 ; found 246.2.

Intermediate A-78. Synthesis of N-methyl-AF(methyl((S)-1 -((fl)-1 -tritylaziridine-2- carbonyl)piperidin-3-yl)carbamoyl)-i.-valine

Step 1: Synthesis of tert-butyl (S)-3-(3-((S)-1 -methoxy-3-methyl-1 -oxobutan-2-yl)-1 ,3- dimethylureido)piperidine-1-carboxylate

A mixture of methyl N-(chlorocarbonyl)-W-methyl-Z.-valinate (1.94 g, 9.34 mmol) in DCM was added to a solution of (S)-tert-butyl 3-(methylamino)piperidine-1-carboxylate (2.80 g, 13.08 mmol) in DCM (18 mL) at 0 °C. The mixture was stirred at 40 °C for 3 h. The mixture was added to saturated aq. NH4CI (80 mL), and the aqueous phase was extracted with DCM (3 x 40 mL). the combined organic phase was washed with brine (50 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (30%→100% EtOAc/pet. ether) to afford the desired product (1.9 g, 55.3% yield). LCMS (ESI) m/z: [M + H] calculated for C19H35N3O5: 386.26; found 386.2.

Step 2. Synthesis of methyl W-methyl-W-(methyl((S)-piperidin-3-yl)carbamoyl)-L-valinate

To a solution of give tert-butyl (S)-3-(3-((S)-1 -methoxy-3-methyl-1 -oxobutan-2-yl)-1 ,3- dimethylureido)piperidine-1-carboxylate (1 g, 2.59 mmol) in DCM (10 mL) was added TFA (3.84 mL,

51.88 mmol) at 0 °C. The reaction was stirred at room temperature for 1 h. The mixture was added into saturated aq. Na2CC>3 (100 mL) at 0 °C to adjust to pH 9. The aqueous phase was extracted with DCM (3 x 50 mL) and the combined organic phases were washed with brine (10 mL), dried with Na2SO<, filtered and concentrated under reduced pressure to afford the desired product (710 mg, crude). LCMS (ESI) m/z: [M + H] calculated for C14H27N3O3: 286.21 ; found 286.1.

Step 3: Synthesis of methyl N-methyl-N-(methyl((S)-1 -((fl)-1 -tritylaziridine-2-carbonyl)piperidin-3- yl)carbamoyl)-L-valinate

To a solution of (fl)-1 -tritylaziridine-2-carboxylic acid (1.24 g, 2.63 mmol, 70% purity) in MeCN (5 mL) at 0 °C was added DIPEA (1.22 mL, 7.01 mmol) and HATU (1.33 g, 3.50 mmol) followed by methyl N-methyl-N-(methyl((S)-piperidin-3-yl)carbamoyl)-Z.-valinate (500 mg, 1.75 mmol). The reaction mixture was warmed to room temperature and stirred 30 min. The mixture was added to saturated aq. NI-LCI (100 mL) and the aqueous phase was extracted with DCM (3 x 50 mL). The combined organic phase was washed with brine (60 mL), dried over Na 2 SO 4 . filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (50%-» 100% EtOAc/pet. ether) to afford the desired product (650 mg, 49.7% yield). LCMS (ESI) m/z: [M + H] calculated for C36H+4N4O4: 597.34; found 597.3 Step 4: Synthesis of N-methyl-N-(methyl((S)-1 -((fl)-1 -tritylaziridine-2-carbonyl)piperidin-3- yl)carbamoyl)-L-valine

NaOH (58.34 mg, 1.46 mmol) was added to a solution of methyl N-methyl-N-(methyl((S)-1 -((fl)-1 - tritylaziridine-2-carbonyl)piperidin-3-yl)carbamoyl)-L-valin ate (640 mg, 857.97 pmol) in THE (4 mL),

MeOH (1.3 mL), and H 2 O (1.3 mL). The mixture was stirred at room temperature for 20 h. The reaction solution was directly lyophilized to afford the desired product (700 mg, crude). LCMS (ESI) m/z: [M + H] calculated for C3SH42N4O4: 583.32; found 583.4.

Intermedlate A-79. Synthesis of N-methyl- fV-(methyl((S)-1 -((fl)-1 -tritylaziridine-2- carbonyl)pyrrolkfin-3-yl)carbamoyl)-i.-vallne

Step 1: Synthesis of tert-butyl (S)-3-(3-((S)-1 -methoxy-3-methyl-1 -oxobutan-2-yl)-1 ,3- dimethylureido)pyrrolidine-1-carboxylate

A solution of methyl AA-ichlorocarbonylJ-fV-methyl-L-valinate (1.14 g, 5.49 mmol) in DCM (10 mL) was added to a solution of tert-butyl (S)-3-(methylamino)pyrrolidine-1-carboxylate (1.54 g, 7.69 mmol) in DCM (10 mL) at 0 °C. The mixture was warmed to room temperature and stirred for 2 h. The mixture was then added to sat. NhUCI (50 mL), and the aqueous phase was extracted with DCM (3 x 30 mL). The combined organic phase was washed with brine (30 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (30%→100% EtOAc/pet. ether) to afford the desired product (1.07 g, 52.5% yield).

Step 2. Synthesis of methyl N-methyl-N-(methyl((S)-pyrrolidin-3-yl)carbamoyl)-L-valinate To a solution of tert-butyl (S)-3-(3-((S)-1 -methoxy-3-methyl-1 -oxobutan-2-yl)-1 ,3- dimethylureido)pyrrolidine-1-carboxylate (1.05 g, 2.83 mmol) in DCM (11 mL) at 0 °C was added TFA (4.19 mL, 56.53 mmol). The reaction was then warmed to room temperature and stirred for 1 h. The mixture was added to sat. NaaCOa (200 mL) at 0 °C dropwise to adjust to pH 9. The aqueous phase was extracted with DCM (3 x 100 mL), and the combined organic phase was washed with brine (100 mL), dried with NaaSO^ filtered and concentrated under reduced pressure to afford the desired product (800 mg, crude).

Step 3: Synthesis of methyl N-methyl-N-(methyl(( S)-1 -(( fl)-1 -tritylaziridine-2-carbonyl)pyrrolidin-3- yl)carbamoyl)-L-valinate

To a solution of (fl)-1 -tritylaziridine-2-carboxylic acid (1 .04 g, 2.21 mmol) in MeCN (4 mL) at 0 °C was added HATU (1 .12 g, 2.95 mmol), and DIPEA (1.03 mL, 5.90 mmol) followed by methyl Af-methyl-A/- (methyl((S)-pyrrolidin-3-yl)carbamoyl)-L-valinate (400 mg, 1 .47 mmol). The mixture was warmed to room temperature and stirred for 0.5 h. The mixture was poured into NH4CI aq. (50 mL) and extracted with DCM (3 x 20 mL). The combined organic phases were washed with brine (30 mL), dried with Na 2 SO 4 , filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (50%→100% EtOAc/pet. ether) to afford the desired product (580 mg, 67.5% yield).

Step 4: Synthesis of N-methyl-N-(methyl((S)-1-((fl)-1-tritylaziridine-2-carbonyl) pyrrolidin-3- yl)carbamoyl)-L-valine

To a solution of methyl N-methyl-N-(methyl((S)-1-((fl)-1-tritylaziridine-2-carbonyl) pyrrolidin-3- yl)carbamoyl)-L-valinate (650 mg, 1.12 mmol) in THE (3.9 mL) and MeOH (1.3 mL) was added a solution of NaOH (89.23 mg, 2.23 mmol) in H 2 O (1.3 mL). The mixture was stirred at room temperature for 16 h. The reaction mixture was diluted with H 2 O (10 mL) and then lyophilized directly to afford the desired product (700 mg, crude). LCMS (ESI) m/z: [M + H] calcd for C34H40N4O4: 569.30; found, 569.4.

Intermediate A-80. Synthesis of W-methyl-N-((S)-3-methyl-4-((/7)-1-tritylazlrldine-2- carbonyl)piperazine-1 -carbonyl^L-vallne

Step 1: Synthesis of tert-butyl (S)-4-(((S)-1 -methoxy-3-methyl-1 -oxobutan-2- yl)(methyl)carbamoyl)-2-methylpiperazine-1-carboxylate

To a solution of methyl tert-butyl (S)-2-methylpiperazine-1 -carboxylate (3.31 g, 16.52 mmol) in DCM (30 mL) at 0 °C was added a solution of methyl N- (chlorocarbonyl)-N-methyl-/.-valinate in DCM (0.55 M, 30 mL). The mixture was stirred at room temperature for 30 min. The reaction mixture was diluted with HzO (30 mL) and extracted with DCM (3 x 20 mL). The combined organic layers were washed with brine (2 x 15 mL), dried over NaaSOA, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel chromatography (20%→50% EtOAc/pet. ether) to afford the desired product (5 g, 81.5% yield). LCMS (ESI) rn/z: [M + H] calcd for Ci8H33N30s:372.2; found 372.1. Sfep 2: Synthesis of methyl W-methyl-N-((S)-3-methylpiperazine-1-carbonyl)-L-valinate To tert-butyl (S)-4-(((S)-1 -methoxy-3-methyl-1 -oxobutan-2-yl)(methyl)carbamoyl)-2- methylpiperazine-1 -carboxylate (3 g, 8.08 mmol) was added a solution of 4M HCI in MeOH (30 mL). The mixture was stirred at room temperature for 1 h. The reaction mixture was adjusted to pH 8 with saturated aq. NaHCOa, and was then diluted with H 2 O (50 mL) and extracted with DCM (3 x 30 mL). The combined organic layers were washed with brine (50 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to afford the desired product (1.8 g, 82.1% yield).

Step 3: Synthesis of methyl N-methyl-N-((S)-3-methyl-4-((F?)-1-tritylaziridine-2- carbonyl)piperazine-1-carbonyl)-L-valinate

To a solution of (2fl)-1 -tritylaziridine-2-carboxylic acid (971.10 mg, 2.95 mmol) in MeCN (10 mL) was added HATU (1.35 g, 3.54 mmol), DIPEA (1.54 mL, 8.84 mmol) and methyl N-methyl-N-((S)-3- methylpiperazine-1-carbonyl)-L-valinate (0.8 g, 2.95 mmol). The mixture was stirred at room temperature for 12 h. The reaction mixture was then diluted with H 2 O (20 mL) and extracted with DCM (3 x 15 mL). The combined organic layers were washed with brine 20 mL (2 x 10 mL), dried over NazS04, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel chromatography (30%→50% EtO Ac/pet. ether) to afford the desired product (0.35 g, 20.4% yield). LCMS (ESI) m/z: [M + H] calculated for C3SH42N4O4: 583.3; found 583.2.

Step 4: Synthesis of N-methyl-AA-((S)-3-methyl-4-((A)-1-tritylaziridine-2-carbony l)piperazine-1- carbonyl)-L-valine

To a solution of methyl N-methyl-N-((S)-3-methyl-4-((/3)-1-tritylaziridine-2-carbony l)piperazine-1- carbonyl) -L-valinate (200 mg, 343.21 pmol) in H 2 O (1 mL), THF (1 mL), and MeOH (1 mL) at 0 °C was added LiOHe^O (14.40 mg, 343.21 pmol). The mixture was stirred at room temperature for 8 h and was then lyophilized directly to afford the desired product (390 mg, 98.7% yield). LCMS (ESI) m/z: [M + Na] calculated for C34H40N4O4: 591.3; found 591.2.

Intermediate A-81. Synthesis of N-methyl-N-((S)-3-methyl-4-((S)-1-trltylazlrldlne-2- carbonyl)piperazine-1 -carbonyl)- L-valine

Step 1: Synthesis of methyl N-methyl-N-((S)-3-methyl-4-((S)-1-tritylaziridine-2- carbonyl)piperazine-1-carbonyl)-L-valinate

To a solution of methyl A/-methyl-AF((S)-3-methylpiperazine-1-carbonyl)-/.-valinate (500 mg, 1.84 mmol) in MeCN (5 mL) at 0 °C was added (S)-1 -tritylaziridine-2-carboxylic acid (1.30 g, 2.76 mmol, 70% purity), HATU (1.05 g, 2.76 mmol) and DIPEA (962.85 pL, 5.53 mmol). The mixture was stirred at toom temperature for 30 min. The reaction mixture was then diluted with H 2 O (10 mL) and extracted with DCM (3 x 5 mL). The combined organic layers were washed with brine (2 x 5 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel chromatography (20%→33% EtO Ac/pet. ether) to afford the desired product (0.5 g, 46.6% yield). LCMS (ESI) m/z: [M + Na] calculated for C 3 sH4aO4N4:605.2; found 605.2

Step 2. Synthesis of N-methyl-N-((S)-3-methyl-4-((S)-1-tritylaziridine-2-carbonyl )piperazine-1- carbonyl)-L-valine

To a solution of methyl N-methyl-N-((S)-3-methyl-4-((S)-1-tritylaziridine-2-carbonyl )piperazine-1- carbonyl)-L-valinate (400 mg, 686.42 pmol) in H 2 O (2 mL), THF (2 mL), and MeOH (2 mL) at 0 °C was added LiOHeH 2 O (28.80 mg, 686.42 pmol). The mixture was stirred at room temperature for 8 h. The mixture was lyophilized directly to afford then desired product (390 mg, 98.7% yield). LCMS (ESI) m/z: [M + Na] calculated for C34H40N4O4: 591.3; found 591.2.

Intermediate A-82. Synthesis of N-methyl-W-((fl)-3-methyl-4-((/?)-1-tritylaziridir»e-2- carbonyl)piperazine-1 -carbonyl)- L-valine

Step 1: Synthesis of tert-butyl (fl)-4-(((S)-1 -methoxy-3-methy I- 1 -oxobutan-2- yl)(methyl)carbamoyl)-2-methylpiperazine-1-carboxylate

To a mixture of methyl methyl-L-valinate hydrochloride (3 g, 16.51 mmol) and DIPEA (17.26 mL, 99.09 mmol) in DCM (60 mL) at 0 °C was added bis(trichloromethyl) carbonate (2.45 g, 8.26 mmol) in one portion. The mixture was stirred at 0 °C for 30 min, then tert-butyl (fi)-2-methylpiperazine-1 -carboxylate (3.31 g, 16.51 mmol) was added to the mixture. The mixture was stirred at 0 °C for 1 h, then the pH of the solution was adjusted to 8 with sat. NaHCOa. The residue was poured into H 2 O (20 mL) and stirred for 5 min. The aqueous phase was extracted with EtOAc (2 x 20 mL), and the combined organic phase was washed with sat. NaHCOa (20 mL), dried with Na 2 SO 4 . filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (1%— *-10% EtO Ac/pet. ether) to afford the desired product (3.1 g, 50.5% yield). LCMS (ESI) m/z: [M + H] calculated for CieHaaNaOs: 372.3; found 372.2.

Step 2. Synthesis of methyl N-methyl-yV-((fl)-3-methylpiperazine-1-carbonyl)-L-valinate

To a mixture of tert-butyl (fl)-4-(((S)-1-methoxy-3-methyl-1-oxobutan-2-yl)(methyl)carb amoyl)-2- methylpiperazine-1 -carboxylate (2.5 g, 6.73 mmol) was added 4M HCI in MeOH (25 mL) at 0 °C. The mixture was stirred at room temperature for 1 h. The mixture was then concentrated under reduced pressure to afford the desired product (2 g, 96.5% yield).

Step 3: Synthesis of methyl N-methyl-N-((/3)-3-methyl-4-((/3)-1 -tritylaziridine-2- carbonyl)piperazine-1-carbonyl)-L-valinate

To a mixture of (fl)-1 -tritylaziridine-2-carboxylic acid (1.03 g, 3.12 mmol) and HATU (1.11 g, 2.92 mmol) in MeCN (1 mL) was added DIPEA (1.36 mL, 7.80 mmol) followed by methyl N-methyl-N-((fl)-3- methylpiperazine-1-carbonyl)-L-valinate (600 mg, 1.95 mmol). The mixture was stirred at room temperature for 1 h. The mixture was then concentrated under reduced pressure. The residue was purified by silica gel chromatography (1%→50% EtOAc/pet. ether) to afford the desired product (450 mg, 39.62% yield). LCMS (ESI) m/z: [M + H] calculated for C35H42N4O4: 583.3; found 583.2.

Step 4: Synthesis of N-methyl-N-((/?)-3-methyl-4-((/?)-1 -tritylaziridine-2-carbonyl)piperazine-1 - carbonyl)-L-valine

To a mixture of methyl AAmethyl-N-((fl)-3-methyl-4-((fl)-1 -tritylaziridine-2-carbonyl)piperazine-1 - carbonyl)-L-valinate (450 mg, 772.23 pmol) in H 2 O (1 mL), MeOH (1 mL), and THF (3 mL) was added LiOHeH 2 O (48.60 mg, 1 .16 mmol). The mixture was stirred at room temperature for 10 h and was then lyophilized to afford the desired product (410 mg, 92.4% yield). LCMS (ESI) m/z: [M + Na] calculated for C34H40N4O4: 591.3; found 591.3.

Intermediate A-83. Synthesis of W-methyl-W-((fl)-3-methyl-4-((S)-1-tritylaziridine-2- carbonyl)plperazine-1 -carbonyl)- L-valine

Step 1: Synthesis of methyl W-methyl-N-((fl)-3-methyl-4-((S)-1-tritylaziridine-2- carbonyl)piperazine-1-carbonyl)-L-valinate

To a mixture of (S)-1-tritylaziridine-2-carboxylic acid (1.03 g, 3.12 mmol) and HATU (1.11 g, 2.92 mmol) in MeCN (1 mL) was added DIPEA (1.36 mL, 7.80 mmol) followed by methyl N-methyl-N-((/3)-3- methylpiperazine-1-carbonyl)-L-valinate (600 mg, 1.95 mmol). The mixture was stirred at room temperature for 1 h and was then concentrated under reduced pressure. The residue was purified by silica gel chromatography (0%→50% EtO Ac/pet. ether) to afford the desired product (430 mg, 37.8% yield). LCMS (ESI) m/z: [M + H] calculated for C35H42N4O4: 583.3; found 583.2

Step 2 Synthesis of AA-methyl-N-((fl)-3-methyl-4-((S)-1 -tritylaziridine-2-carbonyl)piperazine-1 - carbonyl)-L-valine

To a mixture of methyl /\Amethyl-N-((fl)-3-methyl-4-((S)-1 -tritylaziridine-2-carbonyl)piperazine-1 - carbonyl)-L-valinate (430 mg, 737.91 μιτιοΙ) in H 2 O (1 mL), MeOH (1 mL), and THE (3 mL) was added UOHeH 2 O (46.44 mg, 1 .11 mmol). The mixture was stirred at room temperature for 10 h and was then lyophilized to afford the desired product (370 mg, 87.3% yield). LCMS (ESI) m/z: [M + Na] calculated for C34H40N4O4: 591.3; found 591.3.

Intermediate A-84. Synthesis of A/-((fl)-4-(ferM>utoxycarbonyl)-2-methylplperazine-1- caitx>nyl)-A/-methyl-L-valine

Step 1: Synthesis of methyl N-(chlorocarbonyl)-N-methyl-L-valinate

To a solution of methyl methyl-L-valinate hydrochloride (1.8 g, 9.91 mmol) in DCM (20 mL) at 0 °C was added DIPEA (5.18 mL, 29.73 mmol) followed by bis(trichloromethyl) carbonate (1.47 g, 4.95 mmol). The mixture was stirred at 0 °C for 20 min. The reaction mixture used for the next step directly without workup.

Step 2. Synthesis of tert-butyl (fl)-4-(((S)-1 -methoxy-3-methyl-1 -oxobutan-2- yl)(methyl)carbamoyl)-3-methylpiperazine-1-carboxylate

A solution of methyl AA-(chlorocarbonyl)-A/-methyl-L-valinate (1.03 g, 4.96 mmol) in DCM (10 mL) was added to a solution of tert-butyl (3fl)-3-methylpiperazine-1 -carboxylate (993.41 mg, 4.96 mmol) in DCM (1 mL) at 0 °C. The mixture was then stirred at 0 °C for 15 min. The mixture was added to aq. NH4CI (10 mL) and the solution was then extracted with DCM (3 x 10 mL). The combined organic phase was washed with brine (2 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (0%→50% EtOAc/pet. ether) to afford the desired product (750 mg, 36.2% yield).

Step 3: Synthesis of N-((fl)-4-(tert-butoxycarbonyl)-2-methylpiperazine-1 -carbonyl)-N-methyl-L- valine

To a solution of tert-butyl (fl)-4-(((S)-1 -methoxy-3-methyl-1 -oxobutan-2-yl)(methyl)carbamoyl)-3- methylpiperazine-1 -carboxylate (700 mg, 1.88 mmol) in THF (0.5 mL) and H 2 O (0.5 mL) at 0 °C was added LiOH'H 2 O (237.23 mg, 5.65 mmol). The mixture was stirred at room temperature for 3 h. The pH of the reaction mixture was adjusted to 6 - 7 with 1 N HCI. The mixture was extracted with EtOAc (3 x 10 mL), dried over Na 2 SO 4 , and concentrated under reduced pressure to afford the desired product (600 mg, 85.5% yield).

Intermediate A-85. Synthesis of (S)-3-methyl-2-((fl)-2-oxo-3-((S)-1-tritylaziridine-2- carboxamido)pyrrolidin-1 -yl)butanolc acid

Step 1: Synthesis of benzyl (S)-3-methyl-2-((f7)-2-oxo-3-((S)-1-tritylaziridine-2- carboxamido)pyrrolidin-1-yl)butanoate

To a mixture of benzyl (2S)-2-[(3fl)-3-amino-2-oxopyrrolidin-1-yl]-3-methylbutanoat e (420.0 mg,

1.446 mmol), DIPEA (934.73 mg, 7.232 mmol) and (2S)-1-(triphenylmethyl)aziridine-2-carboxylic acid (619.40 mg, 1.880 mmol) in DMF (5 mL) at 0 °C was added HATU (659.99 mg, 1.736 mmol). The resulting mixture was stirred at room temperature for 2 h. The reaction mixture was quenched with H 2 O. The resulting mixture was extracted with EtOAc (2 x 10 mL), and the combined organic layers were washed with brine (10 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (30% EtOAc/pet. ether) to afford the desired product (480 mg, 55.2% yield). LCMS (ESI) m/z. [M - H]- calcd for CMHMNSOA: 600.29; found 600.3

Step 2. Synthesis of (S)-3-methyl-2-((fl)-2-oxo-3-((S)-1-tritylaziridine-2-carbox amido)pyrrolidin-1- yl)butanoic acid

A suspension of benzyl (2S)-3-methyl-2-[(3fl)-2-oxo-3-[(2S)-1-(triphenylmethyl)azir idine-2- amido]pyrrolidin-1 -yljbutanoate (450.0 mg, 0.748 mmol) and Pd/C (200 mg) in THF (5 mL) at room temperature was stirred for 3 h under a hydrogen atmosphere. The mixture was then filtered and concentrated under reduced pressure to afford the desired product (400 mg, crude). LCMS (ESI) m/z. [M - H] calcd for CaiHsaNsOA: 510.24; found 510.2.

Intermediate A-86. Synthesis of (S)-2-(8-(ferFbutoxycarbonyl)-1-oxo-2,8- diazaspiro[4.5]decan-2-yl)-3-methylbutanoic acid

Sfep f: Synthesis of 1 -(tert-butyl) 4-methyl 4-allylpiperidine-1 ,4-dicarboxylate To a solution of 1 -tert-butyl 4-methyl piperidine-1 ,4-dicarboxylate (5.0 g, 20.551 mmol) in THF (50 mL) at -78 °C was added LiHMDS (27 mL, 26.714 mmol, 1 M in THF) followed by allyl bromide (3.23 g, 26.716 mmol). The resulting mixture was stirred overnight at room temperature. The reaction was quenched with sat. aq. NHACI (aq.) and the combined organic layers were washed with brine (3 x 100 mL), dried over Na 2 SO 4 . filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (50% EtOAc/pet. ether) to afford the desired product (4.5 g, 73.4% yield).

Step 2. Synthesis of 1 -(tert-butyl) 4-methyl 4-(2-oxoethyl)piperidine-1 ,4-dicarboxylate

To a solution of 1 -tert-butyl 4-methyl 4-(prop-2-en-1-yl)piperidine-1 ,4-dicarboxylate (1.0 g, 3.529 mmol) and KaOsO^H 2 O (1.3 g, 3.529 mmol) in 1 ,4-dioxane (5 mL) and H 2 O (5mL) at 0 °C was added Nal04 (1.51 g, 7.058 mmol). The resulting mixture was stirred at room temperature for 5 h. The mixture was extracted with EtOAc (3 x 50 mL) and the combined organic layers were washed with H 2 O (3 x 100 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The crude product was used in the next step directly without further purification to afford the desired product (800 mg, 75.% yield). LCMS (ESI) m/z. [M - H] calcd for CuHasNOs: 284.16; found 284.0.

Sfep 3: Synthesis of 1 -(tert-butyl) 4-methyl (S)-4-(2-((1 -(benzyloxy)-3-methyl-1 -oxobutan-2- yl)amino)ethyl)piperidine-1 ,4-dicarboxylate

To a solution of 1 -tert-butyl 4-methyl 4-(2-oxoethyl)piperidine-1 ,4-dicarboxylate (4.0 g, 14.018 mmol) and benzyl (2S)-2-amino-3-methylbutanoate (3.49 g, 16.822 mmol) in MeOH (40 mL) at 0 °C was added ZnCIa (2.10 g, 15.420 mmol) and NaBHaCN (1.76 g, 28.037 mmol). The resulting mixture was stirred at room temperature for 2 h. The reaction was quenched with sat. aq. NhLCI and the combined organic layers were washed with brine (3 x 100 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (50% EtOAc/pet. ether) to afford the desired product. LCMS (ESI) m/z. [M + H] calcd for CzeH^NaOe: 477.29; found 477.3.

Step 4: Synthesis of tert-butyl (S)-2-(1 -(benzyloxy)-3-methyl-1 -oxobutan-2-yl)-1 -oxo-2,8- diazaspiro[4.5]decane-8-carboxylate

To a solution of 1 -tert-butyl 4-methyl 4-(prop-2-en-1 -yl)piperidine-1 ,4-dicarboxylate (2.20 g, 4.616 mmol) and DIPEA (5.97 g, 46.159 mmol) in toluene was added DMAP (0.56 g, 4.616 mmol) in portions at 120 °C. The resulting mixture was stirred overnight at 120 °C. The reaction was cooled to room temperature and quenched with sat. aq. NHACI. The combined organic layers were washed with brine (3 x 100 mL), dried over NazS04, filtered, and concentrated under reduced pressure. The residue was purified by Prep-TLC (50% EtOAc/pet. ether) to afford the desired product (1 .5 g, 50.2% yield). LCMS (ESI) m/z. [M + H] calcd for CzsHaeNaOs: 445.26; found 445.3. Step 5: Synthesis of (S)-2-(8-(tert-butoxycarbonyl)-1-oxo-2,8-diazaspiro[4.5]deca n-2-yl)-3- methylbutanoic acid

To a solution of tert-butyl 2-[(2S)-1 -(benzyloxy)-3-methyl-1 -oxobutan-2-yl]-1 -oxo-2,8- diazaspiro[4.5]decane-8-carboxylate (2.40 g, 5.398 mmol) in toluene (25 mL) at room temperature was added Pd/C (2.40 g, 22.552 mmol). The resulting suspension was stirred overnight at room temperature under an H2 atmosphere. The mixture was concentrated under reduced pressure, filtered, the filter cake washed with EtOAc (3 x 50 mL), and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (50% EtOAc/pet. ether) to afford the desired product (2. 2 g, 72.5% yield). LCMS (ESI) m/z. [M - H] calcd for C18H30N 2 O5: 353.22; found 353.2.

Intermediate A-87. Synthesis of N-methyl- N-(3-oxo-4-((S)-1 -tritylazirldlne-2- carbonyl)plperazlne-1 -carbonyl)- L-valine

Step 1: Synthesis of benzyl N-methyl-N-(3-oxo-4-((S)-1 -tritylaziridine-2-carbonyl)piperazine-1 - carbonyl)-L-valinate

To a solution of (2S)-1-(triphenylmethyl)aziridine-2-carboxylic acid (2.13 g, 6.466 mmol) in THF (10 mL) at 0 °C was added EtsN (0.87 g, 8.598 mmol) and isobutyl chlorocarbonate (1.44 g, 10.54 mmol). The resulting mixture was stirred at room temperature for 1 h, and then benzyl (2S)-3-methyl-2-[methyl(3- oxopiperazine-1 -carbonyl)amino]butanoate (1.50 g, 4.318 mmol) was added. The resulting mixture was stirred overnight at 70 °C. The reaction mixture was then concentrated under reduced pressure. The residue was purified by Prep-TLC (50% EtOAc/pet. ether) to afford the desired product (900 mg, 31.6% yield). LCMS (ESI) m/z. [M - H] calcd for C40H42N4O5: 657.32; found 657.1.

Step 2. Synthesis of N-methyl-N-(3-oxo-4-((S)-1 -tritylaziridine-2-carbonyl)piperazine-1 -carbonyl)-

L-valine

A solution of benzyl N-methyl-N-(3-oxo-4-((S)-1-tritylaziridine-2-carbonyl)pipera zine-1 -carbonyl)- L-valinate (500 mg) and Pd/C (50 mg) in THF (5 mL) was stirred for 2 h at room temperature under a hydrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with MeOH (3 x 30 mL), and the filtrate was concentrated under reduced pressure to afford the desired product (460 mg, crude). LCMS (ESI) m/z. [M - H] calcd for CaaHael^Os: 567.27; found 567.1.

Intermediate A-88. Synthesis of lithium (fl)-1-(3-methoxypropyl)azlridlne-2-carboxylate

Step 1: Synthesis of benzyl (fl)-1-(3-methoxypropyl)aziridine-2-carboxylate To a mixture of benzyl (fl)-aziridine-2-carboxylate (350.0 mg, 1.975 mmol) and K2CO3 (545.95 mg, 3.950 mmol) in DMSO (4 mL) at 60 °C was added 1 -iodo-3-methoxypropane (790.13 mg, 3.950 mmol). The resulting mixture was stirred for 2 h and was then cooled to room temperature, diluted with brine (50 mL), and extracted with EtOAc (3 x 20 mL). The combined organic layers were concentrated under reduced pressure. The crude product was purified by reverse phase chromatography (30%→38% MeCN/EfeO) to afford the desired product (170 mg, 31.1% yield). LCMS (ESI) m/z. [M + H] calcd for C14H19NO3: 250.14; found 250.2.

Step 2. Synthesis of lithium (fl)-1-(3-methoxypropyl)aziridine-2-carboxylate A mixture of benzyl (fl)-1-(3-methoxypropyl)aziridine-2-carboxylate (170 mg, 0.682 mmol) and LiOH (57.23 mg, 1.364 mmol) in MeOH (2 mL) was stirred at 0 °C for 1 h. The mixture was concentrated under reduced pressure to afford the desired product (200 mg, crude). LCMS (ESI) m/z. [M + H] calcd for C7H13NO3: 160.09; found 160.3.

Intermediate A-89. Synthesis of lithium (S)-1-(3-methoxypropyl)aziridlne-2-carboxylate

Step 1: Synthesis of benzyl (S)-1-(3-methoxypropyl)aziridine-2-carboxylate

To a mixture of benzyl (S)-aziridine-2-carboxylate (250 mg, 1.411 mmol) and K2CO3 (389.96 mg,

2.822 mmol) in DMSO (4 mL) at 60 °C was added 1 -iodo-3-methoxypropane (564.38 mg, 2.822 mmol).

The resulting mixture was stirred for 2 h and was then cooled to room temperature, diluted with brine (50 mL), and extracted with EtOAc (3 x 20 mL). The combined organic layers were concentrated under reduced pressure. The crude product was purified by reverse phase chromatography (25%→40% H 2 O/MeCN) to afford the desired product (234 mg, 63.2% yield). LCMS (ESI) m/z. [M + H] calcd for C14H19NO3: 250.14; found 250.2.

Step 2. Synthesis of lithium (S)-1-(3-methoxypropyl)aziridine-2-carboxylate A mixture of benzyl (S)-1 -(3-methoxypropyl) aziridine-2-carboxylate (230 mg, 0.923 mmol) and ϋΟΗ·Η2θ (77.43 mg, 1 .845 mmol) in MeOH (3 mL) was stirred for 1 h at 0 °C. The resulting mixture was concentrated under reduced pressure to afford the desired product (320 mg, crude). LCMS (ESI) m/z. [M + H] calcd for C7H13NO3: 160.09; found 160.1.

Intermediate A-90. Synthesis of terEbutyl (S)-2-((S)-1-(benzyloxy)-3-methyl-1-oxobutan-2- yl)-1-oxo-2,7-dlazasplro[4.5]decane-7-carboxylate and tert-butyl (fl)-2-((S)-1 -(benzyloxy)-3-methyl- 1 -oxobutan-2-yl)-1 -oxo-2,7-diazaspiro[4.5]decane-7-carboxylate

Step 1: Synthesis of 1 -(tert-butyl) 3-methyl 3-allylpiperidine-1 ,3-dicarboxylate To a solution of 1 -tert-butyl 3-methyl piperidine-1 ,3-dicarboxylate (10.0 g, 41.101 mmol) and LiHMDS (82 mL, 82.202 mmol, 1 M in THE) in THE (100 mL) at -78 °C was added allyl bromide (9.94 g, 82.202 mmol). The reaction was warmed to room temperature and stirred overnight. The solution was then quenched with sat. aq. NhUCI and diluted with EtOAc (500 mL). The organic layer was washed with brine (3 x 150 mL), dried over NazSO*, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford the desired product (9.9 g, 85% yield). LCMS (ESI) m/z: [M + H] calcd for C15H25NO4: 284.18; found 284.0.

Step 2. Synthesis of 1 -(tert-butyl) 3-methyl 3-(2-oxoethyl)piperidine-1 ,3-dicarboxylate To a solution of 1 -(tert-butyl) 3-methyl 3-allylpiperidine-1 ,3-dicarboxylate1 -(tert-butyl) 3-methyl 3- allylpiperidine-1 ,3-dicarboxylate (9.1 g, 32.114 mmol) and 2,6-lutidine (6.88 g, 64.227 mmol) in dioxane (180 mL) and H 2 O (180 mL) at 0 °C was added K20s04*2H20 (591.61 mg, 1 .606 mmol). The resulting mixture was stirred for 15 min at room temperature and was then cooled to at 0 °C and Nal04 (27.47 g, 128.455 mmol) was added in portions. The resulting mixture was stirred for 3 h at room temperature. And thee reaction was then quenched with sat. aq. NaaSaOa at 0 °C. The resulting mixture was extracted with EtOAc (2 x 500 mL), and thee combined organic layers were washed with 1 M HCI (2 x 200 mL), brine (2 x 200 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (7.5 g, 81.9% yield). LCMS (ESI) m/z: [M + H] calcd for C14H23NO5: 286.16; found 286.1.

Step 3: Synthesis of 1 -(tert-butyl) 3-methyl 3-(2-(((fl)-1 -(benzyloxy)-3-methyl-1 -oxobutan-2- yl)amino)ethyl)piperidine-1 ,3-dicarboxylate

To a solution of 1 -(tert-butyl) 3-methyl 3-(2-oxoethyl)piperidine-1 ,3-dicarboxylate (9.0 g, 31.541 mmol) and benzyl (2S)-2-amino-3-methylbutanoate (7.19 g, 34.695 mmol) in MeOH (90 mL) at 0 °C was added ZnClz (4.73g, 34.695 mmol) and NaBHaCN (3.96g, 63.083 mmol). The resulting mixture was stirred overnight at room temperature. Desired product could be detected by LCMS, and it was concentrated under reduced pressure and extracted with EtOAc (1200 mL). The organic layer was washed with brine (3 x 150 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by reverse phase chromatography afforded the desired product (9.9 g, 65.9% yield). LCMS (ESI) m/z. [M + H] calcd for C26H40N 2 O6: 477.29; found 477.2.

Step 4: Synthesis of tert-butyl (S)-2-((S)-1-(benzyloxy)-3-methyl-1-oxobutan-2-yl)-1-oxo-2,7 - diazaspiro[4.5]decane-7-carboxylate and tert-butyl (fl)-2-((S)-1 -(benzyloxy)-3-methyl-1 -oxobutan-2-yl)-1 - oxo-2, 7-diazaspiro[4.5]decane-7-carboxylate

To a solution of 1 -(tert-butyl) 3-methyl 3-(2-(((fl)-1 -(benzyloxy)-3-methyl-1 -oxobutan-2- yl)amino)ethyl)piperidine-1 ,3-dicarboxylate (9.9 g, 20.772 mmol) and DIPEA (26.84 g, 207.715 mmol) in toluene (100 mL) was added DMAP (5.07 g, 41.543 mmol). The resulting mixture was stirred at 80 °C for 50 h. The resulting mixture was concentrated under reduced pressure and the residue was taken up in

EtOAc (1000 mL). The organic layer was washed with brine (3 x 150 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The crude product was purified by CHIRAL-HPLC (50%

EtOH/Hex) to afford tert-butyl (S)-2-((S)-1-(benzyloxy)-3-methyl-1-oxobutan-2-yl)-1-oxo-2,7 - diazaspiro[4.5]decane-7-carboxylate (1 .75 g) and tert-butyl (fl)-2-((S)-1-(benzyloxy)-3-methyl-1-oxobutan- 2-yl)-1 -oxo-2, 7-diazaspiro[4.5]decane-7-carboxylate (1 .98 g). LCMS (ESI) m/z. [M + H] calcd for

C25H36N 2 O5: 445.26; found 445.2.

Intermediates A-91 and A-92. Synthesis of (S)-2-{(S)-7-(terM>utoxycarbonyl)-4-oxo-1-oxa- 3,7-diazaspiro[4.4]nonan-3-yl)-3-methylbutanoic acid and (S)-2-((fl)-7-(tert-butoxycarlx>nyl)-4-oxo- 1-oxa-3,7-diazaspiro[4.4lnonan-3-yl)-3-methylbutanoic acid

Step 1: Synthesis of tert-butyl 3-hydroxy-3-(((S)-1 -methoxy-3-methyl-1 -oxobutan-2- yl)carbamoyl)pyrrolidine-1-carboxylate

To a solution of 1-(tert-butoxycarbonyl)-3-hydroxypyrrolidine-3-carboxylic acid (800 mg, 3.46 mmol) and DIPEA (3.01 mL, 17.3 mmol) in DMF (10 mL) at 0 °C was added methyl L-valinate (681 mg, 5.19 mmol) and HATU (1.71 g, 4.497 mmol). The resulting mixture was warmed to room temperature and stirred for 2 h then diluted with H 2 O (20 mL) and extracted into EtOAc (3 x 20 mL). The combined organic layers were washed with brine (2 x 20 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by reverse phase chromatography (30→55% MeCN/H 2 O, 0.1% NH4HCO3) afforded the desired product (1 g, 76% yield). LCMS (ESI) m/z : [M + Na] calcd for CieHaeNaOe: 367.18; found 366.9.

Step 2. Synthesis of (S)-2-((S)-7-(tert-butoxycarbonyl)-4-oxo-1-oxa-3,7-diazaspir o[4.4]nonan-3- yl)-3-methylbutanoic acid and (S)-2-((fl)-7-(tert-butoxycarbonyl)-4-oxo-1 -oxa-3,7-diazaspiro[4.4]nonan-3- yl)-3-methylbutanoic acid

To a solution of tert-butyl 3-hydroxy-3-(((S)-1 -methoxy-3-methyl-1 -oxobutan-2- yl)carbamoyl)pyrroiidine-1-carboxylate (1.0 g, 2.90 mmol) and CsaCOa (1.89 g, 5.81 mmol) in MeCN (15 mL) at 0 °C was added paraformaldehyde (436 mg, 14.5 mmol). The resulting mixture was heated to 80 °C and stirred overnight. Purification by reverse phase chromatography (10→40% MeCN/H 2 O, 0.1% NH4HCO3) afforded a mixture of the desired products. The diastereomers were separated by prep-SFC (30% EtOH/hexanes, 0.3% TFA) to afford (S)-2-((S)-7-(tert-butoxycarbonyl)-4-oxo-1-oxa-3,7- diazaspiro[4.4]nonan-3-yl)-3-methylbutanoic acid (250 mg, 24% yield) and (S)-2-((fl)-7-(tert- butoxycarbonyl)-4-oxo-1-oxa-3,7-diazaspiro[4.4]nonan-3-yl)-3 -methylbutanoic acid (200 mg, 19% yield). LCMS (ESI) m/z. [M + Na] calcd for CieHaeNaOe: 365.17; found 365.0.

Intermediate A-93. Synthesis of (S)-1-((3-methyloxetan-3-yl)methyl)aziridine-2-carboxyllc acid

Step 1: Synthesis of benzyl (S)-1 -tritylaziridine-2-carboxylate

To a mixture of (S)-1 -tritylaziridine-2-carboxylic acid (3.0 g, 9.11 mmol) and benzyl bromide (2.16 mL, 18.22 mmol) in DMF (30 mL) was added K2CO3 (2.25 g, 18.22 mmol) and Kl (76 mg, 455 μιτιοΙ). The reaction mixture was heated to 50 °C and stirred for 30 min then was cooled to room temperature and diluted with H 2 O (30 mL) and EtOAc (30 mL). The aqueous layer was extracted with EtOAc (3 x 40 mL), and the combined organic layers were washed with brine (5 x 70 mL), dried over NaaSO^ filtered, and concentrated under reduced pressure to afford the desired product (4.7 g, crude).

Step 2. Synthesis of benzyl (S)-aziridine-2-carboxylate To a mixture of benzyl (S)-1-tritylaziridine-2-carboxylate (3.4 g, 8.10 mmol) in MeOH (17.5 mL) and CHCla (17.5 mL) at 0 °C was added TFA (9.0 mL, 122 mmol). The reaction mixture was stirred for 30 min then was poured into sat. aq. NaHCOa (50 mL), extracted into DCM (4 x 35 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by silica gel column chromatography (6→100% EtOAc/pet. ether) afforded the desired product (445 mg, 31 % yield).

Step 3: Synthesis of benzyl (S)-1-((3-methyloxetan-3-yl)methyl)aziridine-2-carboxylate

To a mixture of benzyl (S)-aziridine-2-carboxylate (440 mg, 2.48 mmol) and 3-(iodomethyl)-3- methyloxetane (2.11 g, 9.93 mmol) in DMA (5 mL) was added K2CO3 (1.72 g, 12.42 mmol) and 18-crown- 6 (32.8 mg, 124 pmol). The reaction mixture was heated to 80 °C and stirred for 12 h, and was then was diluted with H 2 O (25 mL) and EtOAc (25 mL). The aqueous layer was extracted with EtOAc (3 x 20 mL), and the combined organic layers were washed with brine (5 x 45 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by prep-TLC (50% EtOAc/pet. ether) afforded the desired product (367 mg, 57% yield). LCMS (ESI) m/z: [M + H] calcd for C15H19NO3: 262.14; found 262.0. Step 4: Synthesis of (S)-1-((3-methyloxetan-3-yl)methyl)aziridine-2-carboxylic acid To a mixture of benzyl (S)-1-((3-methyloxetan-3-yl)methyl)aziridine-2-carboxylate (100 mg, 383 pmol) in MeCN (500 pL) and H 2 O (500 pL) at 0 °C was added NaOH (23 mg, 574 pmol). The reaction mixture was stirred at 0 °C for 1 h then was concentrated under reduced pressure to afford the desired product (100 mg, crude). LCMS (ESI) m/z: [M + H] calcd for CeHiaNOa: 172.10; found 172.0.

Intermediate A-94. Synthesis of (2/7,3 fl)-1-(tert-butylsulfinyl)-3-ethylazirldine-2-carboxyllc acid

Step 1: Synthesis of (fl,£)-2-methyl-N-propylidenepropane-2-sulfinamide To a solution of propionaldehyde (6.27 mL, 86.1 mmol) in THF (200 mL) was added (fl)- 2- methylpropane-2-sulfinamide (10.4 g, 86.1 mmol) and titanium ethoxide (51 mL, 170 mmol). The reaction mixture was heated to 70 °C for 3 h then cooled to room temperature and quenched with H 2 O (50 mL), filtered, and extracted into EtOAc (3 x 30 mL). The combined organic layers were washed with brine (30 mL), dried over Na 2 SO 4 . filtered, and concentrated under reduced pressure. Purification by silica gel column chromatography (9→17% EtOAc/pet. ether) afforded the desired product (4.0 g, 29% yield).

Step 2. Synthesis of ethyl (2fl,3f?)-1-(te/1-butylsulfinyl)-3-ethylaziridine-2-carboxyl ate To a solution of ethyl 2-bromoacetate (2.74 mL, 24.8 mmol) in THF (40 mL) at -78 °C was added LiHMDS (24.80 mL, 1 M in THF). After 30 min (f?,E)-2-methyl-N-propylidenepropane-2-sulfinamide (2.0 g, 12.4 mmol) in THF (20 mL) was added to the reaction mixture. The mixture was stirred for 1 h then warmed to room temperature, quenched with H 2 O (50 mL), and extracted into EtOAc (3 x 50 mL). The combined organic layers were washed with brine (2 x 50 mL), dried over Na 2 SO 4 . filtered, and concentrated under reduced pressure. Purification by silica gel column chromatography (17→25% EtOAc/pet. ether) afforded product (1.34 g, 44% yield). LCMS (ESI) m/z: [M + H] calcd for C11H21NO3S: 248.13; found 248.1.

Step 3: Synthesis of (2/?,3f?)-1-(tert-butylsulfinyl)-3-ethylaziridine-2-carboxyl ic acid To a solution of ethyl (2/?,3fl)-1-(iert-butylsulfinyl)-3-ethylaziridine-2-carboxyl ate (600 mg, 2.4 mmol) in MeOH (3 mL) and H 2 O (3 mL) was added LiOH (70 mg, 2.9 mmol). The resulting mixture was stirred for 16 h then diluted with H 2 O (20 mL) and washed with DCM (3 x 10 mL). Lyophilization of the aqueous layer afforded product (600 mg, crude). LCMS (ESI) m/z. [M + H] calcd for C9H17NO3S: 220.10; found 220.3.

Intermediate A-95. Synthesis of (2S,3S)-1-(ferMxjtylsulfinyl)-3-ethylaziridine-2-carboxylic acid

Step 1: Synthesis of (S,£)-2-methyl-N-propylidenepropane-2-sulfinamide To a solution of propionaldehyde (6.27 mL, 86.1 mmol) in THF (50 mL) was added (S)-2- methylpropane-2-sulfinamide (10.4 g, 86.1 mmol) and titanium ethoxide (51 mL, 170 mmol). The reaction mixture was heated to 70 °C for 3 h then cooled to room temperature and quenched with H 2 O (30 mL), filtered, and extracted into DCM (3 x 100 mL). The combined organic layers were washed with brine (10 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by silica gel column chromatography (25% EtO Ac/pet. ether) afforded product (4.6 g, 33% yield).

Step 2. Synthesis of ethyl (2S,3S)-1-(tert-butylsulfinyl)-3-ethylaziridine-2-carboxylat e To a solution of ethyl 2-bromoacetate (2.74 mL, 24.8 mmol) in THF (40 mL) at -78 °C was added □HMDS (24.80 mL, 1M in THF). After 30 min (S,E)-2-methyl-N-propylidenepropane-2-sulfinamide (2.0 g, 12.4 mmol) in THF (20 mL) was added to the reaction mixture. The mixture was stirred for 1 h then warmed to room temperature, quenched with H 2 O (20 mL), and extracted into EtO Ac (3 x 20 mL). The combined organic layers were washed with brine (2 x 25 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by reverse phase chromatography (31→51% MeCN/H 2 O, 10 mM NH4HCO3) afforded product (600 mg, 20% yield). LCMS (ESI) m/z [M + H] calcd for C11H21NO3S: 248.13; found 248.1.

Step 3: Synthesis of (2S,3S)-1-(terFbutylsulfinyl)-3-ethylaziridine-2-carboxylic acid To a solution of ethyl (2S,3S)-1-(tert-butylsulfinyl)-3-ethylaziridine-2-carboxylat e (600 mg, 2.4 mmol) in MeOH (300 pL) and H 2 O (300 pL) was added LiOH (87 mg, 3.6 mmol). The resulting mixture was stirred for 12 h then diluted with H 2 O (20 mL) and washed with DCM (3 x 10 mL). Lyophilization of the aqueous layer afforded product (600 mg, crude). LCMS (ESI) m/z. [M + H] calcd for C9H17NO3S: 220.10; found 220.2. Intermediate A-96. Synthesis of (2fl,3fl)-3-lsopropyl-1-tritylazlrldlne-2-carboxyllc acid

Step 1: Synthesis of ( £)-4-methylpent-2-enoic acid

Two batches of a solution of malonic acid (25.0 mL, 240 mmol), isobutyraldehyde (34.7 mL, 380 mmol) and morpholine (380 μ!_, 4.32 mmol) in pyridine (75 mL) were stirred for 24 h then were heated to 115 °C and stirred for 12 h. The combined reaction mixtures were poured into H2SO4 (1 M, 800 mL) and extracted into EtOAc (3 x 300 mL). The combined organic layers were washed with brine (300 mL), dried over N32S04, filtered, and concentrated under reduced pressure. The residue was dissolved in NaOH (1 M, 500 mL), washed with EtOAc (2 x 200 mL), acidified to pH = 4 - 2 with HCI (4M), and extracted into EtOAc (3 x 300 mL). The combined organic layers were washed with brine (300 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure which afforded product (54 g, 98% yield).

Step 2. Synthesis of benzyl ( E)-4-methy lpent-2-enoate

To two batches of a solution of (E)-4-methylpent-2-enoic acid (6.25 mL, 52.6 mmol) in acetone (90 mL) was added K2CO3 (13.8 g, 100 mmol) and the mixtures were stirred for 30 min. Then a solution of benzyl bromide (6.31 mL, 53.1 mmol) in acetone (10 mL) was added and the mixtures were heated to 75 °C for 5 h. The reaction mixtures were cooled to room temperature and concentrated under reduced pressure. The residue was dissolved in EtOAc (200 mL) and HzO (200 mL) then extracted into EtOAc (2 x 200 mL). The combined organic layers were washed with brine (300 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by silica gel chromatography (0-→10% EtO Ac/pet. ether) afforded product (9.0 g, 42% yield).

Step 3: Synthesis of benzyl (2fl,3S)-2,3-dihydroxy-4-methylpentanoate

To a solution of AD-mix-a (61.7 g) and methanesulfonamide (4.19 g, 44.1 mmol) in tert-BuOH (225 mL) and H 2 O (225 mL) was added benzyl (£)-4-methylpent-2-enoate (9 g, 44.1 mmol). The mixture was stirred at room temperature for 12 h then Na2SOs (67.5 g) was added and stirred for 30 min. The reaction mixture was diluted with EtOAc (300 mL) and H 2 O (300 mL) and extracted into EtOAc (3 x 300 mL), washed with brine (300 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by silica gel chromatography (0→25% EtO Ac/pet. ether) afforded product (8.3 g, 79% yield). LCMS (ESI) m/z. [M + Na] ealed for C13H18O4: 261.11 ; found 261.0.

Step 4: Synthesis of benzyl (4/?,5S)-5-isopropyl-1 ,3,2-dioxathiolane-4-carboxylate 2-oxide To a solution of benzyl (2/?,3S)-2,3-dihydroxy-4-methylpentanoate (10 g, 42.0 mmol) in DCM (100 mL) at 0 °C was added EtsN (17.5 mL, 126 mmol) and SOCI2 (4.26 mL, 58.8 mmol). The reaction mixture was stirred 30 min then was diluted with DCM (30 mL) and H 2 O (100 mL), extracted into DCM (3 x 50 mL), washed with brine (100 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure which afforded product (11.0 g, 92% yield). Step 5: Synthesis of benzyl (4/?,5S)-5-isopropyl-1 ,3,2-dioxathiolane-4-carboxylate 2,2-dioxide To a solution of benzyl (4/?,5S)-5-isopropyl-1 ,3,2-dioxathiolane-4-carboxylate 2-oxide (11 g, 38.7 mmol) in H 2 O (250 mL), MeCN (125 mL), and CCU (125 mL) was added Nal04 (3.22 mL, 58.0 mmol) and RuCl3*H20 (872 mg, 3.87 mmol). The mixture was stirred at room temperature for 1 h then was diluted with EtOAc (200 mL) and H 2 O (50 mL), filtered, and the filtrate was extracted into EtOAc (3 x 200 mL). The combined organic layers were washed sequentially with brine (200 mL) and sat. aq. Na 2 CO 3 (300 mL), dried over Na 2 SO 4 . filtered, and concentrated under reduced pressure. Purification by silica gel chromatography (0-→17% EtO Ac/pet. ether) afforded product (11 g, 95% yield).

Step 6: Synthesis of benzyl (2S,3S)-2-bromo-3-hydroxy-4-methylpentanoate To a solution of benzyl (4/?,5S)-5-isopropyl-1 ,3,2-dioxathiolane-4-carboxylate 2,2-dioxide (11 g, 36.6 mmol) in THF (520 mL) was added LiBr (3.49 mL, 139 mmol). The reaction mixture was stirred at room temperature for 5 h and then concentrated under reduced pressure. The residue was diluted in THF (130 mL) and H 2 O (65 mL), cooled to 0 °C, then HaS04 solution (20% aq., 1.3 L) was added and the mixture was warmed to room temperature and stirred for 24 h. The mixture was diluted with EtOAc (1.0 L), extracted into EtOAc (2 x 300 mL), washed sequentially with Na 2 CO 3 (sat. aq., 300 mL) and brine (300 mL), then was concentrated under reduced pressure. Purification by silica gel chromatography (0→17% EtOAc/pet. ether) afforded product (10 g, 81% yield).

Step 7: Synthesis of benzyl (2fl,3S)-2-azido-3-hydroxy-4-methylpentanoate To a solution of benzyl (2S,3S)-2-bromo-3-hydroxy-4-methylpentanoate (10 g, 33.2 mmol) in DMSO (100 mL) was added NaNa (4.32 g, 66.4 mmol). The reaction mixture was stirred at room temperature for 12 h then was diluted with EtOAc (300 mL) and H 2 O (200 mL). The aqueous phase was extracted into EtOAc (2 x 200 mL), washed with brine (200 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by silica gel chromatography (0→17% EtOAc/pet. ether) afforded product (7.5 g, 79% yield).

Step S. Synthesis of benzyl (2fl,3fl)-3-isopropylaziridine-2-carboxylate To a solution of benzyl (2/?,3S)-2-azido-3-hydroxy-4-methylpentanoate (7.5 g, 28.5 mmol) in MeCN (150 mL) was added PPha (7.70 g, 29.3 mmol). The reaction mixture was stirred at room temperature for 1 h and then heated to 70 °C and stirred for 4 h. The reaction mixture was concentrated under reduced pressure and purification by silica gel chromatography (0→17% EtOAc/pet. ether) afforded product (4.5 g, 66% yield). LCMS (ESI) m/z. [M + H] calcd for C13H17NO2: 220.13; found 220.0.

Step 9: Synthesis of benzyl (2ff,3fl)-3-isopropyl-1-tritylaziridine-2-carboxylate To a solution of benzyl (2f?,3/?)-3-isopropylaziridine-2-carboxylate (2 g, 9.12 mmol) in DCM (30 mL) at 0 °C was added EtsN (3.81 mL, 27.4 mmol) and trityl chloride (3.05 g, 10.9 mmol) followed by DMAP (111 mg, 912 pmol). The reaction mixture was stirred at 0 °C for 1 h and then was diluted with DCM (50 mL) and H 2 O (50 mL) then extracted into DCM (2 x 30 mL). The combined organic layers were washed with brine (50 mL), dried over Na2SC>4, filtered, and concentrated under reduced pressure. Purification by silica gel chromatography (0→25% DCM/pet. ether) afforded product (3.1 g, 72% yield). Step 10·. Synthesis of (2/?,3fl)-3-isopropyl-1-tritylaziridine-2-carboxylic acid Two solutions of benzyl (2/?,3fl)-3-isopropyl-1-tritylaziridine-2-carboxylate (200 mg, 430 pmol) and Pd/C (100 mg) in THF (4 mL) were stirred for 1 h at room temperature under Ha atmosphere. The reaction mixtures were combined, filtered, and concentrated under reduced pressure. Purification by silica gel chromatography (0→50% EtOAc/pet. ether) afforded product (160 mg, 51% yield). Intermediate A-97. Synthesis of (2S,3S)-1-benzyl-3-isopropylaziridine-2-carboxylic acid

Step 1: Synthesis of benzyl (2S,3fl)-2,3-dihydroxy-4-methylpentanoate To a solution of AD-mix-B (61.7 g) and methanesulfonamide (4.19 g, 44.1 mmol) in tert-BuOH (225 mL) and H 2 O (225 mL) was added benzyl (£)-4-methylpent-2-enoate (9 g, 44.1 mmol). The mixture was stirred at room temperature for 12 h then NaaSOs (67.5 g) was added and stirred for 30 min. The reaction mixture was diluted with EtOAc (300 mL) and H 2 O (300 mL) and extracted into EtOAc (3 x 300 mL), washed with brine (300 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by silica gel chromatography (0→25% EtO Ac/pet. ether) afforded product (8.8 g, 84% yield). LCMS (ESI) m/z. [M + Na] calcd for CiaHieO*: 261.11 ; found 261.0.

Step 2: Synthesis of benzyl (4S,5fl)-5-isopropyl-1 ,3,2-dioxathiolane-4-carboxylate 2-oxide To a solution of benzyl (2S,3fl)-2,3-dihydroxy-4-methylpentanoate (11.6 g, 48.7 mmol) in DCM (116 mL) at 0 °C was added EtaN (20.3 mL, 146 mmol) and SOCIa (4.94 mL, 68.2 mmol). The reaction mixture was stirred 30 min then was diluted with DCM (100 mL) and H 2 O (100 mL), extracted into DCM (3 x 100 mL), washed with brine (200 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure which afforded product (13.0 g, 94% yield).

Step 3: Synthesis of benzyl (4S,5fl)-5-isopropyl-1 ,3,2-dioxathiolane-4-carboxylate 2, 2-dioxide To a solution of benzyl (4S,5fl)-5-isopropyl-1 ,3,2-dioxathiolane-4-carboxylate 2-oxide (13 g, 45.7 mmol) in H 2 O (290 mL), MeCN (145 mL), and CCL (145 mL) was added NalO< (3.80 mL, 68.6 mmol) and RuCb-H 2 O (1.03 g, 4.57 mmol). The mixture was stirred at room temperature for 1 h then was diluted with DCM (500 mL) and H 2 O (300 mL), filtered, and the filtrate was extracted into DCM (3 x 200 mL). The combined organic layers were washed sequentially with brine (500 mL) and sat. aq. NaaCOa (300 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by silica gel chromatography (0→17% EtO Ac/pet. ether) afforded product (11.5 g, 80% yield).

Step 4: Synthesis of benzyl (2/?,3fl)-2-bromo-3-hydroxy-4-methylpentanoate To a solution of benzyl (4S,5fl)-5-isopropyl-1 ,3,2-dioxathiolane-4-carboxylate 2,2-dioxide (11.5 g, 38.3 mmol) in THF (520 mL) was added LiBr (3.65 mL, 146 mmol). The reaction mixture was stirred at room temperature for 5 h and then concentrated under reduced pressure. The residue was diluted in THF (130 mL) and H 2 O (65 mL), cooled to 0 °C, then H2SO4 solution (20% aq., 1.3 L) was added and the mixture was warmed to room temperature and stirred for 24 h. The mixture was diluted with EtOAc (1.0 L), washed with Na 2 CO 3 (sat. aq., 300 mL), then was concentrated under reduced pressure. Purification by silica gel chromatography (0→17% EtOAc/pet. ether) afforded product (10 g, 83% yield).

Step 5: Synthesis of benzyl (2S,3fl)-2-azido-3-hydroxy-4-methylpentanoate To a solution of benzyl (2/7,3fl)-2-bromo-3-hydroxy-4-methylpentanoate (10 g, 33.2 mmol) in DMSO (100 mL) was added NaNs (4.33 g, 66.6 mmol). The reaction mixture was stirred at room temperature for 12 h then was diluted with EtO Ac (300 mL) and H 2 O (200 mL). The mixture was extracted into EtO Ac (2 x 200 mL), dried over NaaSOe, filtered, and concentrated under reduced pressure. Purification by silica gel chromatography (0→17% EtO Ac/pet. ether) afforded product (7.5 g, 76% yield). Step 6: Synthesis of benzyl (2S,3S)-3-isopropylaziridine-2-carboxylate To a solution of benzyl (2S,3fl)-2-azido-3-hydroxy-4-methylpentanoate (7.5 g, 28.5 mmol) in MeCN (150 mL) was added PPhs (7.70 g, 29.3 mmol). The reaction mixture was stirred at room temperature for 1 h and then heated to 70 °C and stirred for 3 h. The reaction mixture was concentrated under reduced pressure and purification by silica gel chromatography (0→17% EtO Ac/pet. ether) afforded product (4.5 g, 64% yield). LCMS (ESI) m/z : [M + H] calcd for C13H17NO2: 220.13; found 220.1.

Step 7: Synthesis of benzyl (2S,3S)-1 -benzyl-3-isopropylaziridine-2-carboxylate To a solution of benzyl (2S,3S)-3-isopropylaziridine-2-carboxylate (1 g, 4.56 mmol) in MeCN (10 mL) was added K2CO3 (3.15 g, 22.8 mmol) and benzyl bromide (812 pL, 6.84 mmol). The reaction mixture was stirred at room temperature for 6 h then was diluted with EtO Ac (30 mL) and H 2 O (30 mL), extracted into EtO Ac (2 x 30 mL), washed with brine (50 mL), dried over Na 2 SO 4 . filtered, and concentrated under reduced pressure. Purification by silica gel chromatography (0-→17% EtO Ac/pet. ether) afforded product (1.3 g, 89% yield). LCMS (ESI) m/z. [M + H] calcd for C20H23NO2: 310.18; found 310.1.

Step 8: Synthesis of (2S,3S)-1-benzyl-3-isopropylaziridine-2-carboxylic acid To a solution of benzyl (2S,3S)-1-benzyl-3-isopropylaziridine-2-carboxylate (600 mg, 1.94 mmol) in THF (6 mL), MeCN (3 mL), and HzO (6 mL) at 0°C was added LiOH'H 2 O (163 mg, 3.88 mmol). The reaction mixture was stirred at room temperature for 1 h and was adjusted to pH = 7-8 with HCI (0.5M). Lyophilization afforded product (750 mg, crude). LCMS (ESI) m/z. [M + H] calcd for C13H17NO2: 220.13; found 220.1.

Intermediate A-98, A-99, A- 100, and A-101. Synthesis of ethyl (2/?,3fl)-1 -benzhydryl-3- (oxetan-3-yl)azirldlne-2-carboxylate, ethyl (2S,3S)-1 -benzhydryl-3-(oxetan-3-yl)aziridine-2- carboxylate, ethyl (2/?,3S)-1-benzhydryl-3-(oxetan-3-yl)azlrldlne-2-cart>oxy late, and ethyl (2S,3R)-1- benzhydryl-3-(oxetan-3-yl)azirldine-2-carboxylate

Step 1: Synthesis of N-benzhydryl-1-(oxetan-3-yl)methanimine

To a solution oxetane-3-carbaldehyde (5.0 g, 58 mmol) and MgSO* (6.99 g, 58.1 mmol) in DCM (120 mL) at 0 °C was added diphenylmethanamine (12.1 mL, 69.7 mmol). The mixture was stirred for 12 h at room temperature then filtered and concentrated under reduced pressure to afford the desired compound (14 g, 95.9% yield) which was used without further purification.

Step 2. Synthesis of ethyl c/s-1-benzhydryl-3-(oxetan-3-yl)aziridine-2-carboxylate and ethyl trans- 1-benzhydryl-3-(oxetan-3-yl)aziridine-2-carboxylate

To a solution of W-benzhydryl-1-(oxetan-3-yl)methanimine (10 g, 39.79 mmol) in MeCN (150 mL) was added TfOH (878 mL, 9.95 mmol) and after 5 min ethyl diazoacetate (5.0 mL, 47.8 mmol) was added. The reaction mixture was stirred for 12 h at room temperature then cooled to 0 °C and quenched by the addition of saturated NaHCOs (300 mL). The aqueous layer was extracted with EtOAc (3 x 200 mL) and the combined organic layers were washed with brine, dried with NaaSOA, filtered, and concentrated under reduced pressure. Purification by reverse phase chromatography (50→65% MeCN/HzO, 10 mM NH4HCO3) afforded racemic ethyl c/s-1-benzhydryl-3-(oxetan-3-yl)aziridine-2- carboxylate (1.1 g, 8.2% yield) and racemic ethyl frans-1-benzhydryl-3-(oxetan-3-yl)aziridine-2- carboxylate (780 mg, 5.8% yield)

Step 3: Separation of racemic ethyl c/s-1-benzhydryl-3-(oxetan-3-yl)aziridine-2-carboxylate: ethyl (2f?,3fl)-1 -benzhydryl-3-(oxetan-3-yl)aziridine-2-carboxylate and ethyl (2S,3S)-1 -benzhydryl-3-(oxetan-3- yl)aziridine-2-carboxylate

Racemic ethyl c/s-1-benzhydryl-3-(oxetan-3-yl)aziridine-2-carboxylate (800 mg, 2.37 mmol) was separated by chiral prep-SFC (25% MeOH/COa) to afford ethyl (2fl,3fl)-1-benzhydryl-3-(oxetan-3-yl) aziridine-2-carboxylate (320 mg, 40% yield) and ethyl (2S,3S)-1-benzhydryl-3-(oxetan-3-yl)aziridine-2- carboxylate (320 mg, 40% yield).

Step 4: Separation of racemic ethyl frans-1 -benzhydryl-3-(oxetan-3-yl)aziridine-2-carboxylate: ethyl (2R,3S)-1-benzhydryl-3-(oxetan-3-yl)aziridine-2-carboxylate and ethyl (2S,3fl)-1 -benzhydryl-3- (oxetan-3-yl)aziridine-2-carboxylate

Racemic ethyl trans1-benzhydryl-3-(oxetan-3-yl)aziridine-2-carboxylate (700 mg, 2.07 mmol) was separated by chiral prep-SFC (25% EtOH, 0.1% NH4OH/CO2) to afford ethyl (2f?,3S)-1 -benzhydryl-3- (oxetan-3-yl) aziridine-2-carboxylate (300 mg, 42% yield) and ethyl (2S,3fl)-1 -benzhydryl-3-(oxetan-3- yl)aziridine-2-carboxylate (320 mg, 43% yield).

Intermediate A-102 and A-103. Synthesis of (2/?,3/?)-1"Ι>βηζΙιγά^3-(οχβΐ8η-3-γΙ)8ζί ΓΜίηβ- 2-carboxylic acid and (2S,3S)-1-benzhydryl-3-(oxetan-3-yl)aziridine-2-cait>oxyl ic acid

Step 1: Synthesis of (2fl,3fl)-1-benzhydryl-3-(oxetan-3-yl)aziridine-2-carboxylic acid To a solution of ethyl (2fl,3fl)-1-benzhydryl-3-(oxetan-3-yl)aziridine-2-carboxylat e (156 mg, 463 mmol) in EtOH (3 mL) was added 2M NaOH (347 mL, 696 mmol). The reaction mixture was stirred for 3 h at room temperature and then concentrated under reduced pressure. The concentrate was acidified to pH 5 with 1 M HCI and extracted with DCM (3 x 5 mL) and the combined organic layers were washed with brine, dried with NaaSOA, filtered and concentrated under reduced pressure to afford the desired compound (110 mg, 72.6% yield).

Step 2: Synthesis of (2S,3S)-1 -benzhydryl-3-(oxetan-3-yl)aziridine-2-carboxylic acid To a solution of ethyl (2S,3S)-1-benzhydryl-3-(oxetan-3-yl)aziridine-2-carboxylate (150 mg, 444 mmol) in EtOH (5 mL) was added 2M NaOH (333 mL, 666 mmol). The reaction mixture was stirred for 3 h at room temperature and then acidified to pH 5 with 1 M HCI. The aqueous layer extracted with DCM (3 x 10 mL) and the combined organic layers were washed with brine, dried with NagSO*. filtered, and concentrated under reduced pressure to afford the desired compound (120 mg, 86.1% yield).

Intermediate A-104 and A-105. Synthesis of sodium ^A^SH-benzhydryl-S-Coxetan-S- yl)azlrldlne-2-carboxylate and sodium (2S,3fl)-1-benzhydryl-3-(oxetan-3-yl)azirldine-2-carboxylate

Step 1: Synthesis of sodium (2fl,3S)-1-benzhydryl-3-(oxetan-3-yl)aziridine-2-carboxylate To a solution of ethyl (2f?,3S)-1-benzhydryl-3-(oxetan-3-yl)aziridine-2-carboxylate (150 mg, 444 mmol) in EtOH (3 mL) was added 2M NaOH (333.42 mL, 666 mmol). The reaction mixture was stirred for 3 h at room temperature and then the pH was adjusted to pH 8 with 1 M HCI. The resulting solution was lyophilized to afford the desired compound (165 mg, crude) which was used without further purification. LCMS (ESI) mfr. [M] calcd for CigHieNOa: 308.13; found 308.0.

Step 2. Synthesis of sodium (2S,3fl)-1-benzhydryl-3-(oxetan-3-yl)aziridine-2-carboxylate To a solution of ethyl (2S,3fl)-1-benzhydryl-3-(oxetan-3-yl)aziridine-2-carboxylate (170 mg, 503 mmol) in EtOH (3 mL) was added 2M NaOH (378 mL, 754 mmol). The reaction mixture was stirred for 3 h at room temperature and then the pH was adjusted to pH 8 with 1 M HCI. The resulting solution was lyophilized to afford the desired compound (230 mg, crude) which was used without further purification. LCMS (ESI) m/z·. [M] calcd for CigHieNOa: 308.13; found 308.0.

Intermediate A- 106. Synthesis of (fl)-1-((benzyloxy)carbonyl)-2-methylazlrldlne-2- carboxylic acid Step 1: Synthesis of benzyl (2S,4S)-4-methyl-5-oxo-2-phenyloxazolidine-3-carboxylate To a mixture of ((benzyloxy)carbonyl)-L-alanine (25 g, 111.99 mmol) and (dimethoxymethyl)benzene (71.38 mL, 115.35 mmol) in THF (180 mL) was added SOCb (8.94 g, 123.19 mmol) in one portion at 0 °C. The mixture was stirred for 10 min before ZnCfe (5.77 mL, 123.26 mmol) was added to the solution, then the mixture was stirred at 0 °C for 4 h. The reaction mixture was quenched by dropwise addition of cold H 2 O and adjusted to pH = 5 with sat. NaHCOs, then extracted with EtOAc (2 x 100 mL). The organic phase was washed with a aq. sat. NaHCOs (30 mL) and brine (30 mL), dried over Na 2 SO 4 , and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (1→10% EtOAc/pet. ether) to afford product (15 g, 43% yield).

Step 2. Synthesis of benzyl (2S,4S)-4-(iodomethyl)-4-methyl-5-oxo-2-phenyloxazolidine-3- carboxylate

HMPA (5.22 mL, 29.74 mmol) and LHMDS (1 M, 6.62 mL) were mixed in THF (45 mL) under Na atmosphere at 20 °C. This solution was cooled to -78 °C and a solution of benzyl (2S,4S)-4-methyl-5- oxo-2-phenyloxazolidine-3-carboxylate (2.0 g, 6.42 mmol) in THF (12 mL) was added dropwise with stirring. After stirring an additional 30 min, a solution of CH2I2 (1 .55 mL, 19.27 mmol) in THF (6 mL) was added dropwise. The mixture was stirred at -78 °C for 90 min. The mixture was warmed to 0 °C and quenched with sat. aq. NH4CI (70 mL). The mixture was extracted with EtOAc (2 x 30 mL), and the combined organic layers was washed with sat. aq. NH4CI (20 mL), H 2 O (2 x 20 mL), and brine (30 mL) dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (1→20% EtOAc/pet. ether) to afford product (1.2 g, 41.4% yield). Step 3: Synthesis of methyl (S)-2-(((benzyloxy)carbonyl)amino)-3-iodo-2-methylpropanoate To a mixture of benzyl (2S,4S)-4-(iodomethyl)-4-methyl-5-oxo-2-phenyloxazolidine-3- carboxylate (1.2 g, 2.66 mmol) in THF (20 mL) was added a solution of NaOMe (957.69 mg, 5.32 mmol, 30% purity) in MeOH (9 mL) dropwise over 10 min at -40 °C under N 2 . The mixture was stirred at -40 °C for 2 h, then warmed to -20 °C and stirred for 1 h. The reaction was quenched by addition of H 2 O (20 mL), and the resulting mixture was extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (20 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (1 →20% EtOAc/pet. ether) to afford product (870 mg, 2.24 mmol, 84.4% yield).

Step 4: Synthesis of 1 -benzyl 2-methyl (fl)-2-methylaziridine-1 ,2-dicarboxylate To a mixture of methyl (S)-2-(((benzyloxy)carbonyl)amino)-3-iodo-2-methylpropanoate (0.87 g, 2.31 mmol) in MeCN (125 mL) was added AgaO (1.60 g, 6.92 mmol) in one portion at room temperature. The mixture was stirred at 90 °C for 30 min. The mixture was filtered and concentrated under reduced pressure to afford product (500 mg, 2.01 mmol, 86.9% yield).

Step 5: Synthesis of 1 -benzyl 2-methyl (fl)-2-methylaziridine-1 ,2-dicarboxylate To a mixture of 1 -benzyl 2-methyl ( fl)-2-methylazi ridi ne-1 ,2-dicarboxylate (250 mg, 1.0 mmol) in MeCN (2.5 mL) and H 2 O (2.5 mL) was added NaOH (40.12 mg, 1.0 mmol) in one portion at 0 °C under Na. The mixture was stirred at 0 °C for 30 min. The mixture was concentrated under reduced pressure to afford crude product (256 mg, crude). LCMS (ESI) m/z. [M + H] calcd for C12H12NO4: 234.1 ; found 234.1. Intermediate A-107. Synthesis of (S)-1-((benzyloxy)carbonyl)-2-methylaziridine-2- carboxylic acid

Step 1: Synthesis of benzyl (2fl,4fl)-4-methyl-5-oxo-2-phenyloxazolidine-3-carboxylate Five batches were completed in parallel. To a mixture of ((benzyloxy)carbonyl)-D-alanine (5 g, 22.40 mmol) and (dimethoxymethyl)benzene (3.71 mL, 24.64 mmol) in THF (35 mL) was added SOCI2 (1.79 mL, 24.64 mmol) in one portion at 0 °C. After the mixture was stirred for 10 min, ZnCl 2 (1.15 mL, 24.64 mmol) was added to the solution. Then the mixture was stirred at 0 °C for 4 h. The give batches were combined and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (1 →10% EtO Ac/pet. ether) to afford product (20 g, 57.4% yield).

Step 2. Synthesis of benzyl (2f?,4fl)-4-(iodomethyl)-4-methyl-5-oxo-2-phenyloxazolidine- 3- carboxylate

Four batches were completed in parallel. THF (300 mL), HMPA (13.06 mL, 74.34 mmol) and LHMDS (1 M, 16.54 mL) were mixed under N 2 atmosphere at 20 °C with stirring. The solution was cooled to -78 °C and a solution of benzyl (2/?,4fl)-4-methyl-5-oxo-2-phenyloxazolidine-3-carboxylate (5 g, 16.06 mmol) in THF (84 mL) was added dropwise. After stirring an additional 30 min, a solution of CH2I2 (3.89 mL, 48.18 mmol) in THF (33 mL) was added dropwise. The mixture was stirred at -78 °C for 90 min. The four batches were combined and warmed to 0 °C. Sat. aq. NH4CI (100 mL) was added to the combined solution and the resulting mixture was extracted with EtOAc (2 x 100 mL). The combined EtOAc layers was washed with sat. aq. NH4CI (50 mL), H 2 O (2 x 20 mL), and brine (30 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (1 →17% EtO Ac/pet. ether) to afford product (16 g, 55.2% yield).

Step 3: Synthesis of methyl (fl)-2-(((benzyloxy)carbonyl)amino)-3-iodo-2-methylpropanoat e To a mixture of benzyl (2/?,4fl)-4-(iodomethyl)-4-methyl-5-oxo-2-phenyloxazolidine- 3-carboxylate (16 g, 35.46 mmol) in THF (90 mL) was added NaOMe (12.77 g, 70.91 mmol, 30% purity) dropwise over

10 min at -40 °C under N 2 . The mixture was stirred at -40 °C for 2 h, then warmed to -20 °C and stirred for 1 h. The reaction was quenched by addition of H 2 O (100 mL), and the resulting mixture was extracted with diethyl ether (3x100 mL). The combined organic layers were washed with brine (50 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (1 →17% EtO Ac/pet. ether) to afford product (10 g, 74.8% yield.

Step 4: Synthesis of 1 -benzyl 2-methyl (S)-2-methylaziridine-1 ,2-dicarboxylate Four batches were completed in parallel. To a mixture of methyl (fl)-2- (((benzyloxy)carbonyl)amino)-3-iodo-2-methylpropanoate (8 g, 21.20 mmol) in MeCN (800 mL) was added AgaO (14.76 g, 63.64 mmol) in one portion at 20 °C. The mixture was stirred at 90 °C for 30 min. The four batches were combined, filtered, and concentrated under reduced pressure to afford product (5.1 g, 90.9% yield. Step 5: Synthesis of (S)-1-((benzyloxy)cart)onyl)-2-methylaziridine-2-carboxylic acid To a solution of 1 -benzyl 2-methyl (S)-2-methylaziridine-1 ,2-dicarboxylate (1 g, 4.01 mmol) in MeCN (5 mL) was added a solution of NaOH (240.69 mg, 6.02 mmol) in l½0 (5 mL) at 0 °C, then the mixture was stirred at 0 °C for 30 min. The mixture was lyophilized directly to afford crude product (1.05 g, crude). LCMS (ESI) m/z. [M + H] calcd for C12H12NO4: 234.08; found 234.2.

Intermediates A- 108 and A-109. Synthesis of tert-butyl (fl)-7-((S)-2-(benzyloxy)-1- cyck>pentyl-2-oxoethyl)-6-oxo-2,7-diazaspiro[4.4]nonane-2 -carboxylate and tert-butyl (S)-7-((S)-2- (benzyloxy)-1-cyclopentyl-2-oxoethyl)-6-oxo-2,7-dlazasplro[4 .4]nonane-2-carboxylate

Step 1: Synthesis of 1 -(tert-butyl) 3-methyl 3-allylpyrrolidine-1 ,3-dicarboxylate To a solution of 1 -(tert-butyl) 3-methyl pyrrolidine-1 ,3-dicarboxylate (10.0 g, 43.6 mmol) in THF (100 mL) at -78 °C was added UHMDS (65.0 mL, 65.4 mmol, 1 M in THF). After 1 h allyl bromide (5.63 mL, 65.4 mmol) was added and the resulting mixture was warmed to room temperature overnight. The reaction was quenched at 0 °C by the addition of NH4CI (200 mL). The aqueous layer was extracted with EtOAc (3 x 100 mL) and the combined organic layers were washed with brine, dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by normal phase chromatography (5% EtOAc/pet. ether) afforded the desired product (10.0 g, 76.6% yield).

Step 3: Synthesis of 1 -(tert-butyl) 3-methyl 3-(2-oxoethyl)pyrrolidine-1 ,3-dicarboxylate To a solution of 1 -(tert-butyl) 3-methyl 3-allylpyrrolidine-1 ,3-dicarboxylate (10.0 g, 37.1 mmol) and 2,6-dimethylpyridine (8.65 mL, 80.7 mmol) in dioxane (571 mL) and H 2 O (142 mL) at 0 °C was added K20s04 e 2H20 (0.27 g, 0.73 mmol). After 15 min Nal04 (23.82 g, 111.4mmol) was added and the resulting mixture was stirred overnight at room temperature and then was diluted with H 2 O (200 mL). The aqueous layer extracted with EtOAc (3 x 200 mL) and the combined organic layers were washed with 2 M HCI, dried with Na 2 SO 4 , filtered and concentrated under reduced pressure to afford the desired product (9.7 g, crude) which was used without further purification.

Step 4: Synthesis of 1 -(tert-butyl) 3-methyl 3-(2-(((S)-2-(benzyloxy)-1-cyclopentyl-2- oxoethyl)amino)ethyl)pyrrolidine-1 ,3-dicarboxylate

To a solution of 1 -(tert-butyl) 3-methyl 3-(2-oxoethyl)pyrrolidine-1 ,3-dicarboxylate (9.60 g, 35.4 mmol) in MeOH (100 mL) at 0 °C was added benzyl (S)-2-amino-2-cyclopentylacetate

(12.38 g, 53.075 mmol) and zinc chloride (7.23 g, 53.1 mmol). After 30 min NaBHaCN (4.45 g, 70.8 mmol) was added and the resulting mixture stirred for 2 h at room temperature, concentrated under reduced pressure and the residue diluted with H 2 O (150 mL). The aqueous layer was extracted with EtOAc (3 x 50 mL) and the combined organic layers were washed with brine, dried with Na 2 SO 4 , filtered, and then concentrated under reduced pressure. Purification by normal phase chromatography (20% EtOAc/pet. ether) afforded the desired product (11.1 g, 64.2% yield). LCMS (ESI) m/z: [M + H] calcd for C27H40N 2 O6: 489.30; found 489.3.

Step 5: Synthesis of tert-butyl (fl)-7-((S)-2-(benzyloxy)-1 -cyclopentyl-2-oxoethyl)-6-oxo-2,7- diazaspiro[4.4]nonane-2-carboxylate and tert-butyl (S)-7-((S)-2-(benzyloxy)-1 -cydopentyl-2-oxoethyl)-6- oxo-2, 7-diazaspiro[4.4]nonane-2-carboxylate

To a solution of stirred solution of 1 -(tert-butyl) 3-methyl 3-(2-(((S)-2-(benzyloxy)-1-cyclopentyl-2- oxoethyl)amino)ethyl)pyrrolidine-1 ,3-dicarboxylate (11.1 g, 22.7 mmol) in toluene (120 mL) was added DIPEA (39.6 mL, 227 mmol) and DMAP (2.78 g, 22.7 mmol). The resulting mixture was stirred for 2 days at 80 °C and then concentrated under reduced pressure. Purification by reverse phase chromatography (20→70% MeCN/H 2 O, 0.1% HCOaH) afforded a mixture of desired products. The diastereomers were separated by prep-SFC (30% EtOH/COa) to afford tert-butyl (fl)-7-((S)-2-(benzyloxy)-1 -cyclopentyl-2- oxoethyl)-6-oxo-2,7-diazaspiro[4.4]nonane-2-carboxylate (3.73 g, 44.4% yield) LCMS (ESI) m/zr. [M + H] calcd for CaeHaeNaOs: 457.27; found 457.3 and tert-butyl (S)-7-((S)-2-(benzyloxy)-1 -cyclopentyl-2- oxoethyl)-6-oxo-2,7-diazaspiro[4.4]nonane-2-carboxylate (3.87 g, 46.1% yield) ) LCMS (ESI) m/z : [M + H] calcd for CaeHaeNaOs: 457.27; found 457.3.

Intermediate B-1. Synthesis of N-(3-(3-(4-methoxyphenyl)thloureldo)propanoyl)-A/-methyl·

L-valine

Step 1: Synthesis of methyl N-(3-((iert-butoxycarbonyl)amino)propanoyl)-N-methyl-L-valin ate To a solution of 3-((tert-butoxycarbonyl)amino)propanoic acid (1.04 g, 5.50 mmol) in DMF (6 mL) was added DIPEA (2.38 mL, 13.7 mmol) followed by HATU (2.71 g, 7.15 mmol). The reaction mixture was stirred for 5 min and methyl methyl-L-valinate hydrochloride (1 g, 5.50 mmol) was added. The reaction was stirred at room temperature for 3 h and was then quenched with H 2 O. The aqueous layer was extracted with EtOAc (3 x 10 mL) and the combined organic layers were washed with brine, and dried over Na 2 SO 4 . filtered, and concentrated under reduced pressure to afford the desired crude product. Step 2. Synthesis of methyl N-(3-aminopropanoyl)-N-methyl-L-valinate trifluoroacetic acid To a solution of methyl N-(3-((tert-butoxycarbonyl)amino)propanoyl)-N-methyl-L-valin ate (1.74 g, 5.50 mmol) in DCM (3 mL) was added TFA (2.09 mL, 27.4 mmol). The reaction was stirred at room temperature overnight and was then concentrated under reduced pressure to afford a solution of the desired crude product as a 33.5% solution in TFA.

Step 3: Synthesis of methyl W-(3-(3-(4-methoxyphenyl)thioureido)propanoyl)-N-methyl-Z.-v alinate To a 33.5 wt% solution of methyl N-(3-aminopropanoyl)-N-methyl-L-valinate trifluoroacetic acid (800 mg, 0.811 mmol) in TFA was added DCM (5 mL) followed by EtsN (593 pL, 4.26 mmol) and 4- methoxyphenyl isothiocyanate (117.0 pL, 852 pmol). The reaction was stirred at room temperature for 3 h. The reaction mixture was then washed with H 2 O (2 x 5 mL), aq. NH4CI (5 mL), and brine (5 mL). The organic layer was dried over Na 2 SO 4 and concentrated under reduced pressure to afford the crude product (290.2 mg 89.2% yield) as an oil, which was taken on without purification. LCMS (ESI) m/z:

[M + H] calcd for Cieh^l^S: 382.18; found 382.2.

Step 4: Synthesis of N-(3-(3-(4-methoxyphenyl)thioureido)propanoyl)-W-methyl-L-va line To a solution of methyl N-(3-(3-(4-methoxyphenyl)thioureido)propanoyl)-W-methyl-L-va linate (290.2 mg, 0.76 mmol) in THF (1 mL) was added a solution of LiOHehfeO (41.4 mg, 0.99 mmol) in H 2 O (300 pL). The reaction mixture was stirred overnight and was then acidified with HCI (4 M in dioxane,

120 pL, 0.48 mmol). The solution was then concentrated, the residue was dissolved in EtOAc, and the organic layer washed with H 2 O (3 x 5 mL) and brine (5 mL). The organic layer was dried over Na 2 SO 4 and concentrated under reduced pressure to afford the crude product (215.1 mg 77.0% yield), which was taken forward without further purification. LCMS (ESI) m/z: [M + H] calcd for C17H25N3O4S: 368.16; found 368.2.

The following table of compounds were prepared using the methods or variations thereof used to synthesize Intermediate B-1.

Table 3: Intermediate B

Molecular

Structure Formula Calculated MW Observed MW ft I H H

¾τγ α C17H25N3O4S [M + H] = 368.16 [M + H] = 368.5

OMe

Intermediate B-1 ft I H H

HO Αγ· Ν γ-χ/ Ν γ Ν X ) C18H27N3O3S [M + H] = 366.19 [M + H] = 366.6

A o s

Intermediate B-2

Intermediate B-3

OMe

S I H

Ηο Α γ Ν γ^ γ rcr C19H29N3O5S [M + H] = 412.19 [M + H] = 412.6 A ° s

Intermediate B-4

OMe

I H H \

HO N ^ YA C15H27N3O4S [M + H] = 346.18 [M + H] = 346.6

Intermediate B-5

O

H H

N

HO 396.20 [M + H] = 396.6

Intermediate B-6 Molecular

Structure Formula Calculated MW Observed MW

C19H27N3O4S [M + H] = 394.18 [M + H] = 394.7

OMe

Intermediate B-7

.0

Intermediate B-9

Intermediate B-10

OMe

HO C18H27N3O5S [M + H] = 398.17 [M + H] = 398.6

Intermediate B-11

Intermediate B-12

O I S

N ^s^OMe

HO [M + Na] =

C12H23N3O4S [M + Na] = 328.13

I S H H 328.5

Intermediate B-13

OMe

C16H23N3O4S [M + H] = 354.15 [M + H] = 354.5

Intermediate B-14

C14H27N3O4S [M + H] = 334.18 [M + H] = 334.6

Intermediate B-15

OMe

HO C1BH27N3O4S [M + H] = 382.18 [M + H] = 382.6

Intermediate B-16 Molecular

Structure Formula Calculated MW Observed MW

O

H H

N N. -N

HO 384.14 [M + H] = 384.3

O

Intermediate B-17 Molecular

Structure Formula Calculated MW Observed MW

?\ I H H

HO τ Ν Υ Χ/Ν Υ JO C17H25N3O3S [M + H] = 352.17 [M + H] = 352.4

A o s Ν

Intermediate B-27

MeO

S

O NH

C21H31 N3O4S [M + H] = 422.21 [M + H] = 422.7

N "NH

HO

O

Intermediate B-28

Intermediate B-30

HN

O

0- C14H27N3O3S [M + H] = 318.19 [M + H] = 318.7

HO ! ¾

Intermediate B-31

HO CieHayNaOaS [M + H] = 366.19 [M + H] = 366.7

Intermediate B-32

HO crvA OMe C14H18N 2 O3S [M + H] = 295.11 [M + H] = 295.6

1

Intermediate B-33

H H

^ N yN^ / ^ OMe

HO C9H16N 2 O3S [M + H] = 233.10 [M + H] = 233.4

Ύ

O

Intermediate B-34 Example 1. Synthesis of (3S)-1-((fl)-aziridine-2-carbonyl)-AF((2S)-1-(((6 3 S,4S)-1 1 -ethyF2 6 - hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-di oxo-6 1 ,6 2 ,6 3 ,6 4 ,6 e ,6 6 -hexahydro- 1 1 WJ-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyH - oxobutan-2-yl)-N-methylpyrrolidine-3-carboxamide k k k

Step 1 : Synthesis of (3S)-N- ((2S)-1-(((6 3 S,4S)-1 , -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin- 3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 ® -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)-N-methyl-1 -((R)- 1 -tritylaziridine- 2-carbonyl)pyrrolidine-3-carboxamide

To a solution of (6 3 S,4S)-4-amino-1 , -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10- dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione (520.0 mg, 0.831 mmol) and N-methyl-fV-((S)-1-((fl)-1- tritylaziridine-2-carbonyl)pyrrolidine-3-carbonyl)-L-valine (0.6727 g, 1.25 mmol) in DMF (10 mL) at 0 °C was added COMU (0.5338 mg, 1.25 mmol) followed by DIPEA (1.16 mL, 6.65 mmol). After 2 h, the reaction mixture was extracted with EtOAc (3 x 100 mL) and the combined organic layers were washed with brine (3 x 30 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The crude residue was purified by reverse phase chromatography (10→50% MeCN/H 2 O) to afford the desired product (500 mg, 52.4% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for CegHyeNsOs: 1147.60; found 1147.8.

Step 2. Synthesis of (3S)-1 -((fl)-aziridine-2-carbonyl)-N- ((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 - (4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)-N- methylpyrrolidine-3-carboxamide

To a stirred solution of (3S)-N-((2S)-1-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(4- (methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)-N- methyl-1 -((fl)-1-tritylaziridine-2-carbonyl)pyrrolidine-3-carboxamid e (145.0 mg, 0.126 mmol) in DCM (3 mL) at 0 °C was added EtsSiH (58.8 mg, 0.505 mmol) followed by TFA (57.6 mg, 0.505 mmol). After 1 h, DIPEA was added to the reaction mixture until pH 8. The resulting mixture was concentrated under reduced pressure, and the residue was purified by reverse phase chromatography (10-→50% MeCN/H 2 O) to afford the desired product (70 mg, 61.2% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for CsoHwNeOe: 905.49; found 905.7.

Example 7. Synthesis of (2fl)-W-(2-(((2S)-1-(((6 3 S,4S)-1 1 -ethyF2 5 -hydroxy-1 2 -(4- (methoxymethyl)pyrldln-3-yl)-10,10-dlmethyl-5,7-dloxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)-A/-methylaziridine-2-carboxami de

Step 1: Synthesis of (2fl)-N-(2-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl^-hydroxy-l 2 -(4-(methoxymethyl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl )(methyl)amino)-2-oxoethyl)-N-methyl- 1 -tritylaziridine-2-carboxamide

To a solution of (2S)-N-((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10- dimethyl-5, 7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-3-methyl-2-(N-methyl-2-(methyl amino)acetamido)butanamide (285.7 mg, 0.353 mmol) in DMF (3.0 mL) at 0 °C was added (fl)-1 -tritylaziridine-2-carboxylic acid (232.4 mg, 0.705 mmol) followed by DIPEA (0.61 mL, 4.7 mmol) and COMU (211.4 mg, 0.494 mmol).

The resulting mixture was warmed to room temperature and stirred for 1 h. The reaction mixture was diluted with H 2 O (15 mL) and the mixture was extracted with EtOAc (3 x 4 mL). The combined organic layers were washed with brine (10 mL), dried over NaaSOA, filtered, and concentrated under reduced pressure. The residue was purified by prep-TLC (12% EtOAc/pet. ether) to afford the desired product (301 mg, 68% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for CeyhbeNsOe: 1121.59; found 1121.8. Step 2. Synthesis of (2fl)-N-(2-(((2S)-1-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(4- (methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)-W-methylaziridine-2-carboxamid e

To a solution of (2fl)-/\A(2-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3- yl)-10,10-d imethyl-5 ,7-d ioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl )(methyl)amino)-2-oxoethyl)-N-methyl- 1 -tritylaziridine-2-carboxamide (301.0 mg, 0.268 mmol) in MeOH (3.0 mL) at 0 °C was added HCO2H (1.50 mL). The reaction mixture was stirred for 1 h and then neutralized to pH 8 with DIPEA. The resulting mixture was diluted with H 2 O (15 mL) and extracted with EtOAc (3 x 4 mL). The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by reverse phase chromatography (30→60% MeCN/H 2 O) to afford the desired product (89.9 mg, 38% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for C4eHeaNeOe: 879.48; found 879.7.

Example 15. Synthesis of two Isomers, 15A and 15B, of (2S)-AA((6 3 S,4S)-1 1 -ethyl-1 2 -(4- (methoxymethyl)pyridin-3-yl)-10,10-dlmethyl-5,7-dloxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)- lndola-6(1 ,3)-pyridazlna-2(1 ,3)-benzenacycloundecaphane-4-yl)-2-(3-((((4- methoxyphenyl)lmino)methylene)amino)-N- methylpropanamldo)-3-methylbutanamide

Step 1: Synthesis of (2S)-Λ/-((6 3 S,4S)-1 1 -ethyl·1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl- 5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 Af8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-2-(3-(3-(4-methoxyphenyl)thiou reido)-N-methylpropanamido)-3- methylbutanamide

To a solution of (6 3 S,4S)-4-amino-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl- 6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 e -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane- 5,7-dione (108 mg, 168 pmol) and N-(3-(3-(4-methoxyphenyl)thioureido)propanoyl)-N-methyl·L· valine (61 .9 mg, 168 pmol) in MeCN (2 mL) at 0 °C was added 2,6-lutidine (97.8 pL, 840 pmol) followed by COMU (78.8 mg, 184 pmol). After 1 h at 0 °C the reaction was diluted with EtOAc and the organic portion washed with H 2 O (15 mL) and brine (15 mL), dried over Na 2 SO 4 , and concentrated under reduced pressure. Purification by silica gel chromatography (20-+100% EtOAc/Hex then 0→5% MeOH/EtOAc) afforded the desired product (117.0 mg 72.6% yield). LCMS (ESI) m/z: [M + H] calcd for CMHWNBOS: 959.49; found 959.5.

Step 2. Synthesis of two isomers of (2S)-N-((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-2-(3-((((4-methoxyphenyl)imino )methylene)amino)-N- methylpropanamido)-3-methylbutanamide

To a solution of (2S)-N-((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7- dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-2-(3-(3-(4-methoxyphenyl)thiou reido)-N-methylpropanamido)-3- methylbutanamide (117.0 mg, 121 pmol) in DCM (1 mL) was added DIPEA (63.2 pL, 363 pmol) followed by 2-chloro-1-methylpyridin-1-ium iodide (42.6 mg, 181 pmol). The reaction mixture was stirred overnight, at which point the solid was filtered and the crude solution was purified by reverse phase chromatography (40→100 MeCN/H 2 O + 0.4% NH4OH) to afford two separated isomers as the desired earlier eluting isomer 15A (6.9 mg, 6.2% yield) and later eluting isomer 15B (2.5 mg, 2.2% yield). LCMS (ESI) m/z:

[M + H] calcd for C53H64N8O7: 925.50; found 925.5 and LCMS (ESI) m/z: [M + H] calcd for C53H64NBO7: 925.50; found 925.6. Example 25. Synthesis of (2S)-2-(3-(3-(2-chloroethyl)ureldo)-N-methylpropanamido)-/V ((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-di oxo- 6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H-B-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-3-methylbutanamide

Ο^,-ΟΝ ,N.

N

H

NH 2 °¾ Cl T

Ml «Yr lutidlne Ml s ITTV

I "OT1PS NtoCN mps l

N

<<

,o

TBAF Ml «χτΛ"

NtoCN

I

Step 1: Synthesis of (2S)-2-(3-(3-(2-chloroethyl)ureido)-A/-methylpropanamido)-W- ((6 3 S,4S)-1 1 - ethyl-1 2 -(2-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-di oxo-2 5 -((triisopropylsilyl)oxy)- 6 1 ,6 2 , 6 3 ,6 4 ,6 5 ,6 e -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane- 4-yl)-3-methylbutanamide

To a solution of (2S)-2-(3-amino-N-methylpropanamido)-N-((6 3 S,4S)-1 1 -ethyl-1 2 -(2- (methoxymethyOpyridin-S-yO-10.10-dimethyl-SJ-dioxo^-tttriiso propylsilyOoxyi-S'.e 2 ^ 3 ^ 4 ^ 5 ^- hexahydro-1 ' H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)-3- methylbutanamide (106 mg, 109 pmol) in MeCN (544 μΙ_) at 0 °C was added 1 -chloro-2-isocyanatoethane (9.29 pL, 109 pmol) followed by EtsN (15.1 pL, 109 pmol). After 12 min, the reaction was diluted with DCM (10 mL) and a solution of 1 % formic acid in H 2 O (10 mL). The aqueous layer was extracted with DCM (10 mL) and the combined organic layers were dried over NazSO*, filtered, and then concentrated under reduced pressure to afford the desired product (117 mg, 100% yield), which was used in the next step without purification. LCMS (ESI) m/z: [M + H] calcd for CsyHesCINeOeSi: 1071.59; found 1071.5.

Step 2. Synthesis of (2S)-2-(3-(3-(2-chloroethyl)ureido)-N-methylpropanamido)-N-( (6 3 S,4S)-1 ethyl-2 s -hydroxy-1 2 -(2-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-di oxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro- 1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)-3-methylbutanamide

To a solution of (2S)-2-(3-(3-(2-chloroethyl)ureido)-N- methylpropanamido)-N-((6 3 S,4S)-1 1 -ethyl- 1 2 -(2-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-di oxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6®- hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)-3- methylbutanamide (117 mg, 109 pmol) in MeCN (1.1 mL) at 0 °C was added TBAF (1 M in dioxane,

109 pL, 109 pmol). After 5 min, the reaction was concentrated under reduced pressure and the crude residue was purified by normal phase chromatography (20→100% B/A, B=10% MeOH/EtOAc, A=hexanes) followed by reverse phase chromatography (20→60% MeCN/H 2 O) to afford the final product (82.2 mg, 82% yield). LCMS (ESI) m/z: [M + H] calcd for CwHesCINeOe: 915.45; found 915.7.

Example 30. Synthesis of (2S)-2-(3-((4,5-dihydrooxazol-2-yl)amino)-W-methylpr<> panamido)- W-((6 3 S,4S)-1 1 -ethyl-2 ® -hydroxy-1 2 -(2-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-di oxo- 6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-3-methylbutanamide

A solution of (2S)-2-(3-(3-(2-chloroethyl)ureido)-N-methylpropanamido)-N-( (6 3 S,4S)-1 1 -ethyl-2 5 - hydroxy-1 2 -(2-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-di oxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8- oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)-3-methylbutanamide (55.0 mg, 60.0 pmol) and EtaN (25.1 pL, 180 pmol) in MeOH (1.2 mL) was heated in the microwave at 150 °C for 1 min. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The crude residue was then purified by reverse phase chromatography (30→100% MeCN/H 2 O + 0.4% NhUOH) to afford the final product (21.1 mg, 40% yield). LCMS (ESI) m/z: [M + H] calcd for

Ο^βΗκΝβΟβ: 879.48; found 879.4.

Example 31. Synthesis of (3S)-N- ((2S)-1-(((6 3 S,4S)-1 1 -ethyl-2 6 -hydroxy-1 2 -(4- (methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dloxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)- lndola-6(1 ,3)-pyridazlna-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)- A#-methyl-1-((S)-oxlrane-2-cait>onyl)pyrrolldlne-3-carbox amlde

To a solution of potassium (S)-oxirane-2-carboxylate (16.98 mg, 0.135 mmol), 2-chloro-1 ,3- dimethylimidazolidinium hexafl uorophosphate (87.46 mg, 0.314 mmol), and DIPEA (0.156 mL,

0.897 mmol) in DMF (1.5 mL) at 0 °C was added (3S)-N- ((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-2 ® -hydroxy-1 2 -(4- (methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)-W- methylpyrrolidine-3-carboxamide (75.0 mg, 0.09 mmol). The resulting mixture was stirred overnight at room temperature, at which point it was diluted with EtOAc (100 mL). The organic layer was washed with brine (3 x 5 mL), dried over NaaSCX, filtered, and concentrated under reduced pressure. Purification by reverse phase chromatography (25→55% MeCN/H 2 O) afforded the desired product (6.3 mg, 7.8% yield) as a solid. LCMS (ESI) m/z: [M + H] calcd for CsoHesNyOs: 906.48; found 906.7.

Example 34. Synthesis of (2fl)-1 -acetyl- W-(2-(((2S)-1-(((6 3 S, 4S)-1 1 -et hyP2 5 -hyd roxy-1 2 -(4- (methoxymethyl)pyridln-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)- lndola-6(1 ,3)-pyrldazlna-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)-N-methylazlridlne-2-carboxamld e o_Q

A 9-

TBAF

DCM ΙΠΡ8 men 4W k k

Step 1: Synthesis of (2fl)-N-(2-(((2S)-1-(((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-

10.10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)-N- methyl-1-tritylaziridine-2-carboxamide

To a solution of (6 3 S,4S)-4-amino-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-2 5 - ((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 e -hexahydro-1 ’H-8-oxa-l (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione (260 mg, 0.332 mmol) and A/-methyl-W-(N-methyl-N-((f?)-1- tritylaziridine-2-carbonyl)glycyl)-L-valine (204 mg, 0.399 mmol) in MeCN (3.3 mL) at 0 °C was added lutidine (192 μΙ_, 1.66 mmol) followed by COMU (156 mg, 0.366 mmol). The reaction stirred at 0 °C for 1 h and was then diluted with EtOAc. The mixture was washed with H20/brine (1 :1), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by normal phase chromatography (0→100% EtOAc/hexanes) afforded the desired product (116 mg, 27% yield). LCMS (ESI) m/z. [M + H] calcd for C/eHgeNeOeSi: 1277.72; found 1277.7.

Step 2. Synthesis of (2fl)-N-(2-(((2S)-1-(((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-

10.10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 8 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)-N- methylaziridine-2-carboxamide

To a solution of (2fl)-N-(2-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10, ID- dimethyl-5, 7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 ' H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan- 2-yl)(methyl)amino)-2- oxoethyl)-yV-methyl-1-tritylaziridine-2-carboxamide (400 mg, 0.313 mmol) in MeOH (1.56 mL) and chloroform (1.56 mL) at 0 °C was added TEA (191 pL, 2.50 mmol). The reaction stirred at 0 °C for 2 h and was then quenched with lutidine (364 pL, 3.13 mmol). The reaction mixture was diluted with DCM, washed with H 2 O, and concentrated under reduced pressure. Purification by reverse phase chromatography (10-→100% MeCN/H 2 O) afforded the desired product (100 mg, 31% yield). LCMS (ESI) m/z. [M + H] calcd for CsyHsaNeOeSi: 1035.61 ; found 1035.6. Step 3: Synthesis of (2fl)-1 -acetyl-N-(2-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin- 3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)-N- methylaziridine-2-carboxamide

To a solution of (2fl)-N-(2-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 e -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan- 2-yl)(methyl)amino)-2- oxoethyl)-N-methylaziridine-2-carboxamide (33 mg, 0.032 mmol) in DCM (637 pL) at 0 °C was added EtsN (22.1 pL, 0.159 mmol) followed by acetyl chloride (4.54 pL, 0.064 mmol). The reaction stirred at 0 °C for 1 h. The reaction was then diluted with DCM, washed with NaHCOs, dried over Na2SO<, filtered, and concentrated under reduced pressure to afford the desired crude product (37 mg, 100% yield). LCMS (ESI) m/z : [M + H] calcd for CsgHwNeOgSi: 1077.62; found 1077.6.

Step 4: Synthesis of (2fl)-1 -acetyl-N-(2-(((2S)-1 -(((6 3 S,4S)-1 ' -ethyl-2 5 -hydroxy-1 2 -(4- (methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)-N- methylaziridine-2-carboxamide

To a solution of (2/¾-1 -acetyl-N-(2-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)- 10,10-dimethyl-S.T-dioxo^-tttriisopropylsilyOoxyj-e 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)-N- methylaziridine-2-carboxamide (34 mg, 0.032 mmol) in MeCN (631 pL) at 0 °C was added TBAF (1 M in THE, 31.5 pL, 0.032 mmol). The reaction stirred for 10 min and was then diluted with DCM, washed with brine, dried over NazSC^, filtered, and concentrated under reduced pressure. Purification by reverse phase chromatography (10→100% MeCN/H 2 O) afforded the desired product (8.5 mg, 29% yield). LCMS (ESI) m/z·. [M + H] calcd for CsoHwNeOg: 921.49; found 921 .5.

Example 36. Synthesis of (2fl)-N- (2-(((2S)-1-(((6 3 S;4S)-1 1 -ethyl-2 6 -hydroxy-1 2 -(4- (methoxymethyl)pyrkJin-3-yl)-10,10-dimethyl-5,7-dloxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)-N- methyH-(methylsulfonyl)aziridine-2-cartx)xamide

Step i: Synthesis of (2fl)-N-(2-(((2S)-1-(((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)-N- methyl-1-(methylsulfonyl)aziridine-2-carboxamide

To a solution of (2fl)-N-(2-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan- 2-yl)(methyl)amino)-2- oxoethyl)-N-methylaziridine-2-carboxamide (33 mg, 0.032 mmol) in DCM (637 pL) at 0 °C was added EtsN (22.1 pL, 0.159 mmol) followed by methanesulfonyl chloride (4.93 pL, 0.064 mmol). The reaction was cooled to 0 °C for 1 h and was then diluted with DCM, washed with NaHCOs, dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired crude product (35 mg, 100% yield). LCMS (ESI) m/z. [M + H] calcd for CseHe+NeOioSSi: 1113.59; found 1113.6.

Step 2. Synthesis of (2fl)-W-(2-(((2S)-1-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(4- (methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)-N- methyl-1 -(methylsulfonyl)aziridine-2-carboxamide

To a solution of (2fl)-AA(2-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan- 2-yl)(methyl)amino)-2- oxoethyl)-N-methyl-1-(methylsulfonyl)aziridine-2-carboxamide (35 mg, 0.032 mmol) in MeCN (646 pL) at 0 °C was added TBAF (1 M in THE, 32.3 pL, 0.032 mmol). The reaction stirred for 10 min and was then diluted with DCM, washed with brine, dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by reverse phase chromatography (10→100% MeCN/H 2 O) afforded the desired product (20 mg, 65% yield). LCMS (ESI) m/z: [M + H] calcd for CtgHwNeOioS: 957.45; found 957.5.

Example 38. Synthesis of methyl (2fl)-2-((2-(((2S)-1-(((6 3 S;4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(4- (methoxymethyl)pyridln-3-yl)-10,10-dlmethyl-5,7-dloxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)- lndola-6(1 ,3)-pyrldazlna-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amlno)-2-oxoethyl)(methyl)carbamoyl)aziridine-1-c arboxylate

Step 1: Synthesis of methyl (2fi)-2-((2-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin- 3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)(methyl)carbamoyl)aziridine-1-c arboxylate

To a solution of (2fl)-/\A(2-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 ' H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan- 2-yl)(methyl)amino)-2- oxoethyl)-N-methylaziridine-2-carboxamide (46 mg, 0.044 mmol) in DCM (888 pL) at 0 °C was added BN (30.8 μΙ_, 0.22 mmol) followed by methyl chloroformate (4.46 μΙ_, 0.058 mmol). The reaction stirred at 0 °C for 1 h and then the reaction was diluted with DCM, washed with NaHCOa, dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired crude product (56 mg, 100% yield).

LCMS (ESI) mfr. [M + H] calcd for CsehtaNeOioSi: 1093.62; found 1093.7.

Step 2. Synthesis of methyl (2/3)-2-((2-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-2 s -hydroxy-1 2 -(4- (methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 ® -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)(methyl)carbamoyl)aziridine-1-c arboxylate

To a solution of methyl (2fl)-2-((2-(((2S)-1-(((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)(methyl)carbamoyl)aziridine-1-c arboxylate (56 mg, 0.051 mmol) in MeCN (1.0 mL) at 0 °C was added TBAF (1 M in THF, 51.2 μΙ_, 0.051 mmol). The reaction stirred for 15 min and was then diluted with DCM, washed with brine, dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by reverse phase chromatography (10→100% ΜΘΟΝ/ΗΣΟ) afforded the desired product (17 mg, 36% yield). LCMS (ESI) m/z : [M + H] calcd for CsoHwNeOio: 937.48; found 937.6.

Example 48 and 49. Synthesis of methyl (2S,3fl)-1 -((fl)-fe#t-buty Isulf lnyl)-3-((2-(((2S)-1 - (((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-di oxo- 6 1 ,6 2 ,6 3 ,6 4 ,6 6 ,6 e -hexahydro-1 1 H- 8-oxa-1 (5,3)-lndola-6(1 ,3)-pyrldazlna-2(1 ,3)- benzenacyck>undecaphane-4-yl)amino)-3-methyl-1-oxobutan-2 -yl)(methyl)amino)-2- oxoethyl)(methyl)carbamoyl)azlrldine-2-carboxylate and methyl (2S,3fl)-3-((2-(((2S)-1-(((6 3 S,4S)-1 1 - ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyrkJln-3-yl)-10,10-dimethyl-5,7-di oxo-6 1 , 6*, 6 s , β 4 , 6 s , 6·- hexahydro-1 1 H- 8-oxa-1 (5,3)-lndola-6(1 ,3)-pyrldazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)- 3-methyl-1-oxobutan-2-yl)(methyl)amlno)-2-oxoethyl)(methyl)c arbamoyl)azlrldlne-2-carboxylate

Step 1 : Synthesis of methyl (2S,3/¾-1-((fl)-te/f-butylsulfinyl)-3-((2-(((2S)-1-(((6 3 S,4S)-1 1 -ethyl-2 5 - hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-di oxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8- oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan- 2-yl)(methyl)amino)-2-oxoethyl)(methyl)carbamoyl)aziridine-2 -carboxylate

To a solution of (2S)-N-((63S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-3-methyl-2-(N-methyl-2-(methyl amino)acetamido)butanamide (267.0 mg, 0.33 mmol) and (2/?,3S)-1-((fl)-tert-butylsulfinyl)-3-(methoxycarbonyl)azir idine-2-carboxylic acid (246.5 mg, 0.99 mmol) in DMF (4.5 mL) at 0 °C was added DIPEA (0.574 mL, 3.3 mmol) followed by a solution of COMU (211 .8 mg, 0.49 mmol) in DMF (0.5 mL). The resulting mixture was stirred for 1 h at 0 °C and was then quenched with sat. NhUCI. The aqueous layer was extracted with EtOAc (3 x 20 mL) and the combined organic layers were washed with brine (2 x 50 mL), dried over Na 2 SO 4 , filtered, concentrated under reduced pressure. The crude product was purified by reverse phase chromatography (35→65% MeCN/H20) to afford the desired product (253 mg, 73.7% yield). LCMS (ESI) m/z. [M + H] calcd for CS4H72NBOIIS: 1041 .51 ; found 1041 .8.

Step 2: Synthesis of methyl (2S,3fl)-3-((2-(((2S)-1-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(4- (methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)(methyl)carbamoyl)aziridine-2-c arboxylate

To a solution of methyl (2S,3fl)-1-((/?)-tert-butylsulfinyl)-3-((2-(((2S)-1-(((6 3 S,4S)-1 1 -ethyl-2 5 - hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-di oxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8- oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan- 2-yl)(methyl)amino)-2-oxoethyl)(methyl)carbamoyl)aziridine-2 -carboxylate (200.0 mg, 0.19 mmol) in THF (4.0 mL) at 0 °C was added HI (1 .0 mL), dropwise. The resulting mixture was stirred for 10 min at 0 °C and was then basified to pH 7 with DIPEA. The mixture was extracted with EtOAc (3 x 30 mL) and the combined organic layers were washed with brine (50 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by reverse phase chromatography (35→65% MeCN/H 2 O) to afford the desired product (13.2 mg, 7.3% yield). LCMS (ESI) m/z. [M + H] calcd for CsoHe4NsOio: 937.48; found 938.6.

Example 55. Synthesis of (2S)-2-(2-((1/?,5S)-6-benzyl-2,4-dioxo-3,6- dlazablcyclo[3.1.0]hexan-3-yl)-N- methylacetamido)-N-((6 3 S,4S)-1 1 -ethyF2 5 -hydroxy-1 ^(A-

(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dloxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)- lndola-6(1,3)-pyridazlna-2(1,3)-benzenacycloundecaphane-4-yl )-3-methylbutanamlde Step 1: Synthesis of (2S)-2-(2-(2,5-dioxo-2,5-dihydro-1 //-pyrrol-1-yl)-N-methylacetamido)-N- ((6 3 S,4S)-1 1 -ethyl- 1 2 -(4-(methoxymethyl)py rid in-3-yl)- 10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane- 4-yl)-3-methylbutanamide

To a solution of (2S)-N-((6 3 S,4S)-1 ’-ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5, 7- dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)bu tanamide (600.0 mg, 0.67 mmol) and 2-(2,5-dioxo-2,5-dihydro-1 /-/-pyrrol-1 -yl)acetic acid (124.7 mg, 0.80 mmol) in DCM (6.0 mL) at 0 °C was added DIPEA (0.934 mL, 5.36 mmol) followed by HATU (382.2 mg, 1.01 mmol). The reaction mixture was warmed to room temperature and stirred for 3 h. The reaction was then quenched by the addition of hfeO (20 mL). The aqueous layer was extracted with DCM (2 x 50 mL) and the combined organic layers were washed with brine (2 x 50 mL), dried over Na 2 SO 4 , and concentrated under reduced pressure. The residue was purified by normal phase chromatography (10→20% EtOAc/pet. ether) to afford the desired product (260 mg, 33.8% yield). LCMS (ESI) m/τ. [M + H] calcd for CsyHTrNyOgSi: 1032.56; found 1032.8.

Step 2. Synthesis of (2S)-2-(2-((1 /?,5S)-6-benzyl-2,4-dioxo-3,6-diazabicyclo[3.1.0]hexan-3-yl) -N- methylacetamido)-N-((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-di oxo-2 5 - ((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 'H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-3-methylbutanamide

To a solution of (2S)-2-(2-(2,5-dioxo-2,5-dihydro-1 //-pyrrol-1 -yl)-N-methylacetamido)-N-((6 3 S,4S)- 1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-di oxo-2 5 -((triisopropylsilyl)oxy)-

6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 //-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane- 4-yl)-3-methylbutanamide (250.0 mg, 0.24 mmol) in EtOAc (2.0 mL) was added (azidomethyl)benzene (80.6 mg, 0.61 mmol). The reaction mixture was heated to 80 °C and stirred for 2 h. The reaction mixture was then heated to 120 °C and stirred for 2 days. The reaction mixture was then cooled to room temperature and quenched with H 2 O. The aqueous layer was extracted with EtOAc and the combined organic layers were washed with brine, dried with Na 2 SO 4 . filtered, and concentrated under reduced pressure. The residue was purified by normal phase chromatography to afford the desired product (50 mg, 18.1% yield). LCMS (ESI) m/z. [M + H] calcd for CwHMNeOgSi: 1137.62; found 1138.3.

Step 3: Synthesis of (2S)-2-(2-((1 /?,5S)-6-benzyl-2,4-dioxo-3,6-diazabicyclo[3.1 ,0]hexan-3-yl)-N- methylacetamido)-N-((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7- dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-3-methylbutanamide

To a solution of (2S)-2-(2-((1 f?,5S)-6-benzyl-2,4-dioxo-3,6-diazabicyclo[3.1 ,0]hexan-3-yl)-N- methylacetamido)-N-((6 3 S,4S)-1 1 -ethyl-1 2 -(4-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-di oxo-2 5 - ((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-3-methylbutanamide (50.0 mg, 0.04 mmol) in THF (0.5 mL) at 0 °C was added 1 M TBAF (0.07 mL, 0.07 mmol). The reaction mixture was stirred for 1 h. The reaction mixture was then diluted with H 2 O and extracted with EtOAc. The combined organic layers were washed with brine, dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by prep- TLC followed by reverse phase chromatography (45→72% MeCN/H 2 O) to afford the desired product (20 mg, 46.4% yield). LCMS (ESI) m/z : [M + Na] calcd for CssHwNeOg: 1003.47; found 1003.8. Example 95. Synthesis of (2fl)- N-(2-(((1S)-1 -cyclopentyl-2-(((6 3 S,4S)-1 1 -ethyl-2 5 - hydroxy- 1 2 - (2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 e ,6 6 -hexahydro-1 1 H-8-oxa- 1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-2- oxoethyl)(methyl)amino)-2-oxoethyl)-AFmethylaziridine-2-cart x)xamide

Step 1: Synthesis of (2fl)-N-(2-(((1 S)-1 -cyclopentyl-2-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-2-oxoethyl)(methyl)a mino)-2-oxoethyl)- W-methyl-1-tritylaziridine-2-carboxamide

To a mixture of (2S)-2-cyclopentyl-N-((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)-2-(N-methyl-2-

(methylamino)acetamido)acetamide (321.2 mg, 0.276 mmol), DIPEA (0.472 mL, 2.764 mmol), and (fl)-1- tritylaziridine-2-carboxylic acid (136.59 mg, 0.415 mmol) in DMF (3.0 mL) at 0 °C was added HATU (126.14 mg, 0.332 mmol). The resulting mixture was stirred at 0 °C for 30 min, then diluted with H 2 O (30 mL) and extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (3 x 10 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by prep-TLC (50% EtO Ac/pet. ether) afforded the desired product (200 mg, 62.3% yield). LCMS (ESI) m/z. [M + H] calcd for CroHaoNeOe: 1161.62; found 1161.5.

Step 2. Synthesis of (2fl)-N-(2-(((1 S)-1 -cyclopentyl-2-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacydoundecaphane-4-yl)amino)-2-oxoethyl)(methyl)am ino)-2-oxoethyl)- W-methylaziridine-2-carboxamide

To a mixture of (2fl)-N-(2-(((1 S)-1 -cyclopentyl-2-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-2-oxoethyl)(methyl)a mino)-2-oxoethyl)- N-methyl-1-tritylaziridine-2-carboxamide (195.0 mg, 0.168 mmol) in DCM (2.0 mL) at 0 °C was added EtsSiH (78.09 mg, 0.672 mmol) and TFA (76.57 mg, 0.672 mmol). The resulting mixture was stirred at 0 °C for 30 min then basified to pH 8 with DIPEA and concentrated under reduced pressure. Purification by reverse phase chromatography (25→55% MeCN/H 2 O) to afford the desired product (60 mg, 38.9% yield). LCMS (ESI) m/z. [M + H] calcd for CsiHeeNeOe: 919.51 ; found 919.5. Example 87. Synthesis of 6-((S)-azlridln-2-yl)-yV-((2S)-1-(((6 3 S;4S)-1 1 -ethyl-2 6 -hydroxy-1 2 -(2- ((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa- 1 (5,3)-indola-€(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan- 2-yl)-N- methylnicotinamide

Step 1: Synthesis of 6-((2S)-1 -(tert-butylsulfinyl)aziridin-2-yl)-N-((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2- ((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((tr iisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 e - hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3- methyl-1-oxobutan-2-yl)-fV-methylnicotinamide

To a mixture of (2S)-N-((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl- 5,7-dioxo-2 5 -((triisopropylsilyl)oxy)- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 e -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)bu tanamide (198.24 mg, 0.218 mmol) and DIPEA (0.074 mL, 0.436 mmol) in MeCN (10 mL) at 0 °C was added HATU (200 mg, 0.526 mmol) and the resulting mixture was stirred for 3 min. To the mixture was then added a solution of 6-((2S)-1 - (tert-butylsulfinyl)aziridin-2-yl)nicotinic acid (117.0 mg, 0.436 mmol) in MeCN (10 mL) in portions. The resulting mixture was stirred overnight at 0 °C and then was then quenched with hfeO extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (20 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (430 mg, 85.0% yield). LCMS (ESI) m/z. [M + H] calcd for CwHgoNeOeSSi: 1159.65; found 1159.8.

Step 2. Synthesis of 6-((2S)-1 -(tert-butylsulfinyl)aziridin-2-yl)-N-((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-2 5 - hydroxy-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 - oxobutan-2-yl)-N-methylnicotinamide

To a solution of 6-((2S)-1-(tert-butylsulfinyl)aziridin-2-yl)-N-((2S)-1-(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((tri isopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro- 1 ’ H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1- oxobutan-2-yl)-N-methylnicotinamide (430.0 mg, 0.371 mmol) in THF (50.0 mL) at 0 °C was added TBAF (1 M in THF, 1.1 mL, 1.11 mmol) in portions. The resulting mixture was stirred at 0 °C for 2 h and was then concentrated under reduced pressure. The residue was purified by prep-TLC (5% MeOH/DCM) to afford the desired product (290 mg, 78% yield). LCMS (ESI) m/z: [M + H] calcd for CssHyoNeOeS: 1003.51 ; found 1003.8.

Step 3: Synthesis of 6-((S)-aziridin-2-yl)-W-((2S)-1-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)-N- methylnicotinamide

To a solution of 6-((2S)-1-(fe/t-butylsulfinyl)aziridin-2-yl)-N-((2S)-1-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy- 1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 ® -hexahydro-1 1 H- 8-oxa- 1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)- ΛΑ-methylnicotinamide (150.0 mg, 0.150 mmol) in H 2 O (15.0 mL) and acetone (15.0 mL) at 0 °C was added TFA (7.50 mL, 100.97 mmol) in portions. The resulting mixture was warmed to room temperature and stirred for 48 h and then was neutralized to pH 8 with sat. NaHCOa. The aqueous layer was extracted with EtOAc, dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by reverse phase chromatography (38→58% MeCN/H 2 O) afforded the desired product (10.0 mg, 7.4% yield). LCMS (ESI) m/z\ [M + H] calcd for CsiHeaNeO?: 899.48; found 899.5.

Example 139. Synthesis of (2S)-2-((S)-7-(((fl)-azirldln-2-yl)methyl)-1-oxo-2,7- dlazasplro[4.4]nonan-2-yl)-N- ((6 3 S,4S)-1 ’-ethyl^-hydroxy-l 2 -(2-((S)-1 -methoxyethyl)pyrldin-3-yl)-

10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-lndola-6(1 ,3)-pyridazlna- 2(1,3)-benzenacycloundecaphane-4-yl)-3-methylbutanamkle

Step 1: Synthesis of tert-butyl (5f?)-7-((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-2 5 - hydroxy-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-SJ-dioxo-e 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-

6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)-6-oxo-2,7- diazaspiro[4.4]nonane-2-carboxylate

To a solution of (6 3 S,4S)-4-amino-1 '-ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)- 10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 ® -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione (600 mg, 0.94 mmol) and DIPEA (820 μΙ_, 4.7 mmol) in DMF (8 mL) at 0 °C was added (S)-2-((fl)-7-(tert-butoxycarbonyl)-1-oxo-2,7-diazaspiro[4.4 ]nonan-2-yl)-3- methylbutanoic acid (380 mg, 1.13 mmol) and COMU (440 mg, 1.03 mmol). The reaction mixture was stirred for 1 h then was diluted with H 2 O (100 mL) and extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (30 mL), dried with Na 2 SO 4 , filtered, and the filtrate was concentrated under reduced pressure. Purification by Prep-TLC (EtOAc) afforded the desired product (600 mg, 66% yield). LCMS (ESI) m/z: [M + H] calcd for C54H71N7O9: 962.54; found 962.5.

Step 2: Synthesis of (2S)-N- ((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-3-methyl-2-((S)-1-oxo-2,7-diaz aspiro[4.4]nonan-2-yl)butanamide

To a solution of tert-butyl (5fl)-7-((2S)-1-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacydoundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)-6-oxo-2,7- diazaspiro[4.4]nonane-2-carboxylate (600 mg, 0.62 mmol) in DCM (6 mL) at 0 °C was added TFA (3.0 mL, 40 mmol). The reaction mixture was stirred for 2 h and then was concentrated under reduced pressure. The residue was diluted with H 2 O (100 mL), basified to pH 8 with sat. aq. NaHCOs, and extracted with EtOAc (3 x 60 mL). The combined organic layers were washed with brine (30 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product (430 mg, 79% yield). LCMS (ESI) m/z. [M + H] calcd for C49H63N7O7: 862.49; found 862.5.

Step 3: Synthesis of (2S)-N- ((6 3 S,4S)-1 , -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-

10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-3-methyl-2-((S)-1-oxo-7-(((S)- 1-tritylaziridin-2-yl)methyl)-2,7- diazaspiro[4.4]nonan-2-yl)butanamide

To a solution of (2S)-N-((6 3 S,4S)-1 1 -ethyl-2 s -hydroxy-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-

10.10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 , 6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-3-methyl-2-((S)-1 -oxo-2, 7-diazaspiro[4.4]nonan-2-yl)butanamide (200 mg, 0.23 mmol) and (fl)-1 -tritylaziridine-2-carbaldehyde (110 mg, 0.35 mmol) in MeOH (0.50 mL) and MeCN (4.0 mL) was added NaBHsCN (29 mg, 0.46 mmol). The reaction mixture was stirred for 2 h then was quenched with sat. aq. NH4CI and was extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (30 mL), dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by Prep-TLC (EtOAc) afforded desired product (145 mg, 53% yield). LCMS (ESI) m/z. [M + H] calcd for C71H82N8O7: 1159.64; found 1159.6.

Step 4: Synthesis of (2S)-2-((S)-7-(((fl)-aziridin-2-yl)methyl)-1-oxo-2,7-diazasp iro[4.4]nonan-2-yl)- N-((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-

6 1 ,6 2 6 3 ,6 4 , 6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane- 4-yl)-3-methylbutanamide

To a solution of (2S)-N-((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-

10.10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-3-methyl-2-((S)-1 -oxo-7-(((S)-1 -tritylaziridin-2-yl)methyl)-2,7- diazaspiro[4.4]nonan-2-yl)butanamide (140 mg, 0.12 mmol) in DCM (2.0 mL) 0 °C was added TFA (74 pL, 0.97 mmol) and EtsSiH (150 pL, 0.97 mmol). The reaction mixture was stirred for 30 min then was basified to pH 8 with DIPEA. The resulting mixture was concentrated under reduced pressure. Purification by reverse phase chromatography (30→60% MeCN/H 2 O) afforded the desired product (37.5 mg, 31% yield). LCMS (ESI) m/z. [M + H] calcd for ΟκΗββΝβΟ: 917.53; found 917.4. Example 133. Synthesis of (2/?,3fl)-3-cyclopropyl-N- (2-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyk2 6 - hydroxy-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 e - hexahydro-1 1 M-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)- 3-methyl-1-oxobutan-2-yl)(methyl)amino)-2-oxoethyl)-Af-methy laziridine-2-carboxamide

Step 1: Synthesis of (2/?,3fl)-1-(tert-butylsulfinyl)-3-cyclopropyl-N-(2-(((2S)-1 -(((6 3 S,4S)-1 '-ethyl- 2 5 -hydroxy-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 2 , 6 3 ,6 4 ,6 5 ,6 6 -hexahydro- 1 ' H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1- oxobutan-2-yl)(methyl)amino)-2-oxoethyl)-N-methylaziridine-2 -carboxamide

To a solution of (2S)-N-((6 3 S,4S)-1 '-ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)- 10,10-dimethyl-SJ-dioxo-O' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-3-methyl-2-(N-methyl-2-(methyl amino)acetamido)butanamide (50 mg,

61 pmol) and (2R,3fl)-1 -(tert-butylsulfinyl)-3-cyclopropylaziridine-2-carboxylic acid (21 mg, 91 pmol) in MeCN at 0 °C was added DIPEA (210 μΙ_, 1.2 mmol) and CIP (25 mg, 91 μιτιοΙ). The resulting mixture was stirred for 2 h and was then concentrated under reduced pressure. Purification by Prep-TLC (9%

EtOAc/pet. ether) afforded the desired product (270 mg, 54% yield). LCMS (ESI) m/z. [M + H] calcd for CseHyeNeOgS: 1037.56; found 1037.4.

Step 2: Synthesis of (2 R,3 fl)-3-cyclopropy l-N-(2-(((2 S)-1 -(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2- ((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 6 5 ,6 ® -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)-N-methylaziridine-2-carboxamid e

To a mixture of (2R,3R)-1-(tert-butylsulfinyl)-3-cyclopropyl-A/-(2-(((2S)-1- (((6 3 S,4S)-1 1 -ethyl-2 5 - hydroxy-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 - oxobutan-2-yl)(methyl)amino)-2-oxoethyl)-/SAmethylaziridine- 2-carboxamide (230 mg, 0.22 mmol) in THF at 0 °C was added HI (0.50 mL, 3.8 mmol, 57% wt in H 2 O). The reaction mixture was stirred for 10 min and then neutralized to pH 8 with DIPEA and concentrated under reduced pressure. Purification by reverse phase chromatography (40→60% MeCN/H 2 O) afforded desired product (20 mg, 11% yield) as a white solid. LCMS (ESI) m/z. [M + H] calcd for CsaHeeNeOe: 933.53; found 933.6. Example 177. Synthesis of 4-((/i)-aziridine-2-carbonyl)-A/-((2S)-1-(((6 3 S;4S)-1 1 -ethyl-2 5 - hydroxy-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 e - hexahydro-1 1 M-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)- 3-methyl-1 -oxobutan-2-yl)-N- methylpiperazine-1 -carboxamide

Step f: Synthesis of tert-butyl (S)-4-((1 -(benzyloxy)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamoyl)piperazine-1-carboxylate

Into a 100-mL vial were added benzyl methyl-L-valinate (2.0 g, 9.038 mmol) and triphosgene (0.89 g, 2.982 mmol) in DCM (30 mL) followed by pyridine (2.14 g, 27.113 mmol) in portions at 0 °C under an N 2 atmosphere. The mixture was stirred for 2 h at room temperature. The crude product was used in the next step directly without further purification. Then, the resulting mixture was added to tert-butyl piperazine-1 -carboxylate (2.22 g, 11.912 mmol) in DCM (25 mL) and EtsN (2.78 g, 27.489 mmol) in portions at room temperature under an N 2 atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, (30% EtO Ac/pet. ether) to afford the desired product (3.6 g, 90.6% yield). LCMS (ESI) m/z: [M + H] calcd for C23H35N3O5: 434.26; found 434.2.

Step 2. Synthesis of N-(4-(tert-butoxycarbonyl)piperazine-1 -carbonyl)-A/-methyl-L-valine

Into a 100-mL vial were added tert- butyl (S)-4-((1 -(benzyloxy)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamoyl) piperazine-1 -carboxylate (2.95 g, 6.804 mmol) and Pd/C (1 .48 g) in THF (25 mL). the reaction was stirred for overnight at room temperature under a hydrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with EtOAc (3 x 50 mL), and the combined organic layers were concentrated under reduced pressure to afford the desired product (2.4 g, crude). LCMS (ESI) m/z: [M + H] calcd for C16H29N3O5: 344.21 ; found 344.4.

Step 3: Synthesis of tert-butyl 4-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3- yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6',6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-

6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamoyl)piperazine-1 -carboxylate

Into a 50-mL vial was added (6 3 S,4S)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)- 10,10-dimethyl-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloun decaphane-5,7-dione (1.0 g, 1.256 mmol) and W-(4-(tert- butoxycarbonyl) piperazine-1 -carbonyl)-N-methyl-L-valine (647.04 mg, 1.884 mmol) in DMF (8 mL) followed by HATU (668.63 mg, 1.758 mmol) and DIPEA (811.69 mg, 6.280 mmol) in portions at room temperature. The resulting mixture was extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (2 x 20 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (50% EtOAc/pet. ether) to afford the desired product (1.08 g, 76.7% yield). LCMS (ESI) m/z: [M + H] calcd for CtoHreNeOgSi: 1121.68; found 1122.0.

Step 4: Synthesis of N-((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 e -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan- 2-yl)-N-methylpiperazine- 1 -carboxamide

Into a 100-mL vial was added tert-butyl 4-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6',6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro- 1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 - oxobutan-2-yl)(methyl)carbamoyl)piperazine-1 -carboxylate (1.08 g, 0.963 mmol) and TFA (3.0 mL, 40.39 mmol) in DCM (12 mL). The reaction was stirred for 2 h at room temperature under an h½ atmosphere. The resulting mixture was concentrated under reduced pressure to afford the desired product (907 mg, crude). LCMS (ESI) m/z: [M + Na] calcd for CsrHeaNeOySi: 1042.61 ; found 1043.9.

Step 5: Synthesis of N-((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)-N-methyl-4-((fi)-1 - tritylaziridine-2-carbonyl)piperazine-1 -carboxamide

Into a 40-mL vial was added N-((2S)-1 -(((6 3 S,4S)-1 ' -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-2 s -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)-N- methylpiperazine-1 -carboxamide (400.0 mg, 0.392 mmol) and (fl)-1 -tritylaziridine-2-carboxylic acid (193.49 mg, 0.587 mmol) in DMF (3.5 mL) followed by HATU (208.46 mg, 0.548 mmol) and DIPEA (253.06 mg, 1.958 mmol) in portions at room temperature under an Na atmosphere. The resulting mixture was extracted with EtOAc (3 x 60 mL) and the combined organic layers were washed with brine (2 x 10 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, (50% EtOAc/pet. ether) to afford the desired product (367 mg, 70.3% yield). LCMS (ESI) m/z: [M + H -TIPS] calcd for CTSHIOINSOBSI: 1176.63; found 1176.2.

Step 6: Synthesis of N-((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3- yl)-10,10-d imethyl-5 ,7-d ioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)-N-methyl-4-((fl)-1-tritylaziridine-2- carbonyl)piperazine-1 -carboxamide

Into a 100-mL vial was added N-((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3- yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)-A/-methyl-4- ((fl)-1 -tritylaziridine-2-carbonyl)piperazine-1 -carboxamide (161.0 mg, 0.121 mmol) and CsF (91.75 mg, 0.604 mmol) in DMF (1.5 mL). The reaction was stirred for 2 h at room temperature and was then extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (2 x 10 mL), dried over NaaSO^ filtered, and concentrated under reduced pressure. The residue was purified by Prep- TLC (50% EtOAc/pet. ether) to afford the desired product (101 mg, 71.1% yield). LCMS (ESI) m/z: [M +

H] calcd for CyoHsiNgOs: 1176.62; found 1176.9.

Step 7: Synthesis of 4-((fl)-aziridine-2-carbonyl)-N- ((2S)-1 -(((6 3 S,4S)-1 , -ethyl-2 5 -hydroxy-1 2 -(2- ((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)-N- methylpiperazine-1 -carboxamide

Into a 40-mL vial was added A/-((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-2 5 - hydroxy-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacyclou ndecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)-N-methyl-4- ((fl)-1 -tritylaziridine-2-carbonyl) piperazine-1 -carboxamide (101.0 mg, 0.086 mmol) and EfaSiH (49.91 mg,

0.429 mmol) in DCM (2.0 mL) was added TFA (48.94 mg, 0.429 mmol) in portions at room temperature under an Na atmosphere. The mixture was basified to pH 8 with DIPEA. The crude product was purified by Prep-HPLC to afford the desired product (29.6 mg, 36.9% yield). LCMS (ESI) m/z: [M + H] calcd for CsiHeyNgOe: 934.51 ; found 934.3.

Example 175. Synthesis of (2S)-2-((S)-7-((/I)-azirldlne-2-carbonyl)-1-oxo-2,7- diazasplro[4.4]nonan-2-yl)-2-cyclopentyl-N- ((6 3 S,4S)-1 '-ethyl^-hydroxy-l^-ttSH - methoxyethyl)pyrldln-3-yl)-10,10-dimethyl-S.T-dioxo-e 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)- lndola-6(1 ,3)-pyrldazlna-2(1 ,3)-benzenacycloundecaphane-4-yl)acelamlde

Step 1: Synthesis of (S)-2-((fl)-7-(tert-butoxycarbonyl)-1 -oxo-2, 7-diazaspiro[4.4]nonan-2-yl)-2- cyclopentylacetic acid

To a solution stirred solution of terf-butyl (fl)-7-((S)-2-(benzyloxy)-1-cyclopentyl-2-oxoethyl)-6- oxo-2, 7-diazaspiro[4.4]nonane-2-carboxylate (1.0 g, 2.19 mmol) in MeOH (10 mL) at 0 °C was added Pd/C (200 mg). The resulting mixture was stirred for 1 h at room temperature under a hydrogen atmosphere, filtered, and the filter cake washed with MeOH (5 x10 mL). The filtrate was concentrated under reduced pressure to afford the desired product (895 mg, crude) which was used without further purification. LCMS (ESI) m/z·. [M + H] calcd for C19H30N 2 O5: 376.23; found 367.1.

Step 2: Synthesis of tert-butyl (5fl)-7-((1 S)-1 -cyclopentyl-2-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2- ((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-2-oxoethyl)-6-oxo-2, 7- diazaspiro[4.4]nonane-2-carboxylate

To a stirred solution of (6 3 S,4S)-4-amino-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1-methoxyethyl)pyridin-3- yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione (702 mg, 1.10 mmol) and DIPEA (1.91 mL, 1.10 mmol) in DMF (500 mL) at 0 °C was added (S)-2-((fl)-7-(tert-butoxycarbonyl)-1-oxo-2,7-diazaspiro[4.4 ]nonan-2-yl)-2- cyclopentylacetic acid (523 mg, 1.43 mmol) and COMU (517 mg, 1.21 mmol). After 1 h at room temperature the reaction mixture was diluted with H 2 O (150 mL). The aqueous layer was extracted with EtOAc (3 x 50 mL) and the combined organic layers were washed with brine, dried with NapSO*. filtered, and concentrated under reduced pressure. Purification by normal phase chromatography (40% EtOAc/pet. ether) afforded the desired product (978 mg, 90.2% yield). LCMS (ESI) m/z. [M + H] calcd for C56H73N7O9: 988.56; found 988.7.

Step 3: Synthesis of (2S)-2-cyclopentyl-N- ((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-

6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)-2-((S)-1 -oxo-2,7-diazaspiro[4.4]nonan-2- yl)acetamide

To a stirred solution of terf-butyl (5fl)-7-((1 S)-1 -cyclopentyl-2-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2- ((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dίoxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-2-oxoethyl)-6-oxo-2, 7- diazaspiro[4.4]nonane-2-carboxylate (300 mg, 0.304 mmol) in DCM (3.0 mL) at 0 °C was added TFA (1.5 mL). The resulting mixture was stirred for 30 min at room temperature. The reaction mixture was then diluted with toluene (2 mL) and concentrated under reduced pressure three times to afford the desired product (270 mg, crude) which was used without further purification. LCMS (ESI) m/z: [M + H] calcd for CSIHMNTO/: 888.50; found 888.5.

Step 4: Synthesis of (2S)-2-cyclopentyl-W-((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)-2-((S)-1 -oxo-7-((fl)-1 -tritylaziridine-2-carbonyl)- 2,7-diazaspiro[4.4]nonan-2-yl)acetamide

To a stirred solution of (2S)-2-cyclopentyl-N- ((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)-2-((S)-1 -oxo-2,7-diazaspiro[4.4]nonan-2- yl)acetamide (270 mg, 0.304 mmol) and DIPEA (0.53 mL, 3.0 mmol) in DMF (3.0 mL) at 0 °C was added (fl)-1 -tritylaziridine-2-carboxylic acid (130 mg, 0.395 mmol) and COMU (143 mg, 0.334 mmol). After 1h at room temperature the reaction mixture was diluted with H 2 O (30 mL). The aqueous layer was extracted with EtOAc (3 x 3 mL) and the combined organic layers were washed with brine, dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by prep-TLC (5% MeOH/DCM) afforded the desired product (332 mg, 91.1% yield). LCMS (ESI) m/z. [M + H] calcd for CysHezNeOe: 1199.64; found 1199.7.

Step 5: Synthesis of (2S)-2-((S)-7-((fl)-aziridine-2-carbonyl)-1-oxo-2,7-diazaspi ro[4.4]nonan-2-yl)- 2-cyclopentyl-N-((6 3 S,4S)-1 1 -ethy l-2 5 -hydroxy- 1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7- dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)acetamide

To a stirred solution of (2S)-2-cyclopentyl-N-((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 6 3 ,6 4 6 5 ,6 ® -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)-2-((S)-1 -oxo-7-((fl)-1 -tritylaziridine-2-carbonyl)- 2,7-diazaspiro[4.4]nonan-2-yl)acetamide (309 mg, 0.258 mmol) in DCM (3.0 mL) at 0 °C was added

EtsSiH (164 mL, 1.03 mmol) and TFA (79 mL, 1.03 mmol). After 30 min the reaction mixture was basified to pH 8 with DIPEA and concentrated under reduced pressure. Purification by reverse phase chromatography (30→60% MeCN/H 2 O) afforded the desired product (36 mg, 14.2% yield). LCMS (ESI) m/z. [M + H] calcd for CstHeeNeOe: 957.53; found 957.3.

Example 214. Synthesis of (2S)-2-cyclopentyl-2-((S)-7-((2/?,3fl)-3-cyclopropylazirldln e-2- carbonyl)-1 -oxo-2,7-diazaspiro[4.4]nonan-2-yl)-W-((6 3 S,4S)-1 1 -ethyF2 5 -hydroxy-1 2 -(2-((S)-1 - methoxyethyl)pyrldln-3-yl)-10.10-dlmethyFSJ-dloxo-e 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)- lndola-6(1 ,3)-pyrldazlna-2(1 ,3)-benzenacycloundecaphane-4-yl)acelamlde

Step 1: Synthesis of (2S)-2-((5S)-7-((2fl,3fl)-1-(fe/t-butylsulfinyl)-3-cycloprop ylaziridine-2- carbonyl)-1 -oxo-2,7-diazaspiro[4.4]nonan-2-yl)-2-cyclopentyl-N- ((6 3 S,4S)-1 '-ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)acetamide

To a stirred solution of (2S)-2-cyclopentyl-N-((6 3 S,4S)-1 1 -ethyl-2 s -hydroxy-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)-2-((S)-1 -oxo-2,7-diazaspiro[4.4]nonan-2- yl)acetamide (270 mg, 0.30 mmol) in DMF (3.0 mL) at 0 °C was added DIPEA (530 pL, 3.0 mmol) and (2/?,3fl)-1-(tert-butylsulfinyl)-3-cyclopropylaziridine-2-ca rboxylic acid (105 mg, 0.46 mmol) followed by COMU (140 mg, 0.33 mmol). The resulting mixture was stirred for 1 h at room temperature and was then diluted with H 2 O (30 mL). The reaction mixture was extracted into EtOAc (3 x 7 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by Prep-TLC (6% MeOH/DCM) afforded the desired product (237 mg, 71% yield). LCMS (ESI) m/z. [M + H] calcd for CeiHsoNeOgS: 1101.58; found 1101.3.

Step 2. Synthesis of (2S)-2-cyclopentyl-2-((S)-7-((2f?,3fl)-3-cyclopropylaziridin e-2-carbonyl)-1- oxo-2,7-diazaspiro[4.4]nonan-2-yl)-N-((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1-methoxyethyl)pyridin-3- yl)-10,10-d imethy I-5 ,7-d ioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)acetamide

To a stirred solution of (2S)-2-((5S)-7-((2fl,3fl)-1-(tert-butylsulfinyl)-3-cycloprop ylaziridine-2- carbonyl)-1 -oxo-2,7-diazaspiro[4.4]nonan-2-yl)-2-cyclopentyl-N- ((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)acetamide (230 mg, 0.21 mmol) in THF (2.5 mL) at 0 °C was added EtsSiH (130 pL, 0.83 mmol) and HI (125 pL, 0.41 mmol, 57% in H 2 O). The resulting mixture was stirred for 30 min at room temperature then cooled to 0 °C and neutralized to pH 8. The mixture was concentrated under reduced pressure. Purification by Prep-TLC (8.3% MeOH/DCM) afforded the desired product (46 mg, 21% yield). LCMS (ESI) m/z. [M + H] calcd for Cs/HyaNeOe: 997.55; found 997.2.

Example 209. Synthesis of (2fl)-N- (2-(((1S)-1-cyclopentyl-2-(((6 3 S;4S)-1 1 -ethyl- 25 - h |r <> x y-1 2 - (2-((S)-1 -methoxyethyl)-5-(4-methylpiperazin-1 -yl)pyrkJin-3-yl)-10,10-dimethyl-5,7-dioxo- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)amino)-2-oxoethyl)(methyl)amino )-2-oxoethyl)-N-methylazirldine- 2-carboxamide

-Trt -O N' v oo I o 1 H O j o u* °w\

HATU, DIPEA EtsSIH. TFA 8 ΎΊΓΤ ν

DCM

DMF

O / o N— '

/

Step 1: Synthesis of benzyl ((1 S)-1 -cydopentyl-2-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 - methoxyethyl)-5-(4-methylpiperazin-1 -yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-2- oxoethyl)(methyl)carbamate To a stirred solution of (6 3 S, 4S)-4-amino-1 '-ethyl-2 5 -hydroxy-1 2 -(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1 -yl)pyridine-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-5,7-dione (490 mg, 0.664 mmol) and (S)-2- (((benzyloxy)carbonyl)(methyl)amino)-2-cyclopentylacetic acid (232 mg, 0.797 mmol) in DMF (5 mL) at 0 °C was added DIPEA (1.19 mL, 6.64 mmol) and HATU (303 mg, 0.797 mmol). The resulting mixture was stirred for 1 h at room temperature and then diluted with H 2 O (20 mL). The aqueous phase was extracted with EtOAc (3 x 20 mL) and the combined organic layers were washed with brine, dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by reverse phase chromatography (0→100% MeCN/H 2 O, 0.1% NH4HCO3) afforded the desired product (420 mg, 59.4% yield). LCMS (ESI) m/z. [M + H] calcd for CsehUNeOe: 1011.57; found 1011.6.

Step 2 Synthesis of (2S)-2-cyclopentyl-N-((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1- methoxyethyl)-5-(4-methylpiperazin-1 -yl)pyridin-3-yl)-10,10-dimethyl-SJ-dioxo-e 1 ,β^,β^,β 6 - hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)-2- (methylamino)acetamide

To a stirred solution of benzyl ((1 S)-1-cyclopentyl-2-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1- methoxyethyl)-5-(4-methylpiperazin-1 -yl)pyridin-3-yl)-10,10-dimethyl-SJ-dioxo-e 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-2- oxoethyl)(methyl)carbamate (450 mg, 0.445 mmol) in f-BuOH (10 mL) was added Pd/C (90 mg). The resulting mixture was warmed to 40 °C overnight under a hydrogen atmosphere, then filtered and the filter cake washed with MeOH. The filtrate was concentrated under reduced pressure to afford the desired product (420 mg, crude) which was used without further purification. LCMS (ESI) m/z [M + H] calcd for CsoHeeNeOe: 877.54; found 877.5.

Step 3: Synthesis of (2fl)-N-(2-(((1 S)-1 -cyclopentyl-2-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1- methoxyethyl)-5-(4-methylpiperazin-1 -yl)pyridin-3-yl)-10,10-dimethyl-SJ-dioxo-e 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-2- oxoethyl)(methyl)amino)-2-oxoethyl)-N-methyl-1-tritylaziridi ne-2-carboxamide

To a stirred solution of (2S)-2-cyclopentyl-N-((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1- methoxyethyl)-5-(4-methylpiperazin-1 -yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,β^,β^,β 6 - hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)-2- (methylamino)acetamide (130 mg, 0.148 mmol) and lithium (fl)-N-methyl-N-(1-tritylaziridine-2- carbonyl)glycinate (78.3 mg, 0.193 mmol) in DMF (2 mL) at 0 °C was added DIPEA (264 mL, 1.48 mmol) and HATU (68 mg, 0.178 mmol). The resulting mixture was stirred for 1 h at room temperature and then diluted with H 2 O (20 mL). The aqueous phase was extracted with EtOAc (3 x 10 mL) and the combined organic layers were washed with H 2 O, dried with Na 2 SO 4 , filtered, and concentrated under reduced pressure. Purification by prep-TLC (10% MeOH/DCM) afforded the desired product (100 mg, 50.9% yield). LCMS (ESI) m/z. [M + Na] calcd for CysHgoNtoOe: 1281.69; found 1281.9.

Step 4: Synthesis of (2/7)-N-(2-(((1 S)-1 -cyclopentyl-2-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 - methoxyethyl)-5-(4-methylpiperazin-1 -yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 1 FF8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-2- oxoethyl)(methyl)amino)-2-oxoethyl)-A/-methylaziridine-2-car boxamide

To a stirred solution of (2fl)-N-(2-(((1 S)-1 -cyclopentyl-2-(((6 3 S,4S)-1 , -ethyl-2 5 -hydroxy-1 2 -(2-((S)- 1 -methoxyethyl)-5-(4-methylpiperazin-1 -yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-2- oxoethyl)(methyl)amino)-2-oxoethyl)-N-methyl-1-tritylaziridi ne-2-carboxamide (100 mg, 0.079 mmol) in DCM (1.0 mL) at 0 °C was added EtsSiH (51 mL, 0.318 mmol) and TFA (24 mL, 0.318 mmol). After 30 min the reaction mixture was basified to pH 8 with DIPEA and concentrated under reduced pressure. Purification by reverse phase chromatography (30→55% MeCN/H 2 O) afforded the desired product (14mg, 16.5% yield). LCMS (ESI) m/z. [M + H] calcd for CseHyeNioOe: 1017.59; found 1017.6.

Example 268. Synthesis of (2fl)-/tf-(2-(((2S)-1-(((6 3 S > 4S)-1 1 -ethyl-2 e -hydroxy-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-S^-dioxo-e 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 M-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amlno)-3-methyl-1 -oxobutan-2- yl)(methyl)amlno)-2-oxoethyl)-N- methyH-(3-(2-oxopyrrolldln-1-yl)propyl)azlrldlne-2-carboxaml de

Step 1: Synthesis of (2fl)-N-(2-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 8 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)-N- methyl-1-tritylaziridine-2-carboxamide

To a solution of (1 S)-1 -tritylaziridine-2-carboxylic acid (537.6 mg, 1.63 mmol), (2S)-N-((6 3 S,4S)- 1 '-ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)- 6',6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane- 4-yl)-3-methyl-2-(N-methyl-2-(methylamino)acetamido)butanami de (800 mg, 0.816 mmol) in THF (8 mL) was added DIPEA (0.711 mL, 4.08 mmol), HATU (465.4 mg, 1.22 mmol) at 0 °C, the reaction was warmed to room temperature and stirred for 2 h. To the reaction was added H 2 O (20 mL), the aqueous phase was extracted with DCM (3 x 30 mL) and the combined organic phases were washed with brine (20 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0→100% EtOAc/pet. ether) to afford the desired product (1 g, 94.9% yield). Step 2. Synthesis of (2f7)-N-(2-(((2S)-1-(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-

10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)-N- methylaziridine-2-carboxamide

To a solution of (2fl)-AA(2-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridine-3 -yl)-

10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)-N- methyl-1-tritylaziridine-2-carboxamide (1 g, 0.774 mmol) in MeOH (5 mL) and CHCIs (5 mL) was added TEA (1.15 mL, 15.48 mmol) at 0 °C. The reaction was warmed to room temperature and stirred for 2 h. The reaction mixture was added dropwise to aq. NaHCOs (30 mL) at 0 °C. Then the pH was adjusted to pH 7-8 with using aq. NaHCOs at 0 °C. The mixture was extracted with DCM (3 x 20 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the desired product product (960 mg, crude), which was used directly in the next step without further purification.

Step 3: Synthesis of (2fl)-N-(2-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-

10.10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)-N- methyl-1-(3-(2-oxopyrrolidin-1-yl)propyl)aziridine-2-carboxa mide

To a solution of (2/¾-N-(2-(((2S)-1-(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridine-3-yl)- 10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-

6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)-N- methylaziridine-2-carboxamide (960 mg, 0.915 mmol) in MeCN (10 mL) was added 1-(3-chloropropyl)pyrrolidin-2-one (887.1 mg, 5.49 mmol), K2CO3 (1.14 g, 8.23 mmol), Nal (411.4 mg, 2.74 mmol), the reaction was stirred at 80 °C for 24 h. To the reaction was added HzO (20 mL), the aqueous phase was extracted with EtOAc (3 x 20 mL). The combined organic phase was dried over NazSO*, filtered, and concentrated under reduced pressure. The residue was purified by reverse phase chromatography (73→93% MeCN/H 2 O, 10 mM NH4HCO3) to afford product (80 mg, 7.5% yield). LCMS (ESI) m/z. [M + H] calcd for CesHgeNeOeSi: 1174.7; found 1174.7.

Step 4: Synthesis of (2fl)-N-(2-(((2S)-1-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-

6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)-N- methyl-1 -(3-(2-oxopyrrolidin-1-yl)propyl)aziridine-2-carboxamide

To a solution of (2fl)-N-(2-(((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridine-3- yl)-

10.10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)amino)-2-oxoethyl)-N- methyl-1 -(3-(2-oxopyrrolidin-1 -yl)propyl)aziridine-2-carboxamide (80 mg, 0.068 mmol) in THE (1 mL) was added TBAF (1 M, 0.082 mL). The reaction was stirred for 1 h and then was added to H 2 O (10 mL), the aqueous phase was extracted with EtOAc (3 x 10 mL). The combined organic phase was dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The resulting residue was purified by reverse phase chromatography (25→65% MeCN/H 2 O, 10 mM NH4HCO3 to afford the desired product (42 mg, 60.4% yield). LCMS (ESI) m/z : [M + H] calcd for CseHyeNgOg: 1018.6; found 1018.5. The following table of compounds (Table 4) were prepared using the aforementioned methods or variations thereof, as is known to those of skill in the art.

Table 4: Exemplary Compounds Prepared by Methods of the Present Invention

Molecular Observed MW

Ex# Calculated MW

Formula LCMS (ESI) m/z Molecular Observed MW

Ex# Calculated MW

Formula LCMS (ESI) m/z Molecular Observed MW

Ex# Calculated MW

Formula LCMS ESI) m/z

59 CsHeeNeOe [M + H] = 931.51 [M + H] = 931.8

60 C46H59N7O7 [M + H] = 822.46 [M + H] = 822.7

61A CSSHBBNBOB [M + H] = 969.52 [M + H] = 969.6

61B CSSHBBNBOB [M + H] = 969.52 [M + H] = 969.6

62A CSBHBBNBOB [M + H] = 955.51 [M + H] = 955.5

62B CSSHBBNBOB [M + H] = 955.51 [M + H] = 955.5

82B CSAHBBNBOB [M + H] = 923.52 [M + H] = 923.6

83 CSIHB+NIOOB [M + H] = 945.50 [M + H] = 945.6

84 C51 ΗΒΛΝΙΟΟΒ [M + H] = 945.50 [M + H] = 945.6

85 CSI ΗββΝβΟθ [M + H] = 937.52 [M + H] = 837.6 Molecular Observed MW

Ex# Calculated MW

Formula LCMS ESI) m/z

86 CsgHeeNeOe [M + H] = 933.52 [M + H] = 933.7

87 CsiHtoNeO? [M + H] = 899.48 [M + H] = 899.5

88 CsgHeeNeOe [M + H] = 933.52 [M + H] = 933.6

89 CsaHeeNeOe [M + H] = 933.52 [M + H] = 933.6

90 CsgHyoNeOg [M + H] = 951.53 [M + H] = 951.6

91 C49HeeNe09S [M + H] = 943.48 [M + H] = 943.6

92 C49H66N6O9S [M + H] = 943.48 [M + H] = 943.5

93 CssHeeNeOg [M + H] = 985.52 [M + H] = 985.7

94 CKHMNBOIO [M + H] = 963.50 [M + H] = 963.6

95 C51 ΗββΝβΟβ [M + H] = 919.51 [M + H] = 919.5

96 CssHeeNeOg [M + H] = 985.52 [M + H] = 985.7

97 CssHeeNeOg [M + H] = 985.52 [M + H] = 985.7

98 C4eHseNe09 [M + H] = 891.44 [M + H] = 891.6

99 C46H59N7O7 [M + H] = 822.46 [M + H] = 822.6

100 C46H59N7O7 [M + H] = 822.46 [M + H] = 822.6

101 C55H74N6O9S [M + Na] = 1045.52 [M + Na] = 1045.7

102 CsoHeeNeOe [M + H] = 907.51 [M + H] = 907.7

103 CsiHeeNeOe [M + H] = 919.51 [M + H] = 919.5

104 C51 HeeNeOe [M + H] = 919.51 [M + H] = 919.7

105 C49He4NeOe [M + H] = 893.49 [M + H] = 893.7

106 CstHeeNeO? [M + H] = 903.51 [M + H] = 930.7

107 C49H64N8O7 [M + H] = 877.50 [M + H] = 877.7

108 CsaH/oNeOgS [M + H] = 983.51 [M + H] = 983.8

109 CsiH64Ne08 [M + H] = 917.49 [M + H] = 917.7

110 Cs4H6eNeOe [M + H] = 955.51 [M + H] = 955.7

111 CszHeeNeOio [M + H] = 963.50 [M + H = 963.8

112 C46H59N7O7 [M + H] = 822.46 [M + H] = 822.6

113 CsaHeeNeOe [M + Na] = 953.49 [M + Na] = 953.6

114 CsoHeeNeOe [M + H] = 907.51 [M + H = 907.7

115 CsiHeiNeOe [M + H] = 917.49 [M + H = 917.6

116 Cs4HeeNeOe [M + H] = 955.51 [M + H] = 955.7

117 CsoHeeNeOgS [M + H] = 955.48 [M + H = 955.6

118 CsoHeeNeOgS [M + H] = 955.48 [M + H] = 955.8

119 CsgHyoNeOgS [M + H] = 983.51 [M + H] = 983.7

120 ΟίβΗκΝβΟβ [M + H] = 879.48 [M + H] = 880.2

121 Csi HeeNeOe [M + H] = 889.53 [M + H] = 889.6

122 C51 HeeNeOe [M + H] = 889.53 [M + H] = 889.6

123 CssHyoNeOe [M + H] = 939.55 [M + H] = 939.6 Molecular Observed MW

Ex# Calculated MW

Formula LCMS ESI) m/z

124 CsaHyoNeOe [M + H] = 915.55 [M + H] = 915.6

125 CwH y aNeOe [M + H] = 929.57 [M + H] = 929.6

126 CsyHyoNeOe [M + H] = 963.55 [M + H] = 963.6

127 CsaHroNgOe [M + H] = 942.49 [M + H] = 942.4

128 CssHyoNeOs [M + H] = 971.54 [M + H] = 971.5

129 ΟοΗκΝβΟβ [M + H] = 891.48 [M + H] = 891.4

130 ΟΑΒΗΒΣΝΒΟΒ [M + H] = 891.48 [M + H] = 891.5

131 C52H63N7O7 [M + H] = 898.49 [M + H] = 898.4

132 C52H63N7O7 [M + H] = 910.46 [M + H] = 910.4

133 CsaHeeNeOe [M + H] = 933.52 [M + H] = 933.6

134 CffiHeeNeOe [M + H] = 919.51 [M + H] = 919.5

135 CKHMNBOB [M + H] = 933.52 [M + H] = 933.5

136 CsaHeeNeOe [M + H] = 933.52 [M + H] = 933.6

137 CsaH y aNeOg [M + H] = 965.55 [M + H] = 966.2

138 CsaHeeNeO? [M + H] = 917.53 [M + H] = 918.2

139 CsaHeeNeOy [M + H] = 917.53 [M + H] = 917.4

140 CteHeaNeOe [M + H] = 879.48 [M + H] = 879.4

141 CSIHBBNBOB [M + H] = 919.51 [M + H = 919.1

142 [M + H] = 935.54 [M + H] = 936.3

143 [M + H] = 965.52 [M + H] = 966.5

144 [M + H] = 948.53 [M + H = 948.5

145 [M + H] = 898.49 [M + H = 898.5

146 [M + H] = 931.51 [M + H] = 931.5

147 [M + H] = 879.48 [M + H = 879.5

148 [M + H] = 934.53 [M + H] = 936.3

149 [M + H] = 931.51 [M + H] = 931.5

150 [M + H] = 893.49 [M + H] = 893.5

151 CSIHBBNBOB [M + H] = 919.51 [M + H = 919.5

152 CSIHBBNBOB [M + H] = 919.51 [M + H] = 919.5

153 [M + H] = 933.52 [M + H] = 934.5

154 [M + H] = 933.52 [M + H = 934.5

155 e [M + H] = 1001.53 [M + H] = 1001.9

156 [M + H] = 1013.55 [M + H] = 1013.9

157 [M + H] = 905.49 [M + H] = 905.5

158 [M + H] = 905.49 [M + H] = 905.6

159 [M + H] = 951.53 [M + H] = 951.6

160 [M + H] = 951.53 [M + H] = 951.6

161 CSIHBBNBOB [M + H] = 937.52 [M + H] = 937.6 Molecular Observed MW

Ex# Calculated MW

Formula LCMS (ESI) m/z

162 CsiHeeNeOg [M + H = 937.52 [M + H] = 937.6

163 CsaHeeNeOe [M + H = 931.51 [M + H] = 931.5

164 ΟκΗββΝβΟβ [M + H = 931.51 [M + H] = 931.5

165 CsaHeeNeOe [M + H = 933.53 [M + H] = 933.5

166 CsHeeNeOe [M + H = 931.51 [M + H] = 931.5

167 Csi ΗββΝβΟβ [M + H = 919.51 [M + H] = 919.7

168 C48H64N10O7S [M + H = 925.48 [M + H] = 925.30

169 C48H64N10O7S [M + H = 925.48 [M + H] = 925.30

170 C54H7oNeOs [M + H = 959.54 [M + H] = 959.40

171 CsiHesFNeOe [M + H] = 937.50 [M + H] = 937.3

172 C51 HeeNeOs [M + H = 919.51 [M + H] = 919.3

173 CsaHegNeOs [M + H = 948.54 [M + H] = 948.5

174 CstHeeNeOe [M + H] = 957.53 [M + H] = 957.3

175 CwHeeNeOe [M + H] = 957.53 [M + H] = 957.3

176 CsaHeeNeO? [M + H = 917.53 [M + H] = 918.1

177 C51H67N9O8 [M + H] = 934.52 [M + H] = 934.3

178 CstHeeNeOe [M - H = 917.49 [M - H] = 917.6

179 C51 ΗββΝβΟβ [M - H = 917.49 [M - H] = 917.7

180 CssHeeNeOe [M + H = 945.53 [M + H] = 945.6

181 CssHeeNeOe [M + H = 943.51 [M + H] = 943.6

182 CstHeeNeOe [M + H = 919.51 [M + H] = 919.8

183 CsiHesNgOg [M + H = 918.50 [M + H] = 948.6

184 CsiHesFNeOe [M + H = 937.50 [M + H] = 937.3

185 CsiHesFNeOe [M + H = 937.50 [M + H] = 937.2

186 CstHeeNeOe [M + H = 919.51 [M + H] = 919.5

187 CszHeaNgOe [M + H = 942.49 [M + H] = 942.3

188 CsoHesNgOe [M + H = 920.51 [M + H] = 920.3

189 CsoHesNgOe [M + H = 920.51 [M + H] = 920.3

190 CsaHegNgOe [M + H = 948.54 [M + H] = 948.5

191 CsaHeeNeOe [M + H = 945.53 [M + H] = 945.5

192 CsaHeeNeOe [M + H = 945.53 [M + H] = 945.4

193 CsgHeeNeO? [M + H = 917.53 [M + H] = 918.0

194 CsiHesNgOg [M + HzO] = 965.50 [M + H] = 966.4

195 CsaHeeNeOs [M + H = 945.53 [M + H] = 945.5

196 CssHeeNeOe [M + H = 945.53 [M + H] = 945.4

197 C51H67N9O8 [M + H = 934.52 [M + H] = 934.5

198 Cs4H7oNeOe [M + H = 959.54 [M + H] = 959.3

199 CsaHeeNeOs [M + H = 945.53 [M + H] = 945.4 Molecular Observed MW

Ex# Calculated MW

Formula LCMS ESI) m/z

200 CsaHeeNeOs [M + H] = 945.53 [M + H] = 945.5

201 C53H72N8O9 [M + H] = 965.55 [M + H] = 965.4

202 CssHeeNeOs [M + H] = 945.53 [M + H] = 945.8

203 CsiHezNeOe [M + H] = 934.52 [M + H] = 934.8

204 CsiHegNgOs [M + H] = 936.54 [M + H] = 936.8

205 C51H65N9O9 [M + H] = 948.50 [M + H] = 948.5

206 CffiHeeNeOs [M + H] = 933.53 [M + H] = 933.7

207 CsaHeeNeOe [M + H] = 933.53 [M + H] = 933.5

208 ΟκΗββΝβΟβ [M + H] = 933.53 [M + H] = 933.7

209 CsehbeNioOe [M + H] = 1017.59 [M + H] = 1017.6

210 C51H67N9O7S [M + H] = 950.50 [M + H] = 950.2

211 C51H67N7O8 [M + H] = 906.52 [M + H] = 906.4

212 CsoHeiNgOeS [M + H] = 948.45 [M + H] = 948.1

213 CAgHeaNeOe [M + H] = 891.48 [M + H] = 891.4

214 Cs7H72NeOe [M + H] = 997.56 [M + H] = 997.2

215 C57H72N8O8 [M + H] = 997.56 [M + H] = 997.2

216 C4BH66N10O7S [M + H] = 927.49 [M + H] = 927.5

217 C48H66N10O7S [M + H] = 927.49 [M + H] = 927.4

218 C48H64N10O7S [M + H] = 925.48 [M + H] = 925.4

219 C48H64N10O7S [M + H] = 925.48 [M + H] = 925.1

220 C4eHe4Nio07S [M + H] = 925.48 [M + H] = 925.4

221 θ4βΗβ4Νιοθ78 [M + H] = 925.48 [M + H] = 925.4

222 C49H66N10O7S [M + H] = 939.49 [M + H] = 939.5

223 C49H66N10O7S [M + H] = 939.49 [M + H] = 939.5

224 C49H66N10O7S [M + H] = 939.49 [M + H] = 940.0

225 C49H66N10O7S [M + H] = 939.49 [M + H] = 939.2

226 C49H66N10O7S [M + H] = 939.49 [M + H] = 939.1

227 C49H66N10O7S [M + H] = 939.49 [M + H] = 939.9

228 C49H66N10O7S [M + H] = 939.49 [M + H] = 940.0

229 C49H66N10O7S [M + H] = 939.49 [M + H] = 939.1

230 C57H77N9O8 [M + H] = 1016.60 [M + H] = 1016.6

231 CsiH64Ne09 [M + H] = 933.49 [M + H] = 933.1

232 C51 ΗΒΛΝΒΟΘ [M + H] = 933.49 [M + H] = 933.2

233 CssHeeFgNeO? [M + H] = 991.53 [M + H] = 992.0

234 C54H69N9O7S [M + H] = 988.51 [M + H] = 988.1

235 C49H66N10O7S [M + H] = 939.49 [M + H] = 939.5

236 ΟδΣΗββΝβΟβ [M + H] = 933.53 [M + H] = 933.5

237 ΟδΣΗββΝβΟβ [M + H] = 933.53 [M + H] = 933.5 Molecular Observed MW

Ex# Calculated MW

Formula LCMS ESI) m/z

238 CsaHeeNeOe [M + H] = 933.53 [M + H] = 933.5

239 CsaHeeNeOe [M + H] = 933.53 [M + H] = 933.5

240 CsaHeeNeOe [M + H] = 933.53 [M + H] = 933.5

241 CsaHeeNeOe [M + H] = 933.53 [M + H] = 933.5

242 CsaHeeNeOe [M + H] = 933.53 [M + H] = 933.5

243 CsaHeeNeOe [M + H] = 933.53 [M + H] = 933.5

244 CsaHeeNeOe [M + H] = 931.51 [M + H] = 931.5

245 CstHyoNeOe [M + H] = 959.54 [M + H] = 959.5

246 CstHyoNeOe [M + H] = 959.54 [M + H] = 959.5

247 CsaH y oNeOe [M + H] = 947.54 [M + H] = 947.4

248 CsaHeeNeOg [M + H] = 949.52 [M + H] = 949.5

249 CsaHeeNeOg [M + H] = 949.52 [M + H] = 949.5

250 CsaHgyNgOe [M + H] = 946.52 [M + H] = 946.5

251 CszHeyNgOe [M + H] = 946.52 [M + H] = 946.5

252 CsaHegNgOe [M + H] = 948.54 [M + H] = 948.5

253 CsaHegNgOe [M + H] = 948.54 [M + H] = 948.5

254 CsaHegNgOe [M + H] = 948.54 [M + H] = 948.5

255 CsaHegNgOe [M + H] = 948.54 [M + H] = 948.5

256 CsoHeeNgOe [M + H] = 907.51 [M + H] = 907.5

257 CsoHeeNgOe [M + H] = 907.51 [M + H] = 907.5

258 CsoHeaFaNeOe [M + H] = 961.48 [M + H] = 961.5

259 CsoHeaFaNeOe [M + H] = 961.48 [M + H] = 961.5

260 CsoHeaFaNeOe [M + H] = 961.48 [M + H] = 961.5

261 CsoHeaFaNeOe [M + H] = 961.48 [M + H] = 961.5

262 CstHeeNeOe [M + H] = 921.53 [M + H] = 921.5

263 CsiHeeNeOe [M + H] = 921.53 [M + H] = 921.5

264 CsaHyoNeOe [M + H] = 935.54 [M + H] = 935.3

265 CsaHyoNeOe [M + H] = 935.54 [M + H] = 935.3

266 Cs4H72NeOio [M + H] = 993.55 [M + H] = 993.5

267 C54H72N8O10 [M + H] = 993.55 [M + H] = 993.5

268 CseHysNgOg [M + H] = 1018.58 [M + H] = 1018.6

269 CseHysNgOg [M + H] = 1018.58 [M + H] = 1018.5

270 CsaHyiNgOg [M + H] = 978.55 [M + H] = 978.5

271 CsaHyiNgOg [M + H] = 978.55 [M + H] = 978.5

272 CssH y aNgOio [M + H] = 1020.56 [M + H] = 1020.5

273 CsaHegNgOe [M + H] = 960.54 [M + H] = 960.5

274 CsaHegNgOe [M + H] = 960.54 [M + H] = 960.5

275 CsaHeeNeOg [M + H] = 949.52 [M + H] = 949.3 Molecular Observed MW

Ex# Calculated MW

Formula LCMS ESI) m/z

276 CtoHeeNeOg [M + H] = 949.52 [M + H] = 949.3

277 CsahboNeOe [M + H] = 963.54 [M + H] = 963.5

278 CsaHyoNeOg [M + H] = 963.54 [M + H] = 963.5

279 C54H72NBO9 [M + H] = 977.55 [M + H] = 977.5

280 C54H72N8O9 [M + H] = 977.55 [M + H] = 977.6

281 CsaHeeNeOe [M + H] = 949.52 [M + H] = 949.3

282 CffiHeeNeOg [M + H] = 949.52 [M + H] = 949.3

283 CsaHeeNeOg [M + H] = 949.52 [M + H] = 949.5

284 ΟΚΗΜΝΒΟΒ [M + H] = 949.52 [M + H] = 949.5

285 CsaHeeNeOe [M + H] = 933.53 [M + H] = 933.5

286 CABHBBNTO? [M + H] = 862.49 [M + H] = 862.4

287 CsshfooNeOs [M + H] = 947.54 [M + H] = 947.5

288 CMHZONBOB [M + H] = 947.54 [M + H] = 947.5

289 CsoHeeNioOyS [M + H] = 953.51 [M + H] = 953.5

290 CsoHeeNioOyS [M + H] = 953.51 [M + H] = 953.4

291 CsoHeeNioOzS [M + H] = 953.51 [M + H] = 953.5

292 CsoHeeNioOyS [M + H] = 953.51 [M + H] = 953.5

293 C49H66N10O7S [M + H] = 939.49 [M + H] = 939.5

294 C49H66N10O7S [M + H] = 939.49 [M + H] = 939.5

295 C50H68N10O7S [M + H] = 953.51 [M + H] = 953.5

296 C49H66N10O7S [M + H] = 939.49 [M + H] = 939.4

297 C49H66N10O7S [M + H] = 939.49 [M + H] = 939.5

298 C49H66N10O7S [M + H] = 939.49 [M + H] = 939.5

299 C49H66N10O7S [M + H] = 939.49 [M + H] = 939.5

300 C49H66N10O7S [M + H] = 939.49 [M + H] = 939.4

301 C49H66N10O7S [M + H] = 939.49 [M + H] = 939.4

302 C49H66N10O7S [M + H] = 939.49 [M + H] = 939.4

303 C50H66N10O7S [M + H] = 951.49 [M + H] = 951.5

304 CsoHeeNio07S [M + H] = 951.49 [M + H] = 951.5

305 C55H7ONBOB [M + H] = 971.54 [M + H] = 971.1

306 CsaHeeNeOe [M + H] = 931.51 [M + H] = 931.2

307 CSBHBBNBOB [M + H] = 931.51 [M + H] = 931.2

308 CsshtaNgOe [M + H] = 984.54 [M + H] = 984.5

309 C53H70N8O8 [M + H] = 947.54 [M + H] = 947.5

310 C49HBBNIO07S [M + H] = 939.49 [M + H] = 939.5

311 CSIHBSFNBOB [M + H] = 937.50 [M + H] = 937.2

312 C49HB3N70B [M + H] = 878.48 [M + H] = 878.4

313 C57H72N8O7 [M + H] = 981.56 [M + H] = 981.5 Molecular Observed MW

Ex# Calculated MW

Formula LCMS (ESI) m/z

314 Cs+HraFNeOe [M + H] = 977.53 [M + H] = 977.5

315 CstHtoFNeOe [M + H] = 977.53 [M + H] = 977.5

316 CssHegNgOe (M + H] = 984.54 [M + H] = 984.5

317 CsehbeNeOg [M + H] = 1031.60 [M + H] = 1031.5

318 CseHyaFaNeOy [M + H] = 1031.56 [M + H] = 1031.5

319 CseHreFaNeO? [M + H] = 948.48 [M + H] = 948.4

320 CsiHesNgOyS (M + H] = 993.54 [M + H] = 993.5

321 CsshboFaNeO? [M + H] = 1013.57 [M + H] = 1013.5

322 CsshboFaNeOy [M + H] = 989.55 [M + H] = 989.5

323 CseHyaFNeO? [M + H] = 989.55 [M + H] = 989.5

324 CseHyaFNeO? [M + H] = 1086.60 [M + H] = 1086.6

325 CssHyaNeOg [M + H] = 1057.63 [M + H] = 1057.4

326 CssHyaNeOg [M + H] = 967.52 [M + H] = 967.5

327 C 59 H7 9 N11O7S [M + H] = 973.56 [M + H] = 973.4

328 C59H79N11O7S [M + H] = 973.56 [M + H] = 973.5

329 C54H69N7O11 [M + H] = 992.52 [M + H] = 992.4

330 CstHesNyOg [M + H] = 920.49 [M + H] = 920.6

331 C57H69N7O9 [M + H] = 996.53 [M + H] = 996.6

332 C52H67N7O9 [M + H] = 934.51 [M + H] = 934.6

Biological Assays

Compounds 1 -2,4-18A, 19A-19B, 21A-24A, 27-32A, 33-43A, 44-45, 47B-54, 56-59, 68A, 69A, 71 B, 72A, 73-78, 79B-82A, 83-97, 100-110, 112-117, 119-234, 236-294, and 297-332 exhibited: a) a % cross-linking to KRAS G12D of greater than zero within a 24-hour incubation timeframe in the assay described below; and/or b) an IC50 of 2 μΜ or less in the KRAS^^-B-Raf (AsPC-1 ) disruption assay described below.

Potency assay: pERK

The purpose of this assay is to measure the ability of test compounds to inhibit K-Ras in cells. Activated K-Ras induces increased phosphorylation of ERK at Threonine 202 and Tyrosine 204 (pERK). This procedure measures a decrease in cellular pERK in response to test compounds. The procedure described below in NCI-H358 cells is applicable to K-Ras G12C.

Note: This protocol may be executed substituting other cell lines to characterize inhibitors of other RAS variants, including, for example, AsPC- 1 (K-Ras G 12D), Capan- 1 (K-Ras G12V), or NCI- H1355 (K-Ras G13C).

NCI-H358 cells were grown and maintained using media and procedures recommended by the ATCC. On the day prior to compound addition, cells were plated in 384-well cell culture plates (40 μΙ/well) and grown overnight in a 37°C, 5% C02 incubator. Test compounds were prepared in 10, 3-fold dilutions in DMSO, with a high concentration of 10 mM. On the day of assay, 40 nL of test compound was added to each well of cell culture plate using an Echo550 liquid handler (LabCyte®). Concentrations of test compound were tested in duplicate. After compound addition, cells were incubated 4 hours at 37°C, 5% C02. Following incubation, culture medium was removed and cells were washed once with phosphate buffered saline.

In some experiments, cellular pERK level was determined using the AlphaLISA SureFire Ultra p- ERK1/2 Assay Kit (PerkinElmer). Cells were lysed in 25 μΙ_ lysis buffer, with shaking at 600 RPM at room temperature. Lysate (10 pL) was transferred to a 384-well Opti-plate (PerkinElmer) and 5 pL acceptor mix was added. After a 2-hour incubation in the dark, 5 pL donor mix was added, plate was sealed, and incubated 2 hours at room temperature. Signal was read on an Envision plate reader (PerkinElmer) using standard AlphaLISA settings. Analysis of raw data was carried out in Excel (Microsoft) and Prism (GraphPad). Signal was plotted vs. the decadal logarithm of compound concentration, and ICso was determined by fitting a 4-parameter sigmoidal concentration response model.

In other experiments, cellular pERK was determined by In-Cell Western. Following compound treatment, cells were washed twice with 200 pL tris buffered saline (TBS) and fixed for 15 minutes with 150 pL 4% paraformaldehyde in TBS. Fixed cells were washed 4 times for 5 minutes with TBS containing

0.1% Triton X-100 (TBST) and then blocked with 100 pL Odyssey blocking buffer (LI-COR) for 60 minutes at room temperature. Primary antibody (pERK, CST-4370, Cell Signaling Technology) was diluted 1 :200 in blocking buffer, and 50 pL was added to each well and incubated overnight at 4°C. Cells were washed 4 times for 5 minutes with TBST. Secondary antibody (IR-800CW rabbit, LI-COR, diluted 1 :800) and DNA stain DRAQ5 (LI-COR, diluted 1 :2000) were added and incubated 1-2 hours at room temperature. Cells were washed 4 times for 5 minutes with TBST. Plates were scanned on a Li-COR Odyssey CLx Imager. Analysis of raw data was carried out in Excel (Microsoft) and Prism (GraphPad). Signal was plotted vs. the decadal logarithm of compound concentration, and ICso was determined by fitting a 4-parameter sigmoidal concentration response model.

The following compounds exhibited a pERK EC50 of under 5 uM (AsPC-1 KRAS G12D):

179,157,178,327,205,106,242,121 ,183,36,158,196,84,17AandB, 87,187,114,182,255,254,185,236,124,19 7,1 ,107,192,34,118,296,78,89,104,74,306,310,105,152,269,229,221 ,294,117,119,240,151 ,193,86,245,12 8, 163, 272, 270, 79AandB, 232, 140, 138, 293, 38, 94, 110, 172, 271 , 246, 72AandB, 108, 35, 14, 127, 7, 153, 39, 190, 96,227,13,77,286,215,244,184,284,275,147,295,204,50,161 ,129,176,51 ,290,226,218,164,282,167,162,1 31 ,228,292,233,308,304,48,9,113,298,277,54,57,219,173,220,268, 49,149,247,120,154,307,56,166,11 ,53

,101 ,10,8,238,97,303,132,186,52,297,93,85,83,280,103,200,276,278 ,144,165,199,33,139,112,224,177,2 41 ,273,237,274,191 ,243,319,320,225,59,311 ,207,239,279,160,289,171 ,156, 92, 202, 43A, 266, 208, 281 ,15 9,300,210,223,217,283,216,231 ,299,90,91 ,267,155,259,291 ,258,257,262,222,137,100,256,88,316,142,3 18,146,198,288,302,174,265,322,12,168.42A.201 ,301 ,263,248,287,58,305,260,134,169,313,314,323,23 4,136,148,102,315,141 ,150,309,326,261 ,321 ,175,230,249,264,95,285,135,133,170,317,328,214,209,324

,325.

Determination of Cell Viability In RAS Mutant Cancer Cell Lines

Protocol: CellTiter-Glo® Cell Viability Assay

Note - The following protocol describes a procedure for monitoring cell viability of K-Ras mutant cancer cell lines in response to a compound of the invention. Other RAS isoforms may be employed, though the number of cells to be seeded will vary based on cell line used. The purpose of this cellular assay was to determine the effects of test compounds on the proliferation of three human cancer cell lines (NCI-H358 (K-Ras G12C), AsPC-1 (K-Ras G12D), and Capan-1 (K-Ras G12V)) over a 5-day treatment period by quantifying the amount of ATP present at endpoint using the CellTiter-Glo® 2.0 Reagent (Promega).

Cells were seeded at 250 cells/well in 40 pL of growth medium in 384-well assay plates and incubated overnight in a humidified atmosphere of 5% CO2 at 37°C. On the day of the assay, 10 mM stock solutions of test compounds were first diluted into 3 mM solutions with 100% DMSO. Well-mixed compound solutions (15 pL) were transferred to the next wells containing 30 pL of 100% DMSO, and repeated until a 9-concentration 3-fold serial dilution was made (starting assay concentration of 10 pM). Test compounds (132.5 nL) were directly dispensed into the assay plates containing cells. The plates were shaken for 15 seconds at 300 rpm, centrifuged, and incubated in a humidified atmosphere of 5% CO2 at 37 °C for 5 days. On day 5, assay plates and their contents were equilibrated to room temperature for approximately 30 minutes. CellTiter-Glo® 2.0 Reagent (25 pL) was added, and plate contents were mixed for 2 minutes on an orbital shaker before incubation at room temperature for 10 minutes. Luminescence was measured using the PerkinElmer Enspire. Data were normalized by the following: (Sample signal/Avg. DMSO)*100. The data were fit using a four-parameter logistic fit.

*Key:

+++++: IC50 > 10 uM ++++: 10 uM > IC50 > 1 uM

+++: 1 uM > IC50 ≥ 0.1 uM

++: 0.1 uM > IC50 > 0.01 uM +: IC50 < 0.01 uM

Table 5. H358 Cell Viability assay data (K-Ras G12C, IC50, uM):

IC50* Examples of A compounds

+ 1 ,44,58,59,90,95,106,136,137,142,146,148,150,155,156,159,160, 165,171 ,174,175,186,201 , 207,209,222,231 ,236,241 ,261 ,266,267,273,274,279,280,299,301 ,307,319, 324, 325, 328, 42A,

43A

++ 2,5,7,8,9,10,13,33,41,83,85,100,102,132,133,135,144,168,169, 170,191,208,214,216,217,22 3,224,225,226,227,228,230,234,239,248,249,256,257,258,259,26 0,262,263,264,265,270,27 6,277,283,287,288,289,290,291 ,292,293,296,297,298,300,302,303,316,317,322,323,326,42 B,43B

+++ 3,4,6, 11 ,12,14,40,45,46,48,50,52,56,63,77,103,107,141 ,202,210,243,285,294,304,313,318,3 20,321

++++ 26,29,30,31 ,49,60,66,96,15 A and B,16 A and B,16 A and B,17 A and B,17 A and B,18 A and B,18 A and B,19 A and B,19 A and B,20 A and B,20 A and B,22 A and B,23 A and B,24 A and B,24 A and B,32 A and B,32 A and B,47 A and B,62 A and B,62 A and B,68 A and B,68 A and B,69 A and B,69 A and B

+++++ 15 A and B, 25, 27, 28, 47 A and B,67 Table 6. AsPC-1 Cell Viability assay data (K-Ras G12D, IC50, uM):

IC50* Examples of A compounds

+ 209,325

++ 95,133,170,175,214,230,249,285,309,313,317,321 ,324

+++ 7.12.58.59.77.88.90.100.102.103.135.136.137.141.142.144.146. 148.150.155.156.159.160.1

68.169.171.174.186.198.201.207.210.216.217.222.223.224.22 5.228.231.234.239.241.243.2

48.256.257.258.259.260.261.262.263.264.265.266.267.270.27 3.274.277.279.281.283.287.2

88.289.291.297.298.299.300.301.302.303.305.307.311 .314.315.318.319.320.323.326.328.1 9 A and B,42A,43A

++++ 1 ,8,9,10,11 ,13,14,29,34,35,38,39,44,48,49,52,53,54,56,57,60,63,73,74,75 ,76,78,80,81 ,83,84 ,85,86,87,91 ,92,96,97,101 ,104,105,106,107,108,110,112,113,114,117,119,120,121 ,122,128, 129,132,134,139,140,147,149,151 ,152,153,154,157,158,161 ,162,163,164,165,166,167,172,

173,176,177,184,185,187,190,191 ,192,193,196,197,199,200,202,204,208,215,218,219,220, 221 ,226,227,229,232,233,236,237,238,240,242,244,245,246,247,255 ,268,269,272,275,276, 278,280,282,284,286,290,292,293,294,295,296,304,306,308,310, 316,322,327,16 A and B,16 A and B,17 A and B,17 A and B,18 A and B,18 A and B,22 A and B,23 A and B,32 A and B,42B,61 A and B,61 A and B,62 A and B,62 A and B,64 A and B,65 A and B,65 A and B,68 A and B,69 A and B,69 A and B,70 A and B,71 A and B,72 A and B,72 A and B,79 A and B,79 A and B,82 A and B,82 A and B

+++++ 2,3,4,5,6,28,31 ,36,37,40,41 ,45,46,50,51 ,55,66,67,89,93,94,98,99,109,111 ,115,116,118,126, 127,130,131 ,138,143,145,178,179,180,181 ,182,183,188,189,194,195,203,205,206,211 ,212, 213,235,250,251 ,252,253,254,271 ,312,15 A and B,15 A and B,20 A and B,20 A and B,24 A and B,24 A and B,32 A and B,43B,47 A and B,47 A and B,64 A and B,70 A and B,71 A and

B

Disruption of B-Raf Ras-blndlng Domain (BRAF RBD ) Interaction with K-Ras by Compounds of the Invention (also called a FRET assay or an MOA assay)

Note - The following protocol describes a procedure for monitoring disruption of K-Ras G12C (GMP-PNP) binding to BRAF™ 30 by a compound of the invention. This protocol may also be executed substituting other Ras proteins or nucleotides, such as K-Ras G12D and K-Ras G13D.

The purpose of this biochemical assay was to measure the ability of test compounds to facilitate ternary complex formation between a nucleotide-loaded K-Ras isoform and Cyclophilin A; the resulting ternary complex disrupts binding to a BRAF* 80 construct, inhibiting K-Ras signaling through a RAF effector. Data is reported as IC50 values.

In assay buffer containing 25 mM HEPES pH 7.3, 0.002% Tween20, 0.1% BSA, 100 mM NaCI and 5 mM MgClz, tag less Cyclophilin A, His6-K-Ras-GMPPNP, and GST-BRAF RBD were combined in a 384-well assay plate at final concentrations of 25 μΜ, 12.5 nM, and 50 nM, respectively. Compound was present in plate wells as a 10-point 3-fold dilution series starting at a final concentration of 30 μΜ. After incubation at 25 °C for 3 hours, a mixture of anti-His Eu-W1024 and anti-GST allophycocyanin was then added to assay sample wells at final concentrations of 10 nM and 50 nM, respectively, and the reaction incubated for an additional 1.5 hours. TR-FRET signal was read on a microplate reader (Ex 320 nm, Em 665/615 nm). Compounds that facilitate disruption of a K-Ras:RAF complex were identified as those eliciting a decrease in the TR-FRET ratio relative to DMSO control wells.

Ras-Raf disruption/FRET/MOA assay data (IC50, uM): 'Key:

+++++: IC50 > 10 uM ++++: 10 uM > IC50 > 1 uM

+++: 1 uM > IC50 > 0.1 uM

++: 0.1 uM > IC50 > 0.01 uM +: IC50 < 0.01 uM

Table ?. KRAS G13D FRET data

IC50* Examples of A compounds

+ None

++ 12.58.83.85.86.88.89.95.100.109.113.115.120.129.130.133.135. 136.137.138.139.140.143.1

48.149.150.157.158.162.167.170.171.172.173.174.175.176.17 7.182.186.193.194.195.196.1 97,198,204,209,230,231 ,239,241 ,248,249,256,257,262,263,264,265,274,276,285,286,288,3 06, 307, 309, 325, 329, 330, 332, 258 * ,259*, 260*, 261 *,281 *,282 * , 283*, 284*, 314*.315*, 316*,42A,

43A

+++ 1 ,2, 3, 4, 5, 7, 8, 9,11 ,13,14,33,34,35,36,37,38,39,40,41 ,44,45,48,49,50,51 ,52,53,54,56,57,59,84 ,87,90,91 ,92,93,94,96,97,101 ,102,103,104,105,108,110,111 ,112,116,117,118,119,127,128,1

31.134.141.142.144.145.146.147.151.152.153.155.156.159.16 0.161.163.164.165.166.168.1

69.178.179.180.181.183.184.185.187.188.189.190.191 .199.200.201.202.203.205.206.207.2 08,212,213,214,215,216,217,218,219,220,221 ,222,223,224,225,226,227,228,229,232,236,2

37.238.242.243.244.245.246.247.252.253.254.255.266.267.26 8.269.270.271.272.273.275.2

77.278.279.280.287.289.291.293.294.297.298.299.300.301.30 2.303.305.308.311.317.322.3 23,326,328,331 ,47 A and B,69 A and B

++++ 6,10,25,26,27,28,29,30,31 ,46,55,66,67,73,74,98,106,107,114,126,132,154,192,210,211 ,233, 235,240,250,251 ,290,292,295,296,304,310,312,313,318,319,320,321 ,324,15 A and B,17 A and B,18 A and B,19 A and B,24 A and B,32 A and B,32 A and B,42B,43B,47 A and B,62 A and B,72 A and B

+++++ 60,63,75,76,77,78,80,81 ,99,121 ,122,123,124,125,234,327,15 A and B,16 A and B,16 A and B,17 A and B,18 A and B,19 A and B,20 A and B,20 A and B,22 A and B,23 A and B,24 A and B,61 A and B,61 A and B,62 A and B,64 A and B,64 A and B,65 A and B,65 A and B,68 A and B,68 A and B,69 A and B,70 A and B,70 A and B,71 A and B,71 A and B,72 A and B,79 A and B,79 A and B,82 A and B,82 A and B

Table 8. KRAS G12S FRET data

IC50* Examples of A compounds

+ 325,264,281 ,135

++ 263,258,230,249,287,282,209,139,315,170,283,267,150, 260, 274, 284, 42A,167,276,261 ,133, 137, 12,256, 136,257,285,288,265,88,85,231 ,316,330,269,262,280, 148, 186,278, 182,248,259, 309,162,317,314,332,160,140,175,305,95,308,158,113,273,323,2 68,173, 322, 306, 43A, 214,1 77,171 ,161 ,146,272,207,329,157,197,201 ,147,103,208,200,328,241 ,172,90,89,105,149,279, 100,159,238,93,271 ,91 ,48,174,7,35,155,307,53,36,52,92,190,156,237,302,326,59,8,14 2,37, 33,11 ,141 ,97,127,10,13,277,58,104,275,222,129,266,204,39,191 ,110,176,301 ,96,331 ,54,19 8,239,130,255,134,83,50,1 ,34,289,86,38,47 A and B,168,143,169,270,298

+++ 14,115,152,109,49,102,223,189,44,291 ,144,297,120,202,153,187,185,188,56,138,195,224,2 54,51 ,178,244,286,194,45,225,181 ,246,9,205,166,101 ,196,252,2,116,243,193,216,253,179, 293,217,192,112,245,311 ,199,203,94,165,3,163,232,151 ,5,294,108,183,46,215,299,119,117 ,4,300,164,303,184,213,227,145,57,128,304,6,220,229,218,226, 228,87,251 ,131 ,84,219,111 , 221 ,242,154,310,247,180,40,47 A and B,41 ,290,118,236,292,31 ,324,296,321 ,250,212,206,55,126

++++ 210,240,77, 121 ,211 ,319,42B,313,132,114,98,320,295,318,43B,235, 122,107,62 A and B, 312,24 A and B,106,74,234,78,66,15 A and B, 233, 73, 69 A and B, 63, 29, 67, 22 A and B,80,23 A and B

+++++ 25,124,32 A and 8,327,27,28,26,62 A and B,72 A and B,20 A and B,79 A and B,19 A and B,16 A and B, 123,24 A and B,30,17 A and B,15 A and B,19 A and B,32 A and B,18 A and B,18 A and B,17 A and B,16 A and B,20 A and B,69 A and B,60,68 A and B,68 A and B,65 A and B,65 A and B,64 A and B,64 A and B,70 A and B,71 A and B,70 A and B,71 A and B,72 A and B,61 A and B,61 A and B,82 A and B,82 A and B,81 ,79 A and B.76,75,99,125

Table 9. KRAS G13C FRET data

IC50* Examples of A compounds

+ 84,85,89,129,135,136,139,140,182,195,205,209,230,231 ,256,257,258,260,282,283,284,285, 325, 329, 330, 42A

++ 3, 4, 7, 8,11 ,12,13,14,34,35,36,37,38,39,41 ,44,45,48,49,50,51 ,53,54,56,57,58,59,77,78,83,86, 87,88,90,91 ,92,93,94,95,96,97,100,102,103,104,105,108,109,112,113,115,1 17,118,119,120, 124,127,128,130,131 ,133,134,137,138,141 ,142,143,144,145,146,147,148,149,150,152,155, 156,157,158,159,160,161 ,162,167,169,170,171 ,172,173,174,175,176,177,178,179,180,181 , 183,184,185,186,187,188,189,190,191 ,193,194,196,197,198,200,201 ,203,204,207,208,212, 213,214,215,232,237,238,239,241 ,245,246,248,249,251 ,252,253,254,255,259,261 ,262,263, 264,265,266,267,268,269,270,271 ,272,273,274,275,276,277,278,279,280,281 ,286,287,288, 297,301 ,305,306,307,308,309,311 ,314,315,316,317,322,323,326,328,331 ,332,43A,47 A and B,47 A and B,69 A and B

+++ 1 ,2,5,6,9,10,31 ,33,40,46,52,55,66,74,76,98,101 ,106,107,110,111 ,114,116,121 ,122,123,125, 126,151 ,153,154,163,164,165,166,168,192,199,202,206,211 ,216,217,218,219,220,221 ,222, 223,224,225,226,227,228,229,233,236,243,244,247,250,289,290, 291 ,292,293,294,296,298, 299,300,302,303,310,312,313,318,319,320,321 ,324,22 A and B,42B,70 A and B

++++ 25,27,28,29,63,67,73,80,132,210,234,235,240,242,295,304,327, 15 A and B,16 A and B,17 A and B,17 A and B,18 A and B,19 A and B,20 A and B,23 A and B,24 A and B,32 A and B,43B,62 A and B,65 A and B,68 A and B,69 A and B,72 A and B,79 A and B +++++ 26,30,60,75,81 ,99,15 A and B,16 A and B,18 A and B.19 A and B,20 A and B,24 A and B,32 A and B,61 A and B,61 A and B,62 A and B,64 A and B,64 A and B,65 A and B,68 A and B,70 A and B,71 A and B,71 A and B,72 A and B,79 A and B,82 A and B,82 A and B

Table 10. KRAS G12V FRET data

IC50* Examples of A compounds

+ 325

++ 1 ,11 ,12,13,36,44,48,50,58,59,83,85,88,90,93,95,96,97,100,103,113 ,133,135,136,137,139,14 0,141 ,146,147,148,149,150,151 ,155,156,157,158,159,160,161 ,162,165,167,170,171 ,172,17 3,174,175,177,182,186,189,190,192,197,200,201 ,204,207,208,209,214,230,231 ,239,241 ,24 8,249,256,257,258,259,260,261 ,262,263,264,265,267,268,269,272,273,274,276,278,279,28 0,281 ,282,283,284,285,287,288,302,305,306,307,309,314,315,316,317 ,322,323,326,328,32 9,330,331 , 332, 42A.43A

+++ 2, 3, 4, 5, 6, 7, 8, 9,10,14,31 ,33,34,35,37,38,39,40,41 ,45,49,51 ,52,53,54,55,56,57,84,86,87,89,91 ,92,94,101 ,102,104,105,108,109,110,111 ,112,115,116,117,118,119,120,127,128,129,130,13 1 ,134,142,143,144,145,152,153,154,163,164,166,168,169,176,178 ,179,180,181 ,183,184,18 5,187,188,191 ,194,195,196,198,199,202,203,205,206,212,213,215,216,217,218 ,219,220,22 1 ,222,223,224,225,226,227,228,229,232,236,237,238,243,244,245 ,246,247,250,251 ,252,25 3,254,255,266,270,271 ,275,277,286,289,290,291 ,292,293,294,297,298,299,300,301 ,303,30 4,308,310,311 ,321 ,324,47 A and B

++++ 29,46,66,67,74,77,78,80,98,106,107,114,121 ,122,123,124,126,132,138,193,210,211 ,233,23

4,235,240,242,295,296,312,313,318,319,320,15 A and B,16 A and B,22 A and B,24 A and B,42B,43B,47 A and B,62 A and B,69 A and B,72 A and B,79 A and B

+++++ 73,25,26,27,28,30,60,63,75,76,81 ,99,125,327,15 A and B.16 A and B,17 A and B,17 A and B,18 A and B,18 A and B,19 A and B,19 A and B,20 A and B,20 A and B,23 A and B,24 A and B,32 A and B,32 A and B,61 A and B,61 A and B,62 A and B,64 A and B,64 A and B,65 A and B,65 A and B,68 A and B,68 A and B,69 A and B,70 A and B,70 A and B,71 A and B,71 A and B,72 A and B,79 A and B,82 A and B,82 A and B

Table 11. KRAS G12D FRET data

IC50* Examples of A compounds

+ 214,305,325

++ 59,88,95,103,113,133,135,136,137,139,140,141 ,146,148,150,167,170,171 ,173,175,186,198, 209,230,231 ,241 ,248,249,256,258,260,261 ,263,264,265,267,269,274,276,278,280,281 ,282, 283,284,285,287,309,315,316,330,42A,43A

+++ 1 ,2, 7, 8,12,13,14,33,34,35,36,37,38,39,40,41 ,44,45,48,49,50,51 ,52,53,54,56,58,73,74,83,85,

86.89.90.91.92.93.96.97.100.101.102.104.105.110.112.115.1 20.127.128.129.130.131.134.1

38.142.143.144.147.149.151.152.153.155.156.157.158.159.16 0.161.162.163.164.165.166.1

68.169.172.174.176.177.178.179.181.182.184.185.187.188.18 9.190.191.192.193.194.196.1

97.199.200.201.202.203.204.207.208.215.216.217.222.223.22 4.225.227.236.237.238.239.2

42.243.244.245.252.253.254.255.257.259.262.266.268.270.27 1 .272.273.275.277.279.286.2 88,289,291 ,293,294,297,298,299,300,301 ,302,303,304,306,307,308,311 ,313,314,317,321 ,3 22,323,324,326,328,329,331 ,332,70 A and B

++++ 16 A and

B, 3,4, 5, 6,9, 10,11 ,29,31 ,46,57,63,66,67,76,77,78,80,84,87,94,98,106,107,108,109,111 ,114,1

16.117.118.119.121.122.123.124.125.126.132.145.154.180.18 3.195.205.206.210.211.212.2

13.218.219.220.221.226.228.229.232.233.234.235.240.246.24 7.250.251.290.292.295.296.3

10,312,318,319,320,327,15 A and B,17 A and B,17 A and B,18 A and B,19 A and B,22 A and B,23 A and B,24 A and B,32 A and B,42B,43B,47 A and B,47 A and B,62 A and B,68 A and B,69 A and B,72 A and B,82 A and B

+++++ 25,26,27,28,30,55,60,75,81 ,99,15 A and B,16 A and B,18 A and B,19 A and B,20 A and B,20 A and B,24 A and B,32 A and B,61 A and B,61 A and B,62 A and B,64 A and B,64 A and B,65 A and B,65 A and B,68 A and B,69 A and B,70 A and B,71 A and B,71 A and B,72 A and B,79 A and B,79 A and B,82 A and B

Table 12. KRAS WT FRET data

IC50* Examples of A compounds

+ 264,325

++ 12,13,35,36,37,44,45,52,53,54,58,59,83,85,86,88,89,90,91 ,92,93,95,96,97,100,102,103,105, 109,110,112,113,115,116,120,129,130,133,134,135,136,137,138, 139,140,141 ,143,146,147, 148,149,150,155,156,157,158,159,160,161 ,162,167,168,169,170,171 ,172,173,174,175,176, 177,182,183,186,189,190,191 ,193,195,196,197,198,201 ,204,205,207,208,209,213,214,222, 223,230,231 ,237,238,239,241 ,245,246,248,249,252,253,256,257,258,259,260,261 ,262,263, 265,267,268,269,271 ,272,273,274,276,278,279,280,281 ,282,283,284,285,287,288,289,291 , 297,298,301 ,302,305,306,307,308,309,314,315,316,317,322,323,326,328,329 ,330,331 ,332, 42A.43A

+++ 1 ,2, 3, 4, 5, 6, 7, 8, 9,10,11 ,14,33,34,38,39,40,41 ,48,49,50,51 ,56,57,84,87,94,98,101 ,104,108,11 1 ,117,118,119,121 ,127,128,131 ,142,144,145,151 ,152,153,154,163,164,165,166,178,179,18 0,181 ,184,185,187,188,192,194,199,200,202,203,206,211 ,212,215,216,217,218,219,220,22 1 ,224,225,226,227,228,229,232,236,240,242,243,244,247,250,251 ,254,255,266,270,275,27 7,286,290,292,293,294,295,296,299,300,303,304,310,311 ,312,318,321 ,324,47 A and B

++++ 25,26,27,29,30,31 ,46,55,63,66,67,73,74,76,77,78,80,106,107,114,122,123,124,12 6,132,210,

233,234,235,313,319,320,327,15 A and B,16 A and B,22 A and B,23 A and B,24 A and B,42B,43B,47 A and B,62 A and B,69 A and B,79 A and B

+++++ 28,60,75,81 ,99,125,15 A and B,16 A and B,17 A and B,17 A and B,18 A and B,18 A and B,19 A and B,19 A and B,20 A and B,20 A and B,24 A and B,32 A and B,32 A and B,61 A and B,61 A and B,62 A and B,64 A and B,64 A and B,65 A and B,65 A and B,68 A and B,68 A and B,69 A and B,70 A and B,70 A and B,71 A and B,71 A and B,72 A and B,72 A and B,79 A and B,82 A and B,82 A and B

Table 13. KRAS G12C FRET data

IC50* Examples of A compounds + 1 ,7,41 ,58,88,90,95,109,113,115,135,137,139,140,142,146,148,149,150 ,151 ,156,161 ,162,16 5,167,171 ,174,175,183,184,186,190,192,196,198,200,201 ,203,204,209,230,231 ,241 ,249,25 2,263,264,266,267,268,269,270,271 ,272,273,274,280,281 ,306,307,309,311 ,312,325,329,33 0,331.42A.43A

++ 2,3,4,5,6,8,9,10,11 ,12,13,14,31 ,33,34,35,36,37,38,39,44,45,48,49,50,51 ,52,53,54,56,59,73,7 4,77,78,83,84,85,86,87,89,91 ,92,93,94,96,97,100,101 ,102,103,104,105,106,110,114,117,11

9,124,125,128,129,130,131 ,133,134,136,138,141 ,143,144,147,152,153,154,155,157,158,15 9,160,168,169,170,172,173,176,177,178,179,181 ,182,185,188,189,191 ,193,195,197,202,20 5,206,207,208,211 ,213,214,215,216,217,220,222,223,224,225,232,236,237,238,239 ,243,24 4,245,246,247,248,250,251 ,253,254,255,256,257,258,259,260,261 ,262,265,275,276,277,27 8,279,282,283,284,285,287,288,289,291 ,293,294,297,298,299,301 ,302,305,308,314,315,31 6,317,319,322,323,326,328,332,16 A and B,17 A and B,18 A and B,22 A and B,23 A and B,32 A and B,43B,68 A and B,69 A and B,82 A and B

+++ 40,46,55,57,66,67,75,76,80,81 ,98,107,108,111 ,112,116,118,120,121 ,122,123,126,127,132,1 45,163,164,166,180,187,194,199,210,212,218,219,221 ,226,227,228,229,233,234,235,240,2

42,286,290,292,295,296,300,303,304,310,313,318,320,321 ,324,15 A and B,16 A and B,17 A and B,19 A and B,20 A and B,24 A and B,42B,47 A and B,61 A and B,62 A and B,70 A and B,72 A and B,79 A and B

++++ 25,26,27,28,29,63,327,18 A and B,32 A and B,47 A and B,62 A and B,65 A and B,68 A and B,69 A and B,71 A and B,72 A and B,82 A and B

+++++ 30,60,99,15 A and B,19 A and B,20 A and B,24 A and B,61 A and B,64 A and B,64 A and B,65 A and B,70 A and B,71 A and B,79 A and B

Table 14. KRAS Q61H FRET data

IC50* Examples of A compounds

+ 209,230,231 ,246,249,258,264,281 ,325

++ 36.37.91 .92.108.117.119.141.144.163.164.166.167.170.186.202.203.204. 205.207.208.212.2

13.214.215.216.217.218.220.222.223.224.225.227.228.229.23 2.237.238.239.241.244.245.2

48.252.253.254.255.256.257.259.260.261.262.263.265.266.26 7.268.269.271.272.273.274.2

75.276.277.278.279.280.282.283.284.285.286.287.288.289.29 1 .293.294.297.298.299.300.3 01 ,302,303,304,305,306,307,308,309,311 ,314,315,316,317,322,323,326,328,329,330,331 ,3

32

+++ 165,206,210,211 ,219,221 ,226,233,235,236,240,242,243,247,250,251 ,270,290,292,295,296, 310,312,313,318,319,320,321 ,324

++++ 234,327

+++++ None

Table 15. NRAS G12C FRET data

IC50* Examples of A compounds

+ 1 ,5,7,41 ,58,59,86,88,90,95,109,113,115,135,136,137,138,139,140,142,1 46,148,149,150,151 ,155,156,159,160,161 ,162,165,167,171 ,174,175,184,186,192,196,198,200,201 ,203,204,209, 230,231 ,241 ,249,252,263,264,266,267,268,269,270,271 ,272,273,274,276,278,280,281 ,284, 306,307,309,311 ,312,325,329,330,42A,43A

++ 2,3,6,8,9,10,11 ,12,13,14,33,34,35,36,37,38,39,44,45,48,49,50,52,53,54,73,74 ,77,78,83,84,8 5,87,89,91 ,92,93,96,97,100,101 ,102,103,104,105,106,110,114,124,125,128,129,130,131 ,13 3,134,141 ,143,144,147,152,153,154,157,158,168,169,170,172,173,176,177 ,178,179,181 ,18 2,183,185,188,189,190,191 ,193,195,197,202,205,206,207,208,211 ,213,214,215,216,217,21 8,222,223,224,225,227,228,232,236,237,238,239,243,244,245,24 6,247,248,251 ,253,254,25 5,256,257,258,259,260,261 ,262,265,275,277,279,282,283,285,287,288,289,291 ,293,294,29 7,298,299,300,301 ,302,305,308,314,315,316,317,319,322,323,326,328,331 ,332,16 A and B,17 A and B,18 A and B,22 A and B,23 A and B,43B,68 A and B,69 A and B

+++ 219,4,31 ,40,46,51 ,55,56,57,66,67,75,76,80,94,107,108,111 ,112,116,117,118,119,120,121 ,1

22,123,126,127,132,145,163,164,166,180,187,194,199,210,21 2,220,221 ,226,229,233,235,2 40,242,250,286,290,292,295,296,303,304,310,313,320,321 ,324,15 A and B,16 A and B,17 A and B,19 A and B,20 A and B,32 A and B,42B,47 A and B,61 A and B,62 A and B,70 A and B,71 A and B,72 A and B,72 A and B,79 A and B,82 A and B

++++ 318,26,28,29,63,81 ,98,234,327,18 A and B,24 A and B,47 A and B,62 A and B,68 A and B,69 A and B,82 A and B

+++++ 25,27,30,60,99,15 A and B,19 A and B,20 A and B,24 A and B,32 A and B,61 A and B,64 A and B,64 A and B,65 A and B,65 A and B,70 A and B,71 A and B,79 A and B

Table 16. NRAS WT FRET data

IC50* Examples of A compounds

+ 258,264,315,325

++ 37,91 ,92,117,141 ,167,170,186,204,205,207,208,209,213,214,217,222,223,224,225 ,230,231 , 237,238,239,241 ,245,246,248,249,252,253,255,256,257,259,260,261 ,262,263,265,267,268, 269,272,273,274,276,278,279,280,281 ,282,283,284,285,287,288,289,291 ,297,298,301 ,302, 305,306,307,308,309,311 ,314,316,317,322,323,326,328,329,330,331 ,332

+++ 36.108.119.144.163.164.165.166.202.203.206.211.212.215.216.2 18.219.220.221.226.227.2

28.229.232.236.242.243.244.247.250.251.254.266.270.271.27 5.277.286.290.292.293.294.2 95,296,299,300,303,304,310,312,321 ,324

++++ 210,233,234,235,240,313,318,319,320

+++++ 327

Table 17. NRAS Q61 K FRET data

IC50* Examples of A compounds

+ None

++ 167,170,216,217,218,219,220,222,223,224,225,227,228,229,231 ,246,249,258,260,264,281 , 289,291 ,293,294,297,298,299,300,301 ,302,315,325,326,329

+++ 36.37.91 .92.108.117.119.141.163.186.202.203.204.205.207.208.209.212. 213.214.221.226.2

30.232.235.237.238.239.241.245.248.252.253.254.255.256.25 7.259.261.262.263.265.266.2

67.268.269.271.272.273.274.275.276.277.278.279.280.282.28 3.284.285.286.287.288.290.2 92,295,296,303,304,305,306,307,308,309,310,311 ,314,316,317,319,322,323,324,328,330,3 31,332

++++ 144,164,165,166,206,210,211 ,215,233,234,236,242,243,244,247,250,251 ,270,312,313,318, 320,321

+++++ 240,327

Table 18. NRAS Q61R FRET data

IC50* Examples of A compounds

+ None

++ 170,209,222,223,224,230,249,258,264,289,297,298,301,302,325, 326,329

+++ 36.37.91.92.108.117.141.163.167.186.202.203.204.205.207.208. 212.213.214.216.217.218.2

19.220.221.225.226.227.228.229.231.232.237.238.239.241.24 4.245.246.248.252.253.254.2

55.256.257.259.260.261.262.263.265.266.267.268.269.271.27 2.273.274.275.276.278.279.2

80.281.282.283.284.285.286.287.288.290.291.292.293.294.29 6.299.300.303.304.305.306.3 07,308,309,310,311 ,314,315,316,317,322,323,324,328,330,331 ,332

++++ 119,144,164,165,166,206,210,211 ,215,234,235,236,240,242,243,247,250,251 ,270,277,295, 312,313,318,319,320,321

+++++ 233,327

Cross-linking of Ras Proteins with Compounds of the Invention to Form Conjugates

Note - The following protocol describes a procedure for monitoring cross-linking of K-Ras G12C (GMP-PNP) to a compound of the invention. This protocol may also be executed substituting other Ras proteins or nucleotides, such as such as K-Ras G12D and K-Ras G13D.

The purpose of this biochemical assay was to measure the ability of test compounds to covalently label nucleotide-loaded K-Ras isoforms. In assay buffer containing 12.5 mM HEPES pH 7.4, 75 mM NaCI, 1 mM MgCb, 1 mM BME, 5 μΜ Cyclophilin A, and 2 μΜ test compound, a 5 μΜ stock of GMP- PNP-loaded K-Ras (1-169) G12C was diluted 10-fold to yield a final concentration of 0.5 μΜ; with final sample volume being 100 μΙ~

The sample was incubated at 25 °C for a time period of up to 24 hours prior to quenching by the addition of 10 pL of 5% Formic Acid. Quenched samples were centrifuged at 15000 rpm for 15 minutes in a benchtop centrifuge before injecting a 10 μ!_ aliquot onto a reverse phase C4 column and eluting into the mass spectrometer with an increasing acetonitrile gradient in the mobile phase. Analysis of raw data was carried out using Waters MassLynx MS software, with % bound calculated from the deconvoluted protein peaks for labeled and unlabeled K-Ras. In vitro Cell Proliferation Panels

Potency for inhibition of cell growth was assessed at CrownBio using standard methods. Briefly, cell lines were cultured in appropriate medium, and then plated in 3D methylcellulose. Inhibition of cell growth was determined by CellTiter-Glo® after 5 days of culture with increasing concentrations of compounds. Compound potency was reported as the 50% inhibition concentration (absolute IC50). The assay took place over 7 days. On day 1 , cells in 2D culture were harvested during logarithmic growth and suspended in culture medium at 1x105 cells/ml. Higher or lower cell densities were used for some cell lines based on prior optimization. 3.5 ml of cell suspension was mixed with 6.5% growth medium with 1% methylcellulose, resulting in a cell suspension in 0.65% methylcellulose. 90 μΙ of this suspension was distributed in the wells of 296-well plates. One plate was used for day 0 reading and 1 plate was used for the end-point experiment. Plates were incubated overnight at 37 C with 5% C02. On day 2, one plate (for to reading) was removed and 10 μΙ growth medium plus 100 μΙ CellTiter-Glo® Reagent was added to each well. After mixing and a 10 minute incubation, luminescence was recorded on an EnVision Multi-Label Reader (Perkin Elmer). Compounds in DMSO were diluted in growth medium such that the final, maximum concentration of compound was 10 μΜ, and serial 4-fold dilutions were performed to generate a 9-point concentration series. 10 μΙ of compound solution at 10 times final concentration was added to wells of the second plate. Plate was then incubated for 120 hours at 37C and 5% C02. On day 7 the plates were removed, 100 μΙ CellTiter-Glo® Reagent was added to each well, and after mixing and a 10 minute incubation, luminescence was recorded on an EnVision Multi- Label Reader (Perkin Elmer). Data was exported to GeneData Screener and modeled with a sigmoidal concentration response model in order to determine the IC50 for compound response.

Not all cell lines with a given RAS mutation may be equally sensitive to a RAS inhibitor targeting that mutation, due to differential expression of efflux transporters, varying dependencies on RAS pathway activation for growth, or other reasons. This has been exemplified by the cell line KYSE-410 which, despite having a KRAS G12C mutation, is insensitive to the KRAS G12C (OFF) inhibitor MRTX-849 (Hallin et al., Cancer Discovery 10:54-71 (2020)), and the cell line SW1573, which is insensitive to the KRAS G12C (OFF) inhibitor AMG510 (Canon et al., Nature 575:217-223 (2019)).

Table 19: IC50 values for various cancer cell lines with Compound B

•Key: low sensitivity: IC50 > 1 uM moderately sensitive: 1 uM > IC50 > 0.1 uM very sensitive: IC50 < 0.1 uM blank = not measured

Cell Line Histotype Cancer Driver/Mutant IC50*

A-375 Skin BRAF V600E low sensitivity

KYSE-410 HN/Esophagus KRAS G12C moderately sensitive

MIA PaCa-2 Pancreas KRAS G12C very sensitive

NCI-H358 Lung KRAS G12C very sensitive

SW1573 Lung KRAS G12C low sensitivity

SW837 Intestine/Large/Colorectum KRAS G12C very sensitive

LS513 Intestine/Large/Colorectum KRAS G12D moderately sensitive

HuCCTI Liver/Bile duct KRAS G12D very sensitive

HCC1588 Lung KRAS G12D low sensitivity

HP AC Pancreas KRAS G12D very sensitive

As PC-1 Pancreas KRAS G12D moderately sensitive

AGS Stomach KRAS G12D very sensitive

HEC-1-A Uterus KRAS G12D very sensitive

SW403 Intestine/Large/Colorectum KRAS G12V moderately sensitive

NOZ Liver/Bile duct KRAS G12V low sensitivity

NCI-H441 Lung KRAS G12V moderately sensitive

NCI-H727 Lung KRAS G12V moderately sensitive

OVCAR-5 Ovary KRAS G12V moderately sensitive

Capan-2 Pancreas KRAS G12V moderately sensitive

SW48 Intestine/Large/Colorectum not MAPK (PIK3CA G914R, EGFR G719S)

NCI-H2009 Lung other KRAS (G12A) moderately sensitive

CAL-62 HN/Thyroid other KRAS (G12R) low sensitivity

A549 Lung other KRAS (G12S) low sensitivity

TOV-21G Ovary other KRAS (G13C) low sensitivity

DV-90 Lung other KRAS (G13D) low sensitivity

HCT116 Intestine/Large/Colorectum other KRAS (G13D) moderately sensitive

NCI-H747 Intestine/Large/Colorectum other KRAS (G13D) moderately sensitive

NCI-H460 Lung other KRAS (Q61 H) moderately sensitive

Calu-6 Lung other KRAS (Q61 K) moderately sensitive

SNU-668 Stomach other KRAS (Q61K) moderately sensitive

OZ Liver/Bile duct other KRAS (Q61 L) moderately sensitive

SW948 Intestine/Large/Colorectum other KRAS (Q61 L) low sensitivity other MARK (BRAF

BxPC-3 Pancreas V487 P492delinsA) low sensitivity other MAPK (EGFR

NCI-H1975 Lung T790M, L858R) moderately sensitive other MAPK (EML4-

NCI-H3122 Lung ALK(E13,A20)) moderately sensitive other MAPK (KRAS

YCC-1 Stomach Amp)

MeWo Skin other MAPK (NF1 mut) low sensitivity

NCI-H1838 Lung other MAPK (NF1 mut) moderately sensitive

In vivo PD and Efficacy Data with Compound A, a compound of the present invention

FIG. 1A:

Methods: The human pancreatic adenocarcinoma HPAC KRAS G12D/wt xenograft model was used for a single-dose PD study. Compound A (AsPC-1 pERK K-Ras G12D EC50: 0.036 uM) was administered at 30 and 60 mg/kg by intraperitoneal injection (ip injection). The treatment groups with sample collections at various time points were summarized in Table 20 below. Tumor samples were collected to assess RAS/ERK signaling pathway modulation by measuring the mRNA level of human DUSP6 in qPCR assay.

Table 20. Summary of treatment groups, doses, and time points for single-dose PD study using HPAC tumors.

Compound/group Dose/Regimen PD, n = 3/time point

Vehicle control 10 ml/kg ip 1h, 24h

Compound A 30 mg/kg ip 1 h, 4h, 8h, 24h

Compound A 60 mg/kg ip 1 h, 4h, 6h, 24h

Results: In FIG. 1 A, Compound A at either 30 mg/kg or 60 mg/kg led to inhibition of DUSP6 mRNA levels in tumors at all time points tested, indicating strong MAPK pathway modulation. The inhibitory effects of Compound A on DUSP6 mRNA levels are durable even 24 hours after drug administration.

FIG. 1B:

Methods: Effects of Compound A on tumor cell growth in vivo were evaluated in the human pancreatic adenocarcinoma HPAC KRAS G12D/wt xenograft model using female BALB/c nude mice (6-8 weeks old). Mice were implanted with HPAC tumor cells in PBS (3 x 106 cells/mouse) subcutaneously in the flank. Once tumors reached an average size of ~150 mm3, mice were randomized to treatment groups to start the administration of test articles or vehicle. Compound A was administered by intraperitoneal injection once daily. Body weight and tumor volume (using calipers) was measured twice weekly until study endpoints.

Results: Single-agent Compound A administered at 10 mg/kg ip daily led to a TGI of 89.9% at Day 28, while both 30 mg/kg and 60 mg/kg Compound A dosed ip daily resulted in complete regression of all tumors in the group (complete regression defined as >85% tumor regression from baseline) at the end of treatment (Day 35 after treatment started) in HPAC CDX model with heterozygous KRAS G12D. The anti-tumor activity of all 3 tested doses of Compound A was statistically significant compared with control group (*”p<0.001 , ordinary One-way ANOVA with multiple comparisons via a post-hoc Tukey's test).

While the invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure come within known or customary practice within the art to which the invention pertains and may be applied to the essential features set forth herein.

All publications, patents and patent applications are herein incorporated by reference in their entirety to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety.

Appendix B-1

RAS INHIBITORS

Background

The vast majority of small molecule drugs act by binding a functionally important pocket on a target protein, thereby modulating the activity of that protein. For example, cholesterol-lowering drugs known as statins bind the enzyme active site of HMG-CoA reductase, thus preventing the enzyme from engaging with its substrates. The fact that many such drug/target interacting pairs are known may have misled some into believing that a small molecule modulator could be discovered for most, if not all, proteins provided a reasonable amount of time, effort, and resources. This is far from the case. Current estimates are that only about 10% of all human proteins are targetable by small molecules. Bojadzic and Buchwald, Curr Top Med Chem 18: 674-699 (2019). The other 90% are currently considered refractory or intractable toward above-mentioned small molecule drug discovery. Such targets are commonly referred to as “undruggable.” These undruggable targets include a vast and largely untapped reservoir of medically important human proteins. Thus, there exists a great deal of interest in discovering new molecular modalities capable of modulating the function of such undruggable targets.

It has been well established in literature that Ras proteins (K-Ras, H-Ras and N-Ras) play an essential role in various human cancers and are therefore appropriate targets for anticancer therapy. Indeed, mutations in Ras proteins account for approximately 30% of all human cancers in the United

States, many of which are fatal. Dysregulation of Ras proteins by activating mutations, overexpression or upstream activation is common in human tumors, and activating mutations in Ras are frequently found in human cancer. For example, activating mutations at codon 12 in Ras proteins function by inhibiting both GTPase-activating protein (GAP)-dependent and intrinsic hydrolysis rates of GTP, significantly skewing the population of Ras mutant proteins to the “on * (GTP-bound) state (Ras(ON)), leading to oncogenic

MAPK signaling. Notably, Ras exhibits a picomolar affinity for GTP, enabling Ras to be activated even in the presence of low concentrations of this nucleotide. Mutations at codons 13 (e.g., G13D) and 61 (e.g., Q61 K) of Ras are also responsible for oncogenic activity in some cancers.

Despite extensive drug discovery efforts against Ras during the last several decades, a drug directly targeting Ras is still not approved. Additional efforts are needed to uncover additional medicines for cancers driven by the various Ras mutations.

Summary

Provided herein are Ras inhibitors. The approach described herein entails formation of a high affinity three-component complex, or conjugate, between a synthetic ligand and two intracellular proteins which do not interact under normal physiological conditions: the target protein of interest (e.g., Ras), and a widely expressed cytosolic chaperone (presenter protein) in the cell (e.g., cyclophilin A). More specifically, in some embodiments, the inhibitors of Ras described herein induce a new binding pocket in Ras by driving formation of a high affinity tri-complex, or conjugate, between the Ras protein and the widely expressed cytosolic chaperone, cyclophilin A (CYPA). Without being bound by theory, the inventors believe that one way the inhibitory effect on Ras is effected by compounds of the invention and the complexes, or conjugates, they form is by steric occlusion of the interaction site between Ras and downstream effector molecules, such as RAF and PI3K, which are required for propagating the oncogenic signal.

As such, in some embodiments, the disclosure features a compound, or pharmaceutically acceptable salt thereof, of structural Formula I:

Formula I wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH3)C(0)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 10- membered heteroarylene;

B is absent, -CH(R 9 )-, >C=CR 9 R 9' , or >CR 9 R 9 where the carbon is bound to the carbonyl carbon of -N(R 11 )C(0)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C1-C4 alkylene, optionally substituted Ci-C* alkenylene, optionally substituted C1-C4 heteroalkylene, -C(0)0-CH(R 8 )- where C is bound to -C(R 7 R 8 )-, -C(0)NH-CH(R 8 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C1-C4 heteroalkylene, or 3 to 8-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a vinyl ketone, a vinyl sulfone, an ynone, a haloacetal, or an alkynyl sulfone;

X 1 is optionally substituted C1-C2 alkylene, NR, O, or S(0) n ;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, optionally substituted C2-C4 alkynyl, C(0)R’, C(0)0R\ C(0)N(R') 2 , S(0)R', S(0) 2 R', or S(0) 2 N(R') 2 ; each R’ is, independently, H or optionally substituted C1-C4 alkyl;

Y 1 is C, CH, or N; Y 2 , Y 3 , Υ 4 , and Υ 7 are, independently, C or N;

Y 5 is CH, CHa, or N;

Y 6 is C(O), CH, CH 2 , or N;

R 1 is cyano, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl, or

R 1 and R 2 combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 2 is absent, hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted C 2 -Ce alkenyl, optionally substituted C 2 -Ce alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C1-C4 alkyl optionally substituted with halogen, cyano, hydroxy, or C1-C4 alkoxy, cyclopropyl, or cyclobutyl;

R® is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl, or R® and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R ® is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted C 2 -Ce alkenyl, optionally substituted Ca-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R® combine with the carbon atom to which they are attached to form C=CR r R ® ’; C=N(OH), C=N(0-Ci-Cs alkyl), C=0, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7a and R are, independently, hydrogen, halo, optionally substituted C1-C3 alkyl, or combine with the carbon to which they are attached to form a carbonyl;

R T is hydrogen, halogen, or optionally substituted C1-C3 alkyl; R ®' is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted Ca-Ce alkenyl, optionally substituted Ca-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R ® ’ combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is H, F, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; or

R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 9' is hydrogen or optionally substituted Ci-Ce alkyl; or R 9 and R 9 , combined with the atoms to which they are attached, form a 3 to 6-membered cycloalkyl or a 3 to 6-membered heterocycloalkyl;

R 10 is hydrogen, halo, hydroxy, C1-C3 alkoxy, or C1-C3 alkyl;

R 10a is hydrogen or halo;

R 1 ' is hydrogen or C1-C3 alkyl; and

R 2 ' is hydrogen or C1-C3 alkyl (e.g., methyl).

Also provided are pharmaceutical compositions comprising a compound of Formula I, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.

Further provided is a conjugate, or salt thereof, comprising the structure of Formula IV:

M-L-P Formula IV wherein L is a linker;

P is a monovalent organic moiety; and M has the structure of Formula V:

Formula V wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH3)C(0)-(CH2)- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene;

B is absent, -CH(R 9 )-, >C=CR 9 R 9' , or >CR 9 R 9' where the carbon is bound to the carbonyl carbon of -N(R 11 )C(0)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C1-C4 alkylene, optionally substituted C1-C4 alkenylene, optionally substituted C1-C4 heteroalkylene, -C(0)0-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(0)NH-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C1-C4 heteroalkylene, or 3 to 8-membered heteroarylene;

X 1 is optionally substituted C1-C2 alkylene, NR, O, or S(0) n ; X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2; R is hydrogen, cyano, optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, optionally substituted C2-C4 alkynyl, C(0)R’, C(0)0R', C(0)N(R’) 2 , S(0)R', S(0) 2 R', or S(0) 2 N(R') 2 ; each R ' is, independently, H or optionally substituted C1-C4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 is CH, CH 2 , or N;

Y 6 is C(O), CH, CH 2 , or N;

R 1 is cyano, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl, or

R 1 and R 2 combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 2 is absent, hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted C 2 -Ce alkenyl, optionally substituted Ca-Ce alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C1-C4 alkyl optionally substituted with halogen, cyano, hydroxy, or C1-C4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C1-C 3 alkyl, or R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted C 2 -Ce alkenyl, optionally substituted Ca-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR r R 8' ; C=N(OH), C=N(0-Ci-Cs alkyl), C=0, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7a and R are, independently, hydrogen, halo, optionally substituted C1-C3 alkyl, or combine with the carbon to which they are attached to form a carbonyl;

R T is hydrogen, halogen, or optionally substituted C1-C3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted Ca-Ce alkenyl, optionally substituted Ca-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8 ’ combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl; R 9 is H, F, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl, or

R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 9' is hydrogen or optionally substituted Ci-Ce alkyl; or

R 9 and R 9' , combined with the atoms to which they are attached, form a 3 to 6-membered cycloalkyl or a 3 to 6-membered heterocycloalkyl;

R 10 is hydrogen, halo, hydroxy, C1-C3 alkoxy, or C1-C3 alkyl;

R 10a is hydrogen or halo;

R 11 is hydrogen or C1-C3 alkyl; and R 2 ' is H or C1-C3 alkyl.

Also provided is a method of treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof.

In some embodiments, a method is provided of treating a Ras protein-related disorder in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof.

Further provided is a method of inhibiting a Ras protein in a cell, the method comprising contacting the cell with an effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof.

It is specifically contemplated that any limitation discussed with respect to one embodiment of the invention may apply to any other embodiment of the invention. Furthermore, any compound or composition of the invention may be used in any method of the invention, and any method of the invention may be used to produce or to utilize any compound or composition of the invention.

Brief Description of the Drawings

FIG. 1 A and FIG. 1 B: These figures illustrate a matched pair analysis of potencies of certain compounds of the present invention (Formula BB) (points on the right) and corresponding compounds of Formula AA (points on the left) wherein a H is replaced with (S)Me in the context of two different cell- based assays. The y axes represent pERK EC50 (FIG. 1 A) or CTG IC50 (FIG. 1 B) as measured in an H358 cell line.

FIG. 2A and FIG. 2B: A compound of the present invention, Compound A, drove deep regressions in vivo in a NSCLC (KRAS G12C) xenograft model. Some animals exhibited complete responses (CR) = 3 consecutive tumor measurements ≤ 30 mm3. FIG. 2A shows Compound A dosed at 100 mg/kg by daily oral gavage led to tumor regression in NCI-H358 KRASG12C xenograft model, which is a sensitive model to KRASG12C inhibition alone. The spaghetti titer plot (FIG. 2B) displaying individual tumor growth is shown next to the tumor volume plot (FIG. 2A).

FIG. 3A and FIG. 3B: A compound of the present invention, Compound B, drove tumor xenograft regressions in combination with a MEK inhibitor, cobimetinib, in a NSCLC (KRAS G12C) model. FIG. 3A shows the combination of intermittent intravenous administration of Compound B at 50 mg/kg plus daily oral administration of cobimetinib at 2.5 mg/kg drove tumor regression, whereas each single agent led to tumor growth inhibition. End of study responses were shown as waterfall plots (FIG. 3B), which indicate 6 out 10 mice had tumor regression in the combination group, whereas no tumor regressions recorded in each single agent group.

FIG. 4A and FIG. 4B: A compound of the present invention, Compound C, dosed weekly with daily SHP2 inhibitor, RMC-4550, drove xenograft regressions in a NSCLC (KRAS G12C) model. In FIG. 4A, the combinatorial activity of once weekly intravenous administration of Compound C at 60 mg/kg plus daily oral administration of SHP2 inhibitor at 30 mg/kg is shown. End of study responses in individual tumors were plotted as a waterfall plot (FIG. 4B).

FIG. 5: A compound of the present invention, Compound D, combined with a MEK inhibitor, trametinib, suppressed in vitro growth durably in a long-term cell growth NSCLC (KRAS G12C) model.

Definitions and Chemical Terms

In this application, unless otherwise clear from context, (i) the term "a” means “one or more”; (ii) the term "or" is used to mean "and/or" unless explicitly indicated to refer to alternatives only or the alternative are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and "and/or"; (iii) the terms “comprising" and "including" are understood to encompass itemized components or steps whether presented by themselves or together with one or more additional components or steps; and (iv) where ranges are provided, endpoints are included.

As used herein, the term “about” is used to indicate that a value includes the standard deviation of error for the device or method being employed to determine the value. In certain embodiments, the term “about” refers to a range of values that fall within 25%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less in either direction (greater than or less than) of a stated value, unless otherwise stated or otherwise evident from the context (e.g., where such number would exceed 100% of a possible value).

As used herein, the term “adjacent” in the context of describing adjacent atoms refers to bivalent atoms that are directly connected by a covalent bond.

A “compound of the present invention" and similar terms as used herein, whether explicitly noted or not, refers to Ras inhibitors described herein, including compounds of Formula I and subformula thereof, and compounds of Table 1 and Table 2, as well as salts (e.g., pharmaceutically acceptable salts), solvates, hydrates, stereoisomers (including atropisomers), and tautomers thereof.

The term “wild-type" refers to an entity having a structure or activity as found in nature in a “normal" (as contrasted with mutant, diseased, altered, etc) state or context. Those of ordinary skill in the art will appreciate that wild-type genes and polypeptides often exist in multiple different forms (e.g., alleles).

Those skilled in the art will appreciate that certain compounds described herein can exist in one or more different isomeric (e.g., stereoisomers, geometric isomers, atropisomers, tautomers) or isotopic (e.g., in which one or more atoms has been substituted with a different isotope of the atom, such as hydrogen substituted for deuterium) forms. Unless otherwise indicated or clear from context, a depicted structure can be understood to represent any such isomeric or isotopic form, individually or in combination.

Compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated. Compounds of the present disclosure that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically active starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of olefins, C=N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present disclosure. Cis and trans geometric isomers of the compounds of the present disclosure are described and may be isolated as a mixture of isomers or as separated isomeric forms.

In some embodiments, one or more compounds depicted herein may exist in different tautomeric forms. As will be clear from context, unless explicitly excluded, references to such compounds encompass all such tautomeric forms. In some embodiments, tautomeric forms result from the swapping of a single bond with an adjacent double bond and the concomitant migration of a proton. In certain embodiments, a tautomeric form may be a prototropic tautomer, which is an isomeric protonation states having the same empirical formula and total charge as a reference form. Examples of moieties with prototropic tautomeric forms are ketone - enol pairs, amide - imidic acid pairs, lactam - lactim pairs, amide - imidic acid pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, such as, 1 H- and 3H-imidazole, 1 H-, 2H- and 4H-1 ,2,4-triazole,

1 H- and 2H- isoindole, and 1 H- and 2H-pyrazole. In some embodiments, tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution. In certain embodiments, tautomeric forms result from acetal interconversion.

Unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. Exemplary isotopes that can be incorporated into compounds of the present invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, and iodine, such as 41, 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 0, 17 0, 18 0, Κ Ρ, 33 P, 35 S, 18 F, ^CI, 123 l and 125 l. Isotopically-labeled compounds (e.g., those labeled with 3 H and 14 C) can be useful in compound or substrate tissue distribution assays. Tritiated (i.e., 3 H) and carbon-14 (i.e., 14 C) isotopes can be useful for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements). In some embodiments, one or more hydrogen atoms are replaced by 2 H or 3 H, or one or more carbon atoms are replaced by 13 C- or 14 C-enriched carbon. Positron emitting isotopes such as 15 0, ,3 N, 11 C, and ,e F are useful for positron emission tomography (PET) studies to examine substrate receptor occupancy. Preparations of isotopically labelled compounds are known to those of skill in the art. For example, isotopically labeled compounds can generally be prepared by following procedures analogous to those disclosed for compounds of the present invention described herein, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.

As is known in the art, many chemical entities can adopt a variety of different solid forms such as, for example, amorphous forms or crystalline forms (e.g., polymorphs, hydrates, solvate). In some embodiments, compounds of the present invention may be utilized in any such form, including in any solid form. In some embodiments, compounds described or depicted herein may be provided or utilized in hydrate or solvate form.

At various places in the present specification, substituents of compounds of the present disclosure are disclosed in groups or in ranges. It is specifically intended that the present disclosure include each and every individual subcombination of the members of such groups and ranges. For example, the term “Ci-Ce alkyl” is specifically intended to individually disclose methyl, ethyl, Cs alkyl, C* alkyl, Cs alkyl, and Ce alkyl. Furthermore, where a compound includes a plurality of positions at which substituents are disclosed in groups or in ranges, unless otherwise indicated, the present disclosure is intended to cover individual compounds and groups of compounds (e.g., genera and subgenera) containing each and every individual subcombination of members at each position.

The term “optionally substituted X" (e.g., “optionally substituted alkyl") is intended to be equivalent to “X, wherein X is optionally substituted" (e.g., “alkyl, wherein said alkyl is optionally substituted"). It is not intended to mean that the feature “X" (e.g., alkyl) perse is optional. As described herein, certain compounds of interest may contain one or more “optionally substituted" moieties. In general, the term “substituted", whether preceded by the term “optionally" or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent, e.g., any of the substituents or groups described herein. Unless otherwise indicated, an “optionally substituted” group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. For example, in the term “optionally substituted Ci-Ce alkyl-Cz-Cg heteroaryl,” the alkyl portion, the heteroaryl portion, or both, may be optionally substituted. Combinations of substituents envisioned by the present disclosure are preferably those that result in the formation of stable or chemically feasible compounds. The term "stable", as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein.

Suitable monovalent substituents on a substitutable carbon atom of an "optionally substituted” group may be, independently, deuterium; halogen; -(CH2)o-4R°; -(CH2)o-40R°; -0(CH2)o-4R°; -0-(CH2)O-4C(0)OR°; -(CH2)O-4CH(OR°)2; -(CH2)O-4SR°; -(CH2)o-4Ph, which may be substituted with R°; -(CH2)o-40(CH2)o-iPh which may be substituted with R°; -CH=CHPh, which may be substituted with

R°; -(CH2)o-40(CH2)o-i-pyridyl which may be substituted with R°; 4-8 membered saturated or unsaturated heterocycloalkyl (e.g., pyridyl); 3-8 membered saturated or unsaturated cycloalkyl (e.g., cyclopropyl, cyclobutyl, or cyclopentyl); -NO2; -CN; -N 3 ; -(CH 2 )o-4N(R°)2; -(CH2)o-4N(R°)C(0)R°; -N(R°)C(S)R°;

)(OR°)R°, -SiR°3; -(C1-4 straight or branched alkylene)0-N(R°)2; or -(C1-4 straight or branched alkylene)C(0)0-N(R°)2, wherein each R° may be substituted as defined below and is independently hydrogen, -Ci-e aliphatic, -CFtePh, -0(CH2)o-iPh, -CH2-(5-6 membered heteroaryl ring), or a 3-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, which may be substituted as defined below.

Suitable monovalent substituents on R° (or the ring formed by taking two independent occurrences of R° together with their intervening atoms), may be, independently, halogen, -(Chfelo-aR*, O2, -SiR*3, -OSiR*3, -C(0)SR e , -(C1-4 straight or branched alkylene)C(0)OR·, or -SSR* wherein each R* is unsubstituted or where preceded by “halo" is substituted only with one or more halogens, and is independently selected from C1-4 aliphatic, -CH 2 Ph, -0(CH 2 )o-iPh, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents on a saturated carbon atom of R° include =0 and =S.

Suitable divalent substituents on a saturated carbon atom of an “optionally substituted" group include the following: =0, =S, =NNR‘ 2 , =NNHC(0)R‘, =NNHC(0)0R‘, =NNHS(0) 2 R‘, =NR‘, =NOR * , -OiCiR^J^O-, or -S(C(R‘ 2 )) 2 -3S-, wherein each independent occurrence of R * is selected from hydrogen, C1-6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents that are bound to vicinal substitutable carbons of an “optionally substituted" group include: -OtCR'^rcO-, wherein each independent occurrence of R * is selected from hydrogen, C1-6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.

Suitable substituents on the aliphatic group of R * include halogen, -R·, -(haloR·), -OH, -OR*, -0(haloR e ), -CN, -C(0)0H, -C(0)0R e , -NH 2 , -NHR e , -NR* 2 , or -NCfc, wherein each R e is unsubstituted or where preceded by “halo" is substituted only with one or more halogens, and is independently C1-4 aliphatic, -CH 2 Ph, -0(CH 2 )o-iPh, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.

Suitable substituents on a substitutable nitrogen of an “optionally substituted” group include -Rt, -NRt 2 , -C(0)Rt, -C(0)0Rt, -C(0)C(0)Rt, -C(0)CHsC{0)W, -S(0) 2 Rt, -S(0) 2 NRt 2 , -C(S)NRt 2 , -C(NH)NR+ 2, or -N(Rt)S(0) 2 R ; wherein each Rt is independently hydrogen, Ci-e aliphatic which may be substituted as defined below, unsubstituted -OPh, or an unsubstituted 3-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R , taken together with their intervening atom(s) form an unsubstituted 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.

Suitable substituents on an aliphatic group of Rt are independently halogen, -R·, -(haloR·), -OH, -OR·, -0(haloR·), -CN, -C(0)0H, -C(0)0R e , -NH 2 , -NHR·, -NR* 2 , or -HOz, wherein each R* is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C1-4 aliphatic, -CH 2 Ph, -0(CH 2 )o-iPh, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents on a saturated carbon atom of Rt include =0 and =S.

The term “acetyl,” as used herein, refers to the group -C(0)CH3. The term “alkoxy,” as used herein, refers to a -O-C1-C20 alkyl group, wherein the alkoxy group is attached to the remainder of the compound through an oxygen atom.

The term “alkyl," as used herein, refers to a saturated, straight or branched monovalent hydrocarbon group containing from 1 to 20 (e.g., from 1 to 10 or from 1 to 6) carbons. In some embodiments, an alkyl group is unbranched (i.e., is linear); in some embodiments, an alkyl group is branched. Alkyl groups are exemplified by, but not limited to, methyl, ethyl, n- and /so-propyl, />, sec-, iso- and tert-butyl, and neopentyl.

The term “alkylene," as used herein, represents a saturated divalent hydrocarbon group derived from a straight or branched chain saturated hydrocarbon by the removal of two hydrogen atoms, and is exemplified by methylene, ethylene, isopropylene, and the like. The term “Cx-Cy alkylene" represents alkylene groups having between x and y carbons. Exemplary values for x are 1 , 2, 3, 4, 5, and 6, and exemplary values for y are 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, or 20 (e.g., Ci-Ce, C1-C10, C2-C20,

Cz-Ce, C2-C10, or C2-C20 alkylene). In some embodiments, the alkylene can be further substituted with 1 , 2, 3, or 4 substituent groups as defined herein.

The term "alkenyl," as used herein, represents monovalent straight or branched chain groups of, unless otherwise specified, from 2 to 20 carbons (e.g., from 2 to 6 or from 2 to 10 carbons) containing one or more carbon-carbon double bonds and is exemplified by ethenyl, 1-propenyl, 2-propenyl, 2-methyl-1-propenyl, 1-butenyl, and 2-butenyl. Alkenyls include both cis and trans isomers. The term “alkenylene," as used herein, represents a divalent straight or branched chain groups of, unless otherwise specified, from 2 to 20 carbons (e.g., from 2 to 6 or from 2 to 10 carbons) containing one or more carbon-carbon double bonds.

The term “alkynyl," as used herein, represents monovalent straight or branched chain groups from 2 to 20 carbon atoms (e.g., from 2 to 4, from 2 to 6, or from 2 to 10 carbons) containing a carbon-carbon triple bond and is exemplified by ethynyl, and 1-propynyl.

The term “alkynyl sulfone,” as used herein, represents a group comprising the structure s *'/

S— ≡ _— „ R

, wherein R is any chemically feasible substituent described herein.

The term “amino,” as used herein, represents -N(Rt)2, e.g., -NHz and -N(CH3)z.

The term “aminoalkyl," as used herein, represents an alkyl moiety substituted on one or more carbon atoms with one or more amino moieties.

The term “amino acid,” as described herein, refers to a molecule having a side chain, an amino group, and an acid group (e.g., -CO2H or -SO3H), wherein the amino acid is attached to the parent molecular group by the side chain, amino group, or acid group (e.g., the side chain). As used herein, the term “amino acid" in its broadest sense, refers to any compound or substance that can be incorporated into a polypeptide chain, e.g., through formation of one or more peptide bonds. In some embodiments, an amino acid has the general structure HzN-C(H)(R)-C(X)H. In some embodiments, an amino acid is a naturally-occurring amino acid. In some embodiments, an amino acid is a synthetic amino acid; in some embodiments, an amino acid is a D-amino acid; in some embodiments, an amino acid is an L-amino acid. “Standard amino acid” refers to any of the twenty standard L-amino acids commonly found in naturally occurring peptides. Exemplary amino acids include alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, histidine, optionally substituted hydroxylnorvaline, isoleucine, leucine, lysine, methionine, norvaline, ornithine, phenylalanine, proline, pyrrolysine, selenocysteine, serine, taurine, threonine, tryptophan, tyrosine, and valine.

The term “aryl," as used herein, represents a monovalent monocyclic, bicyclic, or multicyclic ring system formed by carbon atoms, wherein the ring attached to the pendant group is aromatic. Examples of aryl groups are phenyl, naphthyl, phenanthrenyl, and anthracenyl. An aryl ring can be attached to its pendant group at any heteroatom or carbon ring atom that results in a stable structure and any of the ring atoms can be optionally substituted unless otherwise specified.

The term “Co," as used herein, represents a bond. For example, part of the term -N(C(0)-(Co-Cs alkylene-H)- includes -N(C(0)-(Co alkylene-H)-, which is also represented by -N(C(0)-H)-.

The terms “carbocyclic" and “carbocyclyl," as used herein, refer to a monovalent, optionally substituted C3-C12 monocyclic, bicyclic, or tricyclic ring structure, which may be bridged, fused or spirocyclic, in which all the rings are formed by carbon atoms and at least one ring is non-aromatic. Carbocyclic structures include cycloalkyl, cycloalkenyl, and cycloalkynyl groups. Examples of carbocyclyl groups are cyclohexyl, cyclohexenyl, cyclooctynyl, 1 ,2-dihydronaphthyl, 1 ,2,3,4-tetrahydronaphthyl, fluorenyl, indenyl, indanyl, decalinyl, and the like. A carbocyclic ring can be attached to its pendant group at any ring atom that results in a stable structure and any of the ring atoms can be optionally substituted unless otherwise specified.

The term "carbonyl," as used herein, represents a C(O) group, which can also be represented as c=o.

The term "carboxyl,” as used herein, means -CO2H, (C=0)(0H), COOH, or C(0)0H or the unprotonated counterparts.

The term "cyano," as used herein, represents a -CN group.

The term "cycloalkyl,” as used herein, represents a monovalent saturated cyclic hydrocarbon group, which may be bridged, fused or spirocyclic having from three to eight ring carbons, unless otherwise specified, and is exemplified by cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cycloheptyl.

The term “cycloalkenyl," as used herein, represents a monovalent, non-aromatic, saturated cyclic hydrocarbon group, which may be bridged, fused or spirocyclic having from three to eight ring carbons, unless otherwise specified, and containing one or more carbon-carbon double bonds.

The term “diastereomer," as used herein, means stereoisomers that are not mirror images of one another and are non-superimposable on one another.

The term “enantiomer," as used herein, means each individual optically active form of a compound of the invention, having an optical purity or enantiomeric excess (as determined by methods standard in the art) of at least 80% (i.e., at least 90% of one enantiomer and at most 10% of the other enantiomer), preferably at least 90% and more preferably at least 98%.

The term “guanidinyl,” refers to a group having the structure: , wherein each R is, independently, any any chemically feasible substituent described herein.

The term “guanidinoalkyl alkyl," as used herein, represents an alkyl moiety substituted on one or more carbon atoms with one or more guanidinyl moieties. The term “haloacetyl," as used herein, refers to an acetyl group wherein at least one of the hydrogens has been replaced by a halogen.

The term “haloalkyl," as used herein, represents an alkyl moiety substituted on one or more carbon atoms with one or more of the same of different halogen moieties.

The term “halogen," as used herein, represents a halogen selected from bromine, chlorine, iodine, or fluorine.

The term "heteroalkyl," as used herein, refers to an "aikyl" group, as defined herein, in which at least one carbon atom has been replaced with a heteroatom (e.g., an O, N, or S atom). The heteroatom may appear in the middle or at the end of the radical.

The term “heteroaryl," as used herein, represents a monovalent, monocyclic or polycyclic ring structure that contains at least one fully aromatic ring: i.e., they contain 4n+2 pi electrons within the monocyclic or polycyclic ring system and contains at least one ring heteroatom selected from N, O, or S in that aromatic ring. Exemplary unsubstituted heteroaryl groups are of 1 to 12 (e.g., 1 to 11 , 1 to 10, 1 to 9, 2 to 12, 2 to 11 , 2 to 10, or 2 to 9) carbons. The term “heteroaryl” includes bicyclic, tricyclic, and tetracyclic groups in which any of the above heteroaromatic rings is fused to one or more, aryl or carbocyclic rings, e.g., a phenyl ring, or a cyclohexane ring. Examples of heteroaryl groups include, but are not limited to, pyridyl, pyrazolyl, benzooxazolyl, benzoimidazolyl, benzothiazolyl, imidazolyl, thiazolyl, quinolinyl, tetrahydroquinolinyl, and 4-azaindolyl. A heteroaryl ring can be attached to its pendant group at any ring atom that results in a stable structure and any of the ring atoms can be optionally substituted unless otherwise specified. In some embodiment, the heteroaryl is substituted with 1 , 2, 3, or 4 substituents groups.

The term "heterocycloalkyl," as used herein, represents a monovalent monocyclic, bicyclic or polycyclic ring system, which may be bridged, fused or spirocyclic, wherein at least one ring is nonaromatic and wherein the non-aromatic ring contains one, two, three, or four heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur. The 5-membered ring has zero to two double bonds, and the 6- and 7-membered rings have zero to three double bonds. Exemplary unsubstituted heterocycloalkyl groups are of 1 to 12 (e.g., 1 to 11 , 1 to 10, 1 to 9, 2 to 12, 2 to 11 , 2 to 10, or 2 to 9) carbons. The term “heterocycloalkyl" also represents a heterocyclic compound having a bridged multicyclic structure in which one or more carbons or heteroatoms bridges two non-adjacent members of a monocyclic ring, e.g., a quinuclidinyl group. The term “heterocycloalkyl" includes bicyclic, tricyclic, and tetracyclic groups in which any of the above heterocyclic rings is fused to one or more aromatic, carbocyclic, heteroaromatic, or heterocyclic rings, e.g., an aryl ring, a cyclohexane ring, a cyclohexene ring, a cyclopentane ring, a cyclopentene ring, a pyridine ring, or a pyrrolidine ring.

Examples of heterocycloalkyl groups are pyrrolidinyl, piperidinyl, 1 ,2,3,4-tetrahydroquinolinyl, decahydroquinolinyl, dihydropyrrolopyridine, and decahydronapthyridinyl. A heterocycloalkyl ring can be attached to its pendant group at any ring atom that results in a stable structure and any of the ring atoms can be optionally substituted unless otherwise specified.

The term “hydroxy," as used herein, represents a -OH group.

The term “hydroxyalkyl," as used herein, represents an alkyl moiety substituted on one or more carbon atoms with one or more -OH moieties.

The term “isomer,” as used herein, means any tautomer, stereoisomer, atropiosmer, enantiomer, or diastereomer of any compound of the invention. It is recognized that the compounds of the invention can have one or more chiral centers or double bonds and, therefore, exist as stereoisomers, such as double-bond isomers (i.e., geometric E/Z isomers) or diastereomers (e.g., enantiomers (i.e., (+) or (-)) or cis/trans isomers). According to the invention, the chemical structures depicted herein, and therefore the compounds of the invention, encompass all the corresponding stereoisomers, that is, both the stereomerically pure form (e.g., geometrically pure, enantiomerically pure, or diastereomerically pure) and enantiomeric and stereoisomeric mixtures, e.g., racemates. Enantiomeric and stereoisomeric mixtures of compounds of the invention can typically be resolved into their component enantiomers or stereoisomers by well-known methods, such as chiral-phase gas chromatography, chiral-phase high performance liquid chromatography, crystallizing the compound as a chiral salt complex, or crystallizing the compound in a chiral solvent. Enantiomers and stereoisomers can also be obtained from stereomerically or enantiomerically pure intermediates, reagents, and catalysts by well-known asymmetric synthetic methods.

As used herein, the term “linker" refers to a divalent organic moiety connecting moiety B to moiety W in a compound of Formula I, such that the resulting compound is capable of achieving an IC50 of 2 uM or less in the Ras-RAF disruption assay protocol provided in the Examples below, and provided here:

The purpose of this biochemical assay is to measure the ability of test compounds to facilitate ternary complex formation between a nucleotide-loaded Ras isoform and cyclophilin A; the resulting ternary complex disrupts binding to a BRAF* 60 construct, inhibiting Ras signaling through a RAF effector.

In assay buffer containing 25 mM HEPES pH 7.3, 0.002% Tween20, 0.1% BSA, 100 mM NaCI and 5 mM MgCfe, tagless Cyclophilin A, His6-K-Ras-GMPPNP (or other Ras variant), and GST-BRAF RB0 are combined in a 384-well assay plate at final concentrations of 25 μΜ, 12.5 nM and 50 nM, respectively. Compound is present in plate wells as a 10-point 3-fold dilution series starting at a final concentration of 30 μΜ. After incubation at 25°C for 3 hours, a mixture of Anti-His Eu-W1024 and anti-GST allophycocyanin is then added to assay sample wells at final concentrations of 10 nM and 50 nM, respectively, and the reaction incubated for an additional 1.5 hours. TR-FRET signal is read on a microplate reader (Ex 320 nm, Em 665/615 nm). Compounds that facilitate disruption of a Ras:RAF complex are identified as those eliciting a decrease in the TR-FRET ratio relative to DMSO control wells.

In some embodiments, the linker comprises 20 or fewer linear atoms. In some embodiments, the linker comprises 15 or fewer linear atoms. In some embodiments, the linker comprises 10 or fewer linear atoms. In some embodiments, the linker has a molecular weight of under 500 g/mol. In some embodiments, the linker has a molecular weight of under 400 g/mol. In some embodiments, the linker has a molecular weight of under 300 g/mol. In some embodiments, the linker has a molecular weight of under 200 g/mol. In some embodiments, the linker has a molecular weight of under 100 g/mol. In some embodiments, the linker has a molecular weight of under 50 g/mol.

As used herein, a “monovalent organic moiety" is less than 500 kDa. In some embodiments, a “monovalent organic moiety" is less than 400 kDa. In some embodiments, a “monovalent organic moiety" is less than 300 kDa. In some embodiments, a “monovalent organic moiety" is less than 200 kDa. In some embodiments, a “monovalent organic moiety" is less than 100 kDa. In some embodiments, a “monovalent organic moiety" is less than 50 kDa. In some embodiments, a “monovalent organic moiety" is less than 25 kDa. In some embodiments, a “monovalent organic moiety” is less than 20 kDa. In some embodiments, a “monovalent organic moiety” is less than 15 kDa. In some embodiments, a “monovalent organic moiety” is less than 10 kDa. In some embodiments, a “monovalent organic moiety” is less than 1 kDa. In some embodiments, a “monovalent organic moiety" is less than 500 g/mol. In some embodiments, a “monovalent organic moiety * ranges between 500 g/mol and 500 kDa.

The term “stereoisomer," as used herein, refers to all possible different isomeric as well as conformational forms which a compound may possess (e.g., a compound of any formula described herein), in particular all possible stereochemical ly and conformationally isomeric forms, all diastereomers, enantiomers or conformers of the basic molecular structure, including atropisomers. Some compounds of the present invention may exist in different tautomeric forms, all of the latter being included within the scope of the present invention.

The term “sulfonyl," as used herein, represents an -S(0)z- group.

The term “thiocarbonyl," as used herein, refers to a -C(S)- group.

The term “vinyl ketone," as used herein, refers to a group comprising a carbonyl group directly connected to a carbon-carbon double bond.

The term “vinyl sulfone," as used herein, refers to a group comprising a sulfonyl group directed connected to a carbon-carbon double bond.

O

R

The term “ynone,” as used herein, refers to a group comprising the structure wherein R is any any chemically feasible substituent described herein.

Those of ordinary skill in the art, reading the present disclosure, will appreciate that certain compounds described herein may be provided or utilized in any of a variety of forms such as, for example, salt forms, protected forms, pro-drug forms, ester forms, isomeric forms (e.g., optical or structural isomers), isotopic forms, etc. In some embodiments, reference to a particular compound may relate to a specific form of that compound. In some embodiments, reference to a particular compound may relate to that compound in any form. In some embodiments, for example, a preparation of a single stereoisomer of a compound may be considered to be a different form of the compound than a racemic mixture of the compound; a particular salt of a compound may be considered to be a different form from another salt form of the compound; a preparation containing one conformational isomer ((Z) or (E)) of a double bond may be considered to be a different form from one containing the other conformational isomer ((E) or (Z)) of the double bond; a preparation in which one or more atoms is a different isotope than is present in a reference preparation may be considered to be a different form.

Detailed Description

Compounds

Provided herein are Ras inhibitors. The approach described herein entails formation of a high affinity three-component complex, or conjugate, between a synthetic ligand and two intracellular proteins which do not interact under normal physiological conditions: the target protein of interest (e.g., Ras), and a widely expressed cytosolic chaperone (presenter protein) in the cell (e.g., cyclophilin A). More specifically, in some embodiments, the inhibitors of Ras described herein induce a new binding pocket in Ras by driving formation of a high affinity tri-complex, or conjugate, between the Ras protein and the widely expressed cytosolic chaperone, cyclophilin A (CYRA). Without being bound by theory, the inventors believe that one way the inhibitory effect on Ras is effected by compounds of the invention and the complexes, or conjugates, they form is by steric occlusion of the interaction site between Ras and downstream effector molecules, such as RAF, which are required for propagating the oncogenic signal.

Without being bound by theory, the inventors postulate that both covalent and non-covalent interactions of a compound of the present invention with Ras and the chaperone protein (e.g., cyclophilin A) may contribute to the inhibition of Ras activity. In some embodiments, a compound of the present invention forms a covalent adduct with a side chain of a Ras protein (e.g., a sulfhydryl side chain of the cysteine at position 12 or 13 of a mutant Ras protein). Covalent adducts may also be formed with other side chains of Ras. In addition, or alternatively, non-covalent interactions may be at play: for example, van der Waals, hydrophobic, hydrophilic and hydrogen bond interactions, and combinations thereof, may contribute to the ability of the compounds of the present invention to form complexes and act as Ras inhibitors. Accordingly, a variety of Ras proteins may be inhibited by compounds of the present invention (e.g., K-Ras, N-Ras, H-Ras, and mutants thereof at positions 12, 13 and 61 , such as G12C, G12D, G12V, G12S, G13C, G13D, and Q61L, and others described herein).

Methods of determining covalent adduct formation are known in the art. One method of determining covalent adduct formation is to perform a “cross-linking" assay, such as under these conditions ( Note - the following protocol describes a procedure for monitoring cross-linking of K-Ras G12C (GMP-PNP) to a compound of the invention. This protocol may also be executed substituting other Ras proteins or nucleotides).

The purpose of this biochemical assay is to measure the ability of test compounds to covalently label nucleotide- loaded K-Ras isoforms. In assay buffer containing 12.5 mM HEPES pH 7.4, 75 mM

NaCI, 1 mM MgCte, 1 mM BME, 5 μΜ Cyclophilin A and 2 μΜ test compound, a 5 μΜ stock of GMP-PNP- loaded K-Ras (1 -169) G12C is diluted 10-fold to yield a final concentration of 0.5 μΜ; with final sample volume being 100 μΙ_.

The sample is incubated at 25°C for a time period of up to 24 hours prior to quenching by the addition of 10 pL of 5% Formic Acid. Quenched samples are centrifuged at 15000 rpm for 15 minutes in a benchtop centrifuge before injecting a 10 pL aliquot onto a reverse phase C4 column and eluting into the mass spectrometer with an increasing acetonitrile gradient in the mobile phase. Analysis of raw data may be carried out using Waters MassLynx MS software, with % bound calculated from the deconvoluted protein peaks for labeled and unlabeled K-Ras.

Accordingly, provided herein is a compound, or pharmaceutically acceptable salt thereof, having the structure of Formula I:

Formula I wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH3)C(0)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 10- membered heteroarylene;

B is absent, -CH(R 9 )-, >C=CR 9 R 9' , or >CR 9 R 9 where the carbon is bound to the carbonyl carbon of -N(R 11 )C(0)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C1-C4 alkylene, optionally substituted C1-C4 alkenylene, optionally substituted C1-C4 heteroalkylene, -C(0)0-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(0)NH-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C1-C4 heteroalkylene, or 3 to 8-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a vinyl ketone, a vinyl sulfone, an ynone, a haloacetal, or an alkynyl sulfone;

X 1 is optionally substituted C1-C2 alkylene, NR, O, or S(0) n ;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, optionally substituted C2-C4 alkynyl, C(0)R’, C(0)0R\ C(0)N(R') 2 , S(0)R’, S(0) 2 R', or S(0) 2 N(R') 2 ; each R’ is, independently, H or optionally substituted C1-C4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 is CH, CH 2 , or N;

Y 8 is C(O), CH, CH 2 , or N;

R 1 is cyano, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl, or

R 1 and R 2 combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 2 is absent, hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted Ca-Ce alkenyl, optionally substituted Ca-Ce alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C1-C4 alkyl optionally substituted with halogen, cyano, hydroxy, or C1-C4 alkoxy, cyclopropyl, or cyclobutyl;

R® is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl, or R® and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R® is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R® combine with the carbon atom to which they are attached to form C=CR r R ® ’; C=N(OH), C=N(0-CI-C3 alkyl), C=0, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7a and R are, independently, hydrogen, halo, optionally substituted C1-C3 alkyl, or combine with the carbon to which they are attached to form a carbonyl;

R r is hydrogen, halogen, or optionally substituted C1-C3 alkyl; R ® ’ is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted Ca-Ce alkenyl, optionally substituted Ca-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R T and R ® ’ combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is H, F, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl, or

R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 9' is hydrogen or optionally substituted Ci-Ce alkyl; or

R 9 and R 9 , combined with the atoms to which they are attached, form a 3 to 6-membered cycloalkyl or a 3 to 6-membered heterocycloalkyl;

R 10 is hydrogen, halo, hydroxy, C1-C3 alkoxy, or C1-C3 alkyl;

R 10a is hydrogen or halo;

R 11 is hydrogen or C1-C3 alkyl; and R 2 ' is hydrogen or C1-C3 alkyl (e.g., methyl).

In some embodiments, R 9 is H, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl.

In some embodiments, R 21 is hydrogen.

In some embodiments, provided herein is a compound, or pharmaceutically acceptable salt thereof, having the structure of Formula la:

Formula la wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH3)C(0)-(CH2)- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 10- membered heteroarylene;

B is -CH(R 9 )- or >C=CR 9 R 9 where the carbon is bound to the carbonyl carbon of -N(R 11 )C(0)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C1-C4 alkylene, optionally substituted C1-C4 alkenylene, optionally substituted C1-C4 heteroalkylene, -C(0)0-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(0)NH-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C1-C4 heteroalkylene, or 3 to 8-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a vinyl ketone, a vinyl sulfone, an ynone, a haloacetal, or an alkynyl sulfone;

X 1 is optionally substituted C1-C2 alkylene, NR, O, or S(0) n ;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, optionally substituted C2-C4 alkynyl, C(0)R’, C(0)0R’, C(0)N(R')2, S(0)R’, S(0) 2 R', or S(0) 2 N(R’)2; each R’ is, independently, H or optionally substituted C1-C4 alkyl; Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 is CH, CHa, or N;

Y 6 is C(O), CH, CH Z , or N;

R 1 is cyano, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl, or

R 1 and R 2 combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl ;

R 2 is absent, hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted C 2 -Ce alkenyl, optionally substituted C^Ce alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C1-C4 alkyl optionally substituted with halogen, cyano, hydroxy, or Ci-C* alkoxy, cyclopropyl, or cyclobutyl;

R® is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl, or R ® and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R ® is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted C 2 -Ce alkenyl, optionally substituted Ca-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R® combine with the carbon atom to which they are attached to form C=CR r R ® ’; C=N(OH), C=N(0-Ci-Cs alkyl), C=0, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7a and R are, independently, hydrogen, halo, optionally substituted C1-C3 alkyl, or combine with the carbon to which they are attached to form a carbonyl;

R T is hydrogen, halogen, or optionally substituted C1-C3 alkyl; R ®' is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted Ca-Ce alkenyl, optionally substituted C^Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R ® ’ combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl, or

R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl; R 9' is hydrogen or optionally substituted Ci-Ce alkyl;

R 10 is hydrogen, halo, hydroxy, C1-C3 alkoxy, or C1-C3 alkyl;

R 10a is hydrogen or halo; and R 1 ' is hydrogen or C1-C3 alkyl.

In some embodiments, the disclosure features a compound, or pharmaceutically acceptable salt thereof, of structural Formula lb:

Formula lb wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH3)C(0)-(CH2)- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -N(R 11 )C(0)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C1-C4 alkylene, optionally substituted Ci-C* alkenylene, optionally substituted C1-C4 heteroalkylene, -C(0)0-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(0)NH-CH(R 8 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C1-C4 heteroalkylene, or 3 to 8-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a vinyl ketone, a vinyl sulfone, an ynone, a haloacetal, or an alkynyl sulfone;

X 1 is optionally substituted C1-C2 alkylene, NR, O, or S(0) n ;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, optionally substituted C2-C4 alkynyl, C(0)R’, C(0)0R\ C(0)N(R')2, S(0)R’, S(0) 2 R', or S(0)2N(R') 2 ; each R’ is, independently, H or optionally substituted C1-C4 alkyl;

Y 1 is C, CH, or N; Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 and Y® are, independently, CH or N;

R 1 is cyano, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl;

R 2 is hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted Ca-Ce alkenyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C1-C4 alkyl optionally substituted with halogen, cyano, hydroxy, or C1-C4 alkoxy, cyclopropyl , or cyclobutyl ;

R® is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl, or R® and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R® is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R® combine with the carbon atom to which they are attached to form C=CR r R ® ’; C=N(OH), C=N(0-Ci-Cs alkyl), C=0, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R r is hydrogen, halogen, or optionally substituted C1-C3 alkyl; R ® ’ is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted Ca-Ce alkenyl, optionally substituted Ca-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R T and R ® ’ combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 10 is hydrogen, hydroxy, C1-C3 alkoxy, or C1-C3 alkyl; and R 11 is hydrogen or C1-C3 alkyl.

In some embodiments of compounds of the present invention, G is optionally substituted C1-C4 heteroalkylene.

In some embodiments, a compound having the structure of Formula lc is provided, or a pharmaceutically acceptable salt thereof:

Formula lc wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH3)C(0)-(CH2)- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -N(R")C(0)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a vinyl ketone, a vinyl sulfone, an ynone, or an alkynyl sulfone;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C1-C4 alkyl, optionally substituted Ca-C* alkenyl, optionally substituted C2-C4 alkynyl, C(0)R’, C(0)0R', C(0)N(R’) 2 , S(0)R', S(0) 2 R’, or S(0) 2 N(R>; each R ' is, independently, H or optionally substituted C1-C4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 and Y® are, independently, CH or N;

R 1 is cyano, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl;

R 2 is hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted Ca-Ce alkenyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C1-C4 alkyl optionally substituted with halogen, cyano, hydroxy, or C1-C4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted Cz-Ce alkenyl, optionally substituted Cz-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7 R 8' ; C=N(OH), C=N(0-CI-C3 alkyl), C=0, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted Cz-Ce alkenyl, optionally substituted Cz-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 ’ combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 10 is hydrogen, hydroxy, Ci-Ce alkoxy, or C1-C3 alkyl; and R 11 is hydrogen or C1-C3 alkyl.

In some embodiments of compounds of the present invention, X 2 is NH. In some embodiments, X 3 is CH. In some embodiments, R 11 is hydrogen. In some embodiments, R 1 ' is C1-C3 alkyl. In some embodiments, R" is methyl.

In some embodiments, a compound of the present invention has the structure of Formula Id, or a pharmaceutically acceptable salt thereof:

Formula Id wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH3)C(0)-(CH2)- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a vinyl ketone, a vinyl sulfone, an ynone, or an alkynyl sulfone; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C1-C4 alkyl, optionally substituted Ca-C* alkenyl, optionally substituted C2-C4 alkynyl, C(0)R’, C(0)0R', C(0)N(R’) 2 , S(0)R', S(0) 2 R’, or S(0) 2 N(R>; each R ' is, independently, H or optionally substituted C1-C4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 and Y® are, independently, CH or N;

R 1 is cyano, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl;

R 2 is hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted Ca-Ce alkenyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C1-C4 alkyl optionally substituted with halogen, cyano, hydroxy, or C1-C4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted Cz-Ce alkenyl, optionally substituted Cz-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7 R 8' ; C=N(OH), C=N(0-CI-C3 alkyl), C=0, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted Cz-Ce alkenyl, optionally substituted Cz-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 ’ combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; and R 10 is hydrogen, hydroxy, Ci-Ce alkoxy, or C1-C3 alkyl.

In some embodiments of a compound of the present invention, X 1 is optionally substituted Ci-Cz alkylene. In some embodiments, X 1 is methylene. In some embodiments, X 1 is methylene substituted with a Ci-Ce alkyl group or a halogen. In some embodiments, X 1 is -CH(Br)-. In some embodiments, X 1 is -CH(CH3)-. In some embodiments, R 5 is hydrogen. In some embodiments, R 5 is C1-C4 alkyl optionally substituted with halogen. In some embodiments, R 5 is methyl. In some embodiments, Y 4 is C. In some embodiments, R 4 is hydrogen. In some embodiments, Y 5 is CH.

In some embodiments, Y 6 is CH. In some embodiments, Y 1 is C. In some embodiments, Y 2 is C. In some embodiments, Y 3 is N. In some embodiments, R 3 is absent. In some embodiments, Y 7 is C. In some embodiments, a compound of the present invention has the structure of Formula le, or a pharmaceutically acceptable salt thereof:

Formula le wherein A is -N(H or CH3)C(0)-(CH2)- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a vinyl ketone, a vinyl sulfone, an ynone, or an alkynyl sulfone;

R 1 is cyano, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl;

R 2 is hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted Ca-Ce alkenyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 5 is hydrogen, C1-C4 alkyl optionally substituted with halogen, cyano, hydroxy, or C1-C4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl, or

R® and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R® is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7 R 8 ; C=N(OH), C=N(0-Ci-Ca alkyl), C=0, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7 is hydrogen, halogen, or optionally substituted C1-C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkyny!, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 ' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; and R 10 is hydrogen, hydroxy, C1-C3 alkoxy, or C1-C3 alkyl.

In some embodiments of a compound of the present invention, R 6 is hydrogen. In some embodiments, R 2 is hydrogen, cyano, optionally substituted Ci-Ce alkyl, optionally substituted 3 to 6- membered cycloalkyl, or optionally substituted 3 to 6-membered heterocycloalkyl. In some embodiments, R 2 is optionally substituted Ci-Ce alkyl. In some embodiments, R2 is fluoroalkyl. In some embodiments, R 2 is ethyl. In some embodiments, R2 is -CH2CF3. In some embodiments, R2 is C2-C6 alkynyl. In some embodiments, R2 is -CHC=CH. In some embodiments, R2 is -CH2C=CCH3. In some embodiments, R 7 is optionally substituted C1-C3 alkyl. In some embodiments, R 7 is C1-C3 alkyl. In some embodiments, R 8 is optionally substituted C1-C3 alkyl. In some embodiments, R 8 is C1-C3 alkyl.

In some embodiments, a compound of the present invention has the structure of Formula If, or a pharmaceutically acceptable salt thereof:

Formula If wherein A is -N(H or CH3)C(0)-(CH2)- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene; B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalky lene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a vinyl ketone, a vinyl sulfone, an ynone, or an alkynyl sulfone;

R 1 is cyano, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl;

R 2 is Ci-Ce alkyl or 3 to 6-membered cycloalkyl;

R 7 is C1-C3 alkyl;

R 8 is C1-C3 alkyl; and

R 9 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl.

In some embodiments of a compound of the present invention, R 1 is optionally substituted 6 to 10-membered aryl, optionally substituted 3 to 6-membered cycloalkenyl, or optionally substituted 5 to 10- membered heteroaryl. In some embodiments, R 1 is optionally substituted 6-membered aryl, optionally substituted 6-membered cycloalkenyl, or optionally substituted 6-membered heteroaryl.

In some embodiments of a compound of the present invention, stereoisomer (e.g., atropisomer) thereof.

In some embodiments of a compound of the present invention, stereoisomer (e.g., atropisomer) thereof. In some embodiments of a compound of the present invention, Ri is

In some embodiments, a compound of the present invention has the structure of Formula Ig, or a pharmaceutically acceptable salt thereof:

Formula Ig wherein A is -N(H or CH3)C(0)-(CH2)- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a vinyl ketone, a vinyl sulfone, an ynone, or an alkynyl sulfone;

R 2 is Ci-Ce alkyl, Ci-Ce fluoroalkyl, or 3 to 6-membered cycloalkyl;

R 7 is C1-C3 alkyl;

R 8 is C1-C3 alkyl; and

R 9 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl

X e and X* are, independently, N or CH; and

R 12 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, or optionally substituted 3 to 6-membered heterocycloalkylene.

In some embodiments of a compound of the present invention, X ® is N and X* is CH. In some embodiments, X ® is CH and X· is N. In some embodiments of a compound of the present invention, R 12 is optionally substituted Ci-Ce

OMe heteroalkyl. In some embodiments, R' 2 is » l

CH 3

Y^OCHFz

. In some embodiments, R 12 is V^OMe

In some embodiments, a compound of the present invention has the structure of Formula VI, or a pharmaceutically acceptable salt thereof:

Formula VI wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH3)C(0)-(CH2)- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene (e.g., phenyl or phenol), or optionally substituted 5 to 10-membered heteroarylene;

B is absent, -CH(R 9 )-, >C=CR 9 R 9 ', or >CR 9 R 9' where the carbon is bound to the carbonyl carbon of -N(R 11 )C(0)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted Ci-C* alkylene, optionally substituted C1-C4 alkenylene, optionally substituted C1-C4 heteroalkylene, -C(0)0-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(0)NH-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C1-C4 heteroalkylene, or 3 to 8-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a vinyl ketone, a vinyl sulfone, an ynone, a haloacetal, or an alkynyl sulfone;

X 1 is optionally substituted C1-C2 alkylene, NR, O, or S(0) n ;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2; R is hydrogen, cyano, optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, optionally substituted C2-C4 alkynyl, C(0)R’, C(0)0R', C(0)N(R’) 2 , S(0)R', S(0) 2 R', or S(0) 2 N(R') 2 ; each R ' is, independently, H or optionally substituted C1-C4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 is CH, CH 2 , or N;

Y 6 is C(O), CH, CH 2 , or N;

R 2 is absent, hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted C 2 -Ce alkenyl, optionally substituted C 2 -Ce alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C1-C4 alkyl optionally substituted with halogen, cyano, hydroxy, or C1-C4 alkoxy, cyclopropyl, or cyclobutyl;

R® is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C1-C 3 alkyl, or R® and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R® is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted C 2 -Ce alkenyl, optionally substituted C^Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R® combine with the carbon atom to which they are attached to form C=CR r R ® ’; C=N(OH), C=N(0-Ci-Cs alkyl), C=0, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7a and R are, independently, hydrogen, halo, optionally substituted C1-C3 alkyl, or combine with the carbon to which they are attached to form a carbonyl;

R T is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R ®' is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted Cz-Ce alkenyl, optionally substituted Ci!-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R T and R ® ’ combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is H, F, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; or

R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 9' is hydrogen or optionally substituted Ci-Ce alkyl; or

R 9 and R 9 , combined with the atoms to which they are attached, form a 3 to 6-membered cycloalkyl or a 3 to 6-membered heterocycloalkyl; R 10 is hydrogen, halo, hydroxy, C1-C3 alkoxy, or C1-C3 alkyl;

R 10a is hydrogen or halo;

R 1 ' is hydrogen or C1-C3 alkyl;

R 2 ' is hydrogen or C1-C3 alkyl (e.g., methyl); and X ® and X* are, independently, N or CH.

In some embodiments, a compound of the present invention has the structure of Formula Via, or a pharmaceutically acceptable salt thereof:

Formula Via wherein A optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene (e.g., phenyl or phenol), or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a vinyl ketone, a vinyl sulfone, an ynone, or an alkynyl sulfone;

X 1 is optionally substituted C1-C2 alkylene, NR, O, or S(0) n ;

X 2 is O or NH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, optionally substituted C2-C4 alkynyl, C(0)R’, C(0)0R\ C(0)N(R’)2, S(0)R’, S(0) 2 R’, or S(0)2N(R’)2; each R ' is, independently, H or optionally substituted C1-C4 alkyl;

R 2 is Ci-Ce alkyl, Ci-Ce fluoroalkyl, or 3 to 6-membered cycloalkyl;

R 7 is C1-C3 alkyl;

R® is C1-C3 alkyl; and

R 9 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; X ® and X* are, independently, N or CH;

R 11 is hydrogen or C1-C3 alkyl; and R 21 is hydrogen or C1-C3 alkyl. In some embodiments of a compound of the present invention, X ® is N and X* is CH. In some embodiments, X ® is CH and X* is N.

In some embodiments, a compound of the present invention has the structure of Formula Vlb, or a pharmaceutically acceptable salt thereof:

Formula Vlb wherein A optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene (e.g., phenyl or phenol), or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

R 9 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

L is absent or a linker; and

W is a cross-linking group comprising a vinyl ketone, a vinyl sulfone, an ynone, or an alkynyl sulfone.ln some embodiments of a compound of the present invention, A is optionally substituted 6- membered arylene.

In some embodiments, a compound of the present invention has the structure of Formula Vic (corresponding for Formula BB of FIG. 1 A and FIG. 1 B), or a pharmaceutically acceptable salt thereof:

Formula Vic wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH3)C(0)-(CH2)- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene (e.g., phenyl or phenol), or optionally substituted 5 to 10-membered heteroarylene;

B is absent, -CH(R 9 )-, >C=CR 9 R 9' , or >CR 9 R 9 where the carbon is bound to the carbonyl carbon of -N(R 11 )C(0)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C1-C4 alkylene, optionally substituted C1-C4 alkenylene, optionally substituted C1-C4 heteroalkylene, -C(0)0-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(0)NH-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C1-C4 heteroalkylene, or 3 to 8-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a vinyl ketone, a vinyl sulfone, an ynone, a haloacetal, or an alkynyl sulfone;

X 1 is optionally substituted C1-C2 alkylene, NR, O, or S(0) n ;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, optionally substituted C2-C4 alkynyl, C(0)R’, C(0)0R', C(0)N(R')2, S(0)R', S(0) 2 R', or S(0)2N(R') 2 ; each R’ is, independently, H or optionally substituted C1-C4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 is CH, CH2, or N;

Y® is C(O), CH, CH2, or N;

R 2 is absent, hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C1-C4 alkyl optionally substituted with halogen, cyano, hydroxy, or C1-C4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl, or R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted Cz-Ce alkenyl, optionally substituted Cz-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R B combine with the carbon atom to which they are attached to form C=CR 7 R 8 '; C=N(OH), C=N(0-CI-C3 alkyl), C=0, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7a and R 8 * are, independently, hydrogen, halo, optionally substituted C1-C3 alkyl, or combine with the carbon to which they are attached to form a carbonyl;

R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl; R 8 ' is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted Cz-Ce alkenyl, optionally substituted Cz-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 ’ combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is H, F, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; or

R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl ;

R 9' is hydrogen or optionally substituted Ci-Ce alkyl; or

R 9 and R 9 , combined with the atoms to which they are attached, form a 3 to 6-membered cycloalkyl or a 3 to 6-membered heterocycloalkyl;

R 10 is hydrogen, halo, hydroxy, C1-C3 alkoxy, or C1-C3 alkyl;

R 10a is hydrogen or halo;

R 11 is hydrogen or C1-C3 alkyl; and R 21 is hydrogen or C1-C3 alkyl (e.g., methyl).

In some embodiments, A has the structure: wherein R 13 is hydrogen, halo, hydroxy, amino, optionally substituted Ci-Ce alkyl, or optionally substituted Ci-Ce heteroalkyl; and R ,3a is hydrogen or halo. In some embodiments, R 13 is hydrogen. In some embodiments, R 13 and R 13a are each hydrogen. In some embodiments, R 13 is hydroxy, methyl, fluoro, or difluoromethyl. In some embodiments, A is optionally substituted 5 to 6-membered heteroarylene. In some

In some embodiments, A is optionally substituted Ci-C< heteroalkylene. In some embodiments, A

O

/AA n A

I is: ^^3 . in some embodiments, A is optionally substituted 3 to 6-membered heterocycloalkylene. In some embodiments, A is:

*0* AQA ¾ £ Α,ί^Α A^jS o X

A N A

A^XA ANAA O k/O

. or c . In some embodiments, A is

In some embodiments of a compound of the present invention, B is -CHR 9 -. In some embodiments, R 9 is H, F, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl. In some embodiments, R 9 is: A F

AY' cf 3 Αγ" CF 3

Ao' CHa CH 3 CF 3 I J I J A3 J I

V^CH 3 ¾

I A X /0 vQ rV 0° . In some embodiments, R 9 is:

CH 3

V^CK 3 . In some embodiments, R 9 is H, optionally substituted Ci-Ce alkyl, optionally substituted Ci- Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7- membered heterocycloalkyl.

In some embodiments of a compound of the present invention, B is optionally substituted 6- membered arylene. In some embodiments, B is 6-membered arylene. In some embodiments, B is:

In some embodiments of a compound of the present invention, R 7 is methyl.

In some embodiments of a compound of the present invention, R 8 is methyl.

In some embodiments, R 21 is hydrogen.

In some embodiments of a compound of the present invention, the linker is the structure of Formula II:

A 1 -(B ' )t-(C 1 )g-(B 2 )h-(D ' )-(B 3 )i-(C 2 )j-(B 4 )k— A 2 Formula II where A 1 is a bond between the linker and B; A 2 is a bond between W and the linker; B 1 , B 2 , B 3 , and B 4 each, independently, is selected from optionally substituted C1-C2 alkylene, optionally substituted C1-C3 heteroalkylene, O, S, and NR N ; R N is hydrogen, optionally substituted Ci-t alkyl, optionally substituted C2-C4 alkenyl, optionally substituted C2-C4 alkynyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted C1-C7 heteroalkyl; C and C 2 are each, independently, selected from carbonyl, thiocarbonyl, sulphonyl, or phosphoryl; f, g, h, i, j, and k are each, independently, 0 or 1 ; and D' is optionally substituted C1-C10 alkylene, optionally substituted C2-C10 alkenylene, optionally substituted C2-C10 alkynylene, optionally substituted 3 to 14- membered heterocycloalkylene, optionally substituted 5 to 10-membered heteroarylene, optionally substituted 3 to 8-membered cycloalkylene, optionally substituted 6 to 10-membered arylene, optionally substituted C2-C10 polyethylene glycolene, or optionally substituted C1-C10 heteroalkylene, or a chemical bond linking A 1 -(B 1 )t-(C 1 ) g -(B 2 )h- to -(B 3 )i-(C 2 )j-(B 4 )n-A 2 . In some embodiments, the linker is acyclic. In some embodiments, linker has the structure of Formula lla:

R 14

Formula lla wherein X a is absent or N; R 14 is absent, hydrogen or optionally substituted Ci-Ce alkyl; and L 2 is absent, -SO2-, optionally substituted C1-C4 alkylene or optionally substituted C1-C4 heteroalkylene, wherein at least one of X a , R 14 , or L 2 is present. In some embodiments, the linker has the structure:

In some embodiments, the linker is or comprises a cyclic moiety. In some embodiments, the linker has the structure of Formula lib:

Formula Mb wherein o is 0 or 1 ;

R 15 is hydrogen or optionally substituted Ci-Ce alkyl, optionally substituted 3 to 8-membered cycloalkylene, or optionally substituted 3 to 8-membered heterocycloalkylene ;

X 4 is absent, optionally substituted C1-C4 alkylene, O, NCH3, or optionally substituted C1-C4 heteroalkylene;

Cy is optionally substituted 3 to 8-membered cycloalky lene, optionally substituted 3 to 8- membered heterocycloalkylene, optionally substituted 6-10 membered arylene, or optionally substituted 5 to 10-membered heteroarylene; and

L 3 is absent, -SO2-, optionally substituted Ci-C* alkylene or optionally substituted C1-C4 heteroalky lene.

. In some embodiments, the linker has the structure of Formula lib-1 :

Formula llb-1 wherein o is 0 or 1 ;

R 15 is hydrogen or optionally substituted Ci-Ce alkyl, optionally substituted 3 to 8-membered cycloalkylene, or optionally substituted 3 to 8-membered heterocycloalkylene ;

Cy is optionally substituted 3 to 8-membered cycloalkylene, optionally substituted 3 to 8- membered heterocycloalkylene, optionally substituted 6-10 membered arylene, or optionally substituted 5 to 10-membered heteroarylene; and

L 3 is absent, -S02-, optionally substituted C1-C4 alkylene or optionally substituted C1-C4 heteroalkylene. In some embodiments, the linker has the structure of Formula lie:

Formula lie wherein R 15 is hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted 3 to 8- membered cycloalkylene, or optionally substituted 3 to 8-membered heterocycloalky lene; and

R 1Sa , R ,Sb , R 15c , R 15d , R 15e , R 19 , and R 15 o are, independently, hydrogen, halo, hydroxy, cyano, amino, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce alkoxy, or , or R ,5b and R ,5d combine with the carbons to which they are attached to form an optionally substituted 3 to 8-membered cycloalkylene, or optionally substituted 3 to 8-membered heterocycloalkylene.

In some embodiments, the linker has the structure:

. or o

In some embodiments, the linker has the structure:

X

N

N

Y

In some embodiments, the linker has the structure o

CH3 N" X

In some embodiments, the linker has the structure .In some embodiments of a compound of the present invention, W is a cross-linking group comprising a vinyl ketone. In some embodiments, W has the structure of Formula Ilia:

Formula Ilia wherein R 16a , R 16b , and R 16c are, independently, hydrogen, -CN, halogen, or -C1-C3 alkyl optionally substituted with one or more substituents independently selected from -OH, -O-C1-C3 alkyl, -IMH2, -NH(Ci-Cs alkyl), -N(CI-C3 alkyl)2, or a 4 to 7-membered saturated heterocycloalkyl. In some embodiments, W is:

O

O O O

. In some embodiments, W is a cross- linking group comprising an ynone. In some embodiments, W has the structure of Formula lllb: o

R 17

Formula lllb wherein R 17 is hydrogen, -C1-C3 alkyl optionally substituted with one or more substituents independently selected from -OH, -O-C1-C3 alkyl, -NH2, -NH(CI-C3 alkyl), -N(CI-C3 alkyl)2, or a 4 to 7- membered saturated heterocycloalkyl, or a 4 to 7-membered saturated heterocycloalkyl. In some embodiments, W is:

In some embodiments, W is a cross-linking group comprising a vinyl sulfone. In some embodiments, W has the structure of Formula lllc:

Rite Formula lllc wherein R 18a , R ,8b , and R 18c are, independently, hydrogen, -CN, or -C1-C3 alkyl optionally substituted with one or more substituents independently selected from -OH, -O-C1-C3 alkyl,

-NHa, -NH(CI-C3 alkyl), -N(CI-C3 alkyl>2, or a 4 to 7-membered saturated heterocycloalkyl. In some embodiments, W is: In some embodiments, W is a cross-linking group comprising an alkynyl sulfone. In some embodiments, W has the structure of Formula Hid:

%t°

His

Formula Mid wherein R 19 is hydrogen, -C1-C3 alkyl optionally substituted with one or more substituents independently selected from -OH, -O-C1-C3 alkyl, -IMH2, -NH(CI-C3 alkyl), -N(CI-C3 alkyl>2, or a 4 to 7- membered saturated heterocycloalkyl, or a 4 to 7-membered saturated heterocycloalkyl. In some embodiments, W is: In some embodiments, W has the structure of Formula llle:

O vV

Formula llle wherein X ® is a halogen; and

R 20 is hydrogen, -C1-C3 alkyl optionally substituted with one or more substituents independently selected from -OH, -O-C1-C3 alkyl, -NH2, -NH(CI-C3 alkyl), -N(CI-C3 alkyl>2, or a 4 to 7-membered saturated heterocycloalkyl. In some embodiments, W is haloacetal. In some embodiments, W is not haloacetal.

In some embodiments, a compound of the present invention is selected from Table 1 , or a pharmaceutically acceptable salt or stereoisomer thereof. In some embodiments, a compound of the present invention is selected from Table 1 , or a pharmaceutically acceptable salt or atropisomer thereof.

Table 1 : Certain Compounds of the Present Invention

Ex# Structure Ex# Structure

V

A M t JO

A5 ¾

‘OH Ex# Structure Ex# Structure Ex# Structure Ex# Structure Ex# Structure

° rCv° 0 V

,0 H .0

A28 q

6 OH Ex# Structure Ex# Structure

V

M

A39

‘OH

O

°Y N-

Ja H

A42 -o. ¼ " ° Ex# Structure Ex# Structure Ex# Structure Ex# Structure Ex# Structure Ex# Structure Ex# Structure Ex# Structure Ex# Structure Ex# Structure

0. r

A88

‘OH to V

J H z

A92

‘OH Ex# Structure Ex# Structure

V

A M JO

A100 ¾

‘OH Ex# Structure Ex# Structure

,N.

,0 H

A110 MeO

OH

<s Ex# Structure Ex# Structure Ex# Structure v W" w.

M

A123 H o k0H k Ex# Structure Ex# Structure

°r w. W"

A133 H JJO o k Ex# Structure

N o.

A140 H o q. λ

0 H r

A142

‘OH Ex# Structure Ex# Structure

0 Y ,0 H 0v

A149 -o. :¾

‘OH

N" r

°γΧ

„o H r

A152 Ex# Structure Ex# Structure

¥

A159 -q

‘OH Ex# Structure Ex# Structure

A168 Ex# Structure Ex# Structure

Ex# Structure Ex# Structure Ex# Structure v ow O v

,0 H . oc .0

A193 r

OH

N Ex# Structure Ex# Structure Ex# Structure Ex# Structure Ex# Structure

H

.o o /

X O I « .N >5 « O I

H o

A218 l¾X OH

N*

O

°γΟ

\

O i H

A219 =W >

OH

N'

I

X °γ\/γ O , o J H I N 1 J O I

H

A220 o

/T% OH

N

O, o v O

\

O I H Jj y Xj

A221 Λ *

‘OH

N· t

°ΎΌ*

\ ,o

A222 ,° r ¾9oc

ΌΗ

N 1 <5- i Ex# Structure Ex# Structure Ex# Structure Ex# Structure Ex# Structure Ex# Structure

O, vOy° o •N

H 11 I o H

A248 *1, S ^N

6.

N /

//

°y Cv o

1 H 11 I o *LN

N

H

A249 'b.

&1

IH o u

H

%,·θ N

H V> o

ΆΝ.

N

H

A250 o

&1

ΌΗ

41 N

H vCv o N'

H ii ti-N, I

N N'

H

A251 11

» ~\-J ΌΗ

41 N

<x

H

N vOy° o

H 11 I ΊΡ*

11-N si O

N

¾'

A252 11

N-< y // \—p 1 ΌΗ Ex# Structure Ex# Structure Ex# Structure

° γ Ογ°ο . I I

A264 ί -M x y^-

41 I

// ν_/ητ ΌΗ

VW « I ?

A265 41 t ' M ΌΗ rCf'

OKJ

O,

T N r

H ii \

A266 'b

41 ■W 3

</ v-fl iH vOv 0 0 O

H 41 N·

N il^N.

H o /

A267 41, \

N-K A // 1 IH Ex# Structure Ex# Structure Ex# Structure Ex# Structure Ex# Structure Ex# Structure Ex# Structure Ex# Structure Ex# Structure Ex# Structure Ex# Structure Ex# Structure

>·0

O H ^°ο ,0

41 ,Ν·

N 11, N

H

A324 0

11 %

Fx —N

1-1J \

N y^L .

H

.0 n 8 41, N I

N 04

</ H

A325 &1

» V ¾_1/

O. »·· Q V N', - °N>^x ,

O H 11 »i,N I

N

H ,0 o

A326 c/

11

11

N o,

° v Q y o r-N

D

A327 F,

41 j¾6

11 N

O,

<¾ V Oi r-N

11,N I

H ,P

A328 41 1

(/ V-f E

O / Ex# Structure Ex# Structure Ex# Structure Ex# Structure

O

O .o o

J H o'

A344 mo. m er. Ex# Structure

A350

<<

A353 y Ex# Structure

A3S5 >o m

N-

M" i

<< Ex# Structure

•O

A362

<< Ex# Structure

.0 o

J M

A365 4 <>

<< Ex# Structure Ex# Structure Ex# Structure

A382 Ex# Structure

A384

A387 Ex# Structure

A390

'Y 0 v Ex# Structure Ex# Structure

A400 Ex# Structure Ex# Structure vOv o

J H

A412

<< Ex# Structure

A415 A Ex# Structure Ex# Structure Ex# Structure

°H

A429

KJ Ex# Structure Ex# Structure Ex# Structure

,N.

Ϊ

A444 Ex# Structure

0 vCv o

J H JU 0'

A453 wo m

K cr. Ex# Structure Ex# Structure Ex# Structure

CL 0

°γ·" - i H

A467 °x"

<x Ex# Structure

,o 0 Ή

J H 9 0 /

A46 m

O

0

A473 Ex# Structure

>

A477

>

A478 »

<v Ex# Structure Ex# Structure q.

.0

V; o

.0

A486 m Ex# Structure

A490 m

<< Ex# Structure Ex# Structure Ex# Structure Ex# Structure

A512 k Ex# Structure o,

, 0 H

Ml

A514 1 4V° i N· Ex# Structure Ex# Structure Ex# Structure

°fr

A529 -V· Ex# Structure

A536

‘OH

_v

Ex# Structure Ex# Structure

Ο y >

Α547

Ex# Structure

γψ

A554 4 Ex# Structure

Ύ QΊΤ

A557

A559 »

<< Ex# Structure

A561 k

A564 k Ex# Structure

<>

A565

<<

Ex# Structure Ex# Structure

A577

Ex# Structure

A581 iff "*

<< Ex# Structure Ex# Structure Ex# Structure

A596 k

A597 k

A599 ■ Ύ Ex# Structure

“V

TTY

A603 Ex# Structure Ex# Structure cv.

A613 ■

Ex# Structure

A617 k

A618 Ex# Structure

A620

<<

A622 k

A624

A k Ex# Structure

A629 Ex# Structure

A631

°r

A632

V 2¾

A634 Ex# Structure Ex# Structure

A644

<< Ex# Structure

A647 m

<<

A648 m

<< Ex# Structure

T QY

A652 °v. k

A653 <- k Ex# Structure

A655 Ex# Structure

.0

A661

Ex# Structure Ex# Structure Ex# Structure

.o o

' ΐ Μ

A675 X

<< Ex# Structure o

Ov. Cr

A682 4

<v

Ex# Structure

A686

A687

Ex# Structure

N o

A692

Ex# Structure

Y9

A699

Ex# Structure Ex# Structure

A707 4 k

°Y

A708 m Ex# Structure

A711

A714 m

<< Ex# Structure Ex# Structure

A722 3% V

MO

A723 m fV

CF,

Ex# Structure

.O

A725

CP.

A726 Ex# Structure

,0 v¾ O

A731 m Ex# Structure

A739 k Ex# Structure

V O K N y°

1 H ff I

N

M if JO

A740 eQ N H O

* vV

N

O V* ,0 n o H v O

A741 MeQ vM

O

N'

Note that some compounds are shown with bonds as flat or wedged. In some instances, the relative stereochemistry of stereoisomers has been determined; in some instances, the absolute stereochemistry has been determined. In some instances, a single Example number corresponds to a mixture of stereoisomers. All stereoisomers of the compounds of the foregoing table are contemplated by the present invention. In particular embodiments, an atropisomer of a compound of the foregoing table is contemplated.

Brackets are to be ignored.

*The activity of this stereoisomer may, in fact, be attributable to the presence of a small amount of the stereoisomer with the (S) configuration at the -NC(0)-£H(CH 3 )2-N(CH 3 )- position.

In some embodiments, a compound of Table 2 is provided, or a pharmaceutically acceptable salt thereof. In some embodiments, a compound of the present invention is selected from Table 2, or a pharmaceutically acceptable salt or atropisomer thereof. Table 2: Certain Compounds of the Present Invention

Ex# Structure EX# Structure EX# Structure vO i H 0 J

Bll MeO.

ΌΗ

N- EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure

W r-NH

¾ N I a ...o

B107 MeO

(/

N 1

<<

\

Oi - VN —f-

O

B109 MeO /=¾

(/

N

/

\

O V -N -f*

Hv° o

,o I

,o

Bill Mel a

N^S 1

N-

(/

N

/

'"0v° O ,-N ^·

.O

N N

B112 Mi H ..o

N^ .S o

N-

(/

N

/

O,

O r-N

I H O

O N

N ,o

B113 Mei H

N

(t

N

/ EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure o. o, .·· O H 11

Λ1.Ν

N

■Q H

B179 41 IL

<t V-/ 1 k o,

O; V O r-N

O H « N ALN P

— o H

B180 11 IL

'/ V ii- \f _ I 11I x -

N

O,

O; 11

.*·· N

H O

11 ALN.

N V &

H O1

B181 &1 k N o. v I O H ,y/ o » O

B182 11’ N^ S m

» V-/

41 N I 4& k " EX# Structure EX# Structure o.

T N •N

H 4i

1

8189 — o

8,1 ■¾v

N.

(/ v_/

41 N k EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure

O

»>· O o I r-H

O H " 1LN

N 0 n γΡ

B30 •O

11

" N

X O,

— q

B301 *1

¥ N ^ ¾1

O z X o.

O; V y

H it

11.N

N

H ,P q

B302 41 > I n ¾t B

O / X

P

VVv 0 o -N

$1,N *’·

B303 — Q H O

11 zs

¾1 B

< < o

Y¾V 5 I N

B304 — Q H u

P X t-iJ

N i EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure EX# Structure o vO H 4v1 ’o fi-N. cry

¾ o / N

B353 \

41

(TV/

11 'N

O

VW H &1 >o

S,N.

B354 — Q O / N s

41

11 'N

<x EX# Structure

756

SUBSTITUTE SHEET (RULE 26) EX# Structure EX# Structure

758

SUBSTITUTE SHEET (RULE 26) EX# Structure ο.

T Ν r

Η 11 9 11. Ν I

Ν D

Η

Β370 — ο ,,Ν.

11 Ο

L //

#\i

41 Ν

Ο,

Ο; r-N

Η 11 9 ιι.Ν I

Q ¾ ' Ρ

B371 41 Ν I ' Ο

Ν />

41 Ν κ

Ο,

Ο; ip,

- Α Ν' Ν r

Η ιι ι I - N ι.Ν

H ' ,·Ρ

Β372 Q

41 Ν ι >

41 Ν

Κ

0,

<vO r-N

Η 1v1 > ο

11. Ν

Ν ,·Ρ

Β373 Q Η ϊ

11 Ν ^Ν

* i

41 Ν κ

0, ο r-N

Η 11 11.N

— Ή Ύ 4 Ρ

Β374 ο ,,Ν. ο

41 ! //

41 Ν

Κ

759

SUBSTITUTE SHEET (RULE 26) EX# Structure

760

SUBSTITUTE SHEET (RULE 26) WO 2021/257736 PCT/US2021/037679

EX# Structure

O, vOy •N

H 41 °o I

N Y 1 1

H

B380 -Q o

41

41

N c¾ > .·Ό

H 41

B381 -Q

41 r41

T N r

H 41

- J wo-C

B382 O.

41

'41 vOy° o ,o

H 61

N^-'N

H

B383 -Q

41 fXJ

¾i

•o

°vO

H M fs k

B384 -Q

41 H P r41 <?

<v

761

SUBSTITUTE SHEET (RULE 26) EX# Structure

762

SUBSTITUTE SHEET (RULE 26) EX# Structure

763

SUBSTITUTE SHEET (RULE 26) EX# Structure

764

SUBSTITUTE SHEET (RULE 26) EX# Structure

765

SUBSTITUTE SHEET (RULE 26) EX# Structure a V Q

V N' ,0 >1 ^

H O

11 11 N I N-

N ¾1

H

B406 Q O

11 rw ¾1

<vO .0 O in \\

H 41 1L I N·

N N 11

H

B407 -Q O

41

P ' V r n-/

HQ

\

O- .·· O

H ii

B408 Q ¾w

41

P X 1-1J

N

N, vOy .11 \\

H ii 0 o

11-N I N-

N 11

H

B409 &i

P x '1^1r

766

SUBSTITUTE SHEET (RULE 26) EX# Structure

0v° O \\

O H 11 11 N N-

N « in

H

B410 Q

11

* v_z

41 'N k

°γΟγ° o \\ .o H " 11- N I N·

N n in

-Q H

B411 11

11

N k

HO

%"0 11

H ii O

N Ά,Ν

H 11

B412 -Q O //

11 !

F WS^-fN k

V.C" \\

H 11 e fl·

N o 41

H

B413 O

&1

P \ r 4-1J k

\\

H 11 9 ii, YN N-

N 11

H

B414 41 rl1

767

SUBSTITUTE SHEET (RULE 26) EX# Structure

768

SUBSTITUTE SHEET (RULE 26) EX# Structure

769

SUBSTITUTE SHEET (RULE 26) EX# Structure

Note that some compounds are shown with bonds as flat or wedged. In some instances, the relative stereochemistry of stereoisomers has been determined; in some instances, the absolute stereochemistry has been determined. All stereoisomers of the compounds of the foregoing table are contemplated by the present invention. In particular embodiments, an atropisomer of a compound of the foregoing table is 5 contemplated.

In some embodiments, a compound of the present invention is or acts as a prodrug, such as with respect to administration to a cell or to a subject in need thereof.

Also provided are pharmaceutical compositions comprising a compound of the present invention, 10 or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.

Further provided is a conjugate, or salt thereof, comprising the structure of Formula IV:

M-L-P Formula IV wherein L is a linker;

15 P is a monovalent organic moiety; and M has the structure of Formula Va:

Formula Va wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

20 A is -N(H or CH3)C(0)-(CH2)- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene;

B is absent, -CH(R 9 )-, >C=CR 9 R 9 ’, or >CR 9 R 9' where the carbon is bound to the carbonyl carbon 25 of -N(R 11 )C(0)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-

770

SUBSTITUTE SHEET (RULE 26) membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C1-C4 alkylene, optionally substituted C1-C4 alkenylene, optionally substituted C1-C4 heteroalkylene, -C(0)0-CH(R®)- where C is bound to -C(R 7 R®)-, -C(0)NH-CH(R 6 )- 5 where C is bound to -C(R 7 R®)-, optionally substituted C1-C4 heteroalkylene, or 3 to 8-membered heteroarylene;

X 1 is optionally substituted C1-C2 alkylene, NR, O, or S(0) n ;

X 2 is O or NH;

X 3 is N or CH;

10 n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, optionally substituted C2-C4 alkynyl, C(0)R’, C(0)0R’, C(0)N(R') 2 , S(0)R’, S(0) 2 R', or S(0) 2 N(R’) 2 ; each R’ is, independently, H or optionally substituted C1-C4 alkyl;

Y 1 is C, CH, or N;

15 Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 is CH, CH 2 , or N;

Y 6 is C(O), CH, CH 2 , or N;

R 1 is cyano, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally 20 substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl, or

R 1 and R 2 combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 2 is absent, hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted C 2 -Ce alkenyl,

25 optionally substituted C 2 -Ce alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

30 R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C1-C4 alkyl optionally substituted with halogen, cyano, hydroxy, or C1-C4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl, or R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally 35 substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R® is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted C 2 -Ce alkenyl, optionally substituted Ca-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

40 R 7 and R® combine with the carbon atom to which they are attached to form C=CR 7' ' ; C=N(OH), C=N(0-CI-C3 alkyl), C=0, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

771

SUBSTITUTE SHEET (RULE 26) R 7a and R are, independently, hydrogen, halo, optionally substituted C1-C3 alkyl, or combine with the carbon to which they are attached to form a carbonyl;

R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted Ca-Ce 5 alkenyl, optionally substituted Cz-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 ’ combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

10 R 9 is H, optionally substituted Ci-Ce alkyl, optionally substituted CvCe heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl, or

R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 9 ' is hydrogen or optionally substituted Ci-Ce alkyl; or

15 R 9 and R 9 ', combined with the atoms to which they are attached, form a 3 to 6-membered cycloalkyl or a 3 to 6-membered heterocycloalkyl;

R 10 is hydrogen, halo, hydroxy, C1-C3 alkoxy, or C1-C3 alkyl;

R i °a i s hydrogen or halo; and R 11 is hydrogen or C1-C3 alkyl.

20 In some embodiments the conjugate, or salt thereof, comprises the structure of Formula IV:

M-L-P Formula IV wherein L is a linker;

P is a monovalent organic moiety; and

25 M has the structure of Formula Vb:

Formula Vb wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH3)C(0)-(CH2)- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, 30 optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalky!ene, optionally substituted 6-membered ary!ene, or optionally substituted 5 to 6- membered heteroarylene;

772

SUBSTITUTE SHEET (RULE 26) B is -CH(R 9 )- or >C=CR 9 R 9 where the carbon is bound to the carbonyl carbon of -N(R 11 )C(0)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted Ci-C* alkylene, optionally substituted C1-C4 alkenylene, optionally 5 substituted C1-C4 heteroalkylene, -C(0)0-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(0)NH-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C1-C4 heteroalkylene, or 3 to 8-membered heteroarylene;

X 1 is optionally substituted C1-C2 alkylene, NR, O, or S(0) n ;

X 2 is O or NH;

10 X 3 is N or CH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, optionally substituted C2-C4 alkynyl, C(0)R’, C(0)0R’, C(0)N(R') 2 , S(0)R’, S(0) 2 R', or S(0) 2 N(R’) 2 ; each R’ is, independently, H or optionally substituted C1-C4 alkyl;

15 Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 is CH, CH 2 , or N;

Y 8 is C(O), CH, CH 2 , or N;

R 1 is cyano, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally 20 substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl, or

R 1 and R 2 combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

25 R 2 is absent, hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted C 2 -Ce alkenyl, optionally substituted C 2 -Ce alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 30 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C1-C4 alkyl optionally substituted with halogen, cyano, hydroxy, or C1-C4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl, or

35 R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted C 2 -Ce alkenyl, optionally substituted Ca-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl,

40 optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

773

SUBSTITUTE SHEET (RULE 26) R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7 R 8 ; C=N(OH), C=N(0-Ci-Ca alkyl), C=0, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7a and R are, independently, hydrogen, halo, optionally substituted C1-C3 alkyl, or combine 5 with the carbon to which they are attached to form a carbonyl;

R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or 10 optionally substituted 6 to 10-membered aryl, or

R 7 and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl, or

15 R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 9' is hydrogen or optionally substituted Ci-Ce alkyl;

R 10 is hydrogen, halo, hydroxy, C1-C3 alkoxy, or C1-C3 alkyl;

R i °a j s hydrogen or halo; and

20 R 11 is hydrogen or C1-C3 alkyl.

In some embodiments, the conjugate has the structure of Formula IV:

M-L-P Formula IV wherein L is a linker;

25 P is a monovalent organic moiety; and M has the structure of Formula Vc:

Formula Vc wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

30 A is -N(H or CH3)C(0)-(CH2)- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered

774

SUBSTITUTE SHEET (RULE 26) heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -N(R 11 )C(0)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, 5 optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C1-C4 alkylene, optionally substituted C1-C4 alkenylene, optionally substituted C1-C4 heteroalkylene, -C(0)0-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(0)NH-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C1-C4 heteroalkylene, or 3 to 8-membered heteroarylene;

10 X 1 is optionally substituted C1-C2 alkylene, NR, O, or S(0) n ;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl,

15 optionally substituted C2-C4 alkynyl, C(0)R’, C(0)OR’, C(0)N(R') 2 , S(0)R’, S(0) 2 R', or S(0) 2 N(R’) 2 ; each R’ is, independently, H or optionally substituted C1-C4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 and Y 6 are, independently, CH or N;

20 R 1 is cyano, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl;

R 2 is hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted Cz-Ce alkenyl, optionally 25 substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

30 R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C1-C4 alkyl optionally substituted with halogen, cyano, hydroxy, or C1-C4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl, or R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally 35 substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted C 2 -Ce alkenyl, optionally substituted Ca-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

40 R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(0-CI-C3 alkyl), C=0, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

775

SUBSTITUTE SHEET (RULE 26) R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or 5 optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 ’ combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

10 R 10 is hydrogen, hydroxy, C1-C3 alkoxy, or C1-C3 alkyl; and R 11 is hydrogen or C1-C3 alkyl.

In some embodiments, a compound of the present invention has the structure of of Formula IV:

M-L-P Formula IV

15 wherein L is a linker;

P is a monovalent organic moiety; and M has the structure of Formula Vd:

Formula Vd

20 wherein A optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene (e.g., phenyl or phenol), or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, 25 optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

X 1 is optionally substituted C1-C2 alkylene, NR, O, or S(0) n ;

X 2 is O or NH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl,

30 optionally substituted C2-C4 alkynyl, C(0)R\ C(0)0R', C(0)N(R’)2, S(0)R', S(0) 2 R', or S(0)2N(R') 2 ; each R ' is, independently, H or optionally substituted C1-C4 alkyl;

R 2 is Ci-Ce alkyl, Ci-Ce fiuoroalkyl, or 3 to 6-membered cycloalkyl;

776

SUBSTITUTE SHEET (RULE 26) R 7 is C1-C3 alkyl;

R 8 is C1-C3 alkyl; and

R 9 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

5 X ® and X* are, independently, N or CH;

R 1 ' is hydrogen or C1-C3 alkyl; and R 2 ' is hydrogen or C1-C3 alkyl.

In some embodiments of a compound of the present invention, X ® is N and X* is CH. In some embodiments, X ® is CH and X* is N.

10 In some embodiments, a compound of the present invention has the structure of of Formula IV:

M-L-P Formula IV wherein L is a linker;

P is a monovalent organic moiety; and

15 M has the structure of Formula Ve:

Formula Ve wherein A is optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene (e.g., phenyl or phenol), or 20 optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene; and

R 9 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally 25 substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl.

In some embodiments of a conjugate of the present invention, the linker has the structure of

Formula II:

A 1 -(B 1 )f-(C 1 )g-(B 2 )h-(D 1 )-(B 3 )i-(C 2 )j-(B 4 )k— A 2 Formula II

30 where A 1 is a bond between the linker and B; A 2 is a bond between P and the linker; B 1 , B 2 , B 3 , and B 4 each, independently, is selected from optionally substituted C1-C2 alkylene, optionally substituted

777

SUBSTITUTE SHEET (RULE 26) C1-C3 heteroalkylene, O, S, and NR N ; R N is hydrogen, optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, optionally substituted C2-C4 alkynyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted C1-C7 heteroalkyl; C 1 and C 2 are each, independently, selected from carbonyl, thiocarbonyl, sulphonyl, or phosphoryl; f, g, h, 5 I, j, and k are each, independently, 0 or 1 ; and D 1 is optionally substituted C1-C10 alkylene, optionally substituted C2-C10 alkenylene, optionally substituted C2-C10 alkynylene, optionally substituted 3 to 14- membered heterocycloalkylene, optionally substituted 5 to 10-membered heteroarylene, optionally substituted 3 to 8-membered cycloalkylene, optionally substituted 6 to 10-membered arylene, optionally substituted C2-C10 polyethylene glycolene, or optionally substituted C1-C10 heteroalkylene, or a chemical 10 bond linking A 1 -(B 1 ) f -(C')g-(B 2 )h- to -(B 3 )i-(C 2 )j-(B 4 )ic-A 2 .

In some embodiments of a conjugate of the present invention, the monovalent organic moiety is a protein, such as a Ras protein. In some embodiments, the Ras protein is K-Ras G12C, K-Ras G13C, H- Ras G12C, H-Ras G13C, N-Ras G12C, or N-Ras G13C. Other Ras proteins are described herein. In some embodiments, the linker is bound to the monovalent organic moiety through a bond to a sulfhydryl 15 group of an amino acid residue of the monovalent organic moiety. In some embodiments, the linker is bound to the monovalent organic moiety through a bond to a carboxyl group of an amino acid residue of the monovalent organic moiety.

Further provided is a method of treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of the present 20 invention, or a pharmaceutically acceptable salt thereof. The cancer may, for example, be pancreatic cancer, colorectal cancer, non-small cell lung cancer, acute myeloid leukemia, multiple myeloma, thyroid gland adenocarcinoma, a myelodysplastic syndrome, or squamous cell lung carcinoma. In some embodiments, the cancer comprises a Ras mutation, such as K-Ras G12C, K-Ras G13C, H-Ras G12C, H-Ras G13C, N-Ras G12C, or N-Ras G13C. Other Ras mutations are described herein.

25 Further provided is a method of treating a Ras protein-related disorder in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof.

Further provided is a method of inhibiting a Ras protein in a cell, the method comprising contacting the cell with an effective amount of a compound of the present invention, or a pharmaceutically 30 acceptable salt thereof. For example, the Ras protein is K-Ras G12C, K-Ras G13C, H-Ras G12C, H-Ras G13C, N-Ras G12C, or N-Ras G13C. Other Ras proteins are described herein. The cell may be a cancer cell, such as a pancreatic cancer cell, a colorectal cancer cell, a non-small cell lung cancer cell, an acute myeloid leukemia cell, a multiple myeloma cell, a thyroid gland adenocarcinoma cell, a myelodysplastic syndrome cell, or a squamous cell lung carcinoma cell. Other cancer types are 35 described herein. The cell may be in vivo or in vitro.

With respect to compounds of the present invention, one stereoisomer may exhibit better inhibition than another stereoisomer. For example, one atropisomer may exhibit inhibition, whereas the other atropisomer may exhibit little or no inhibition.

778

SUBSTITUTE SHEET (RULE 26) Methods of Synthesis

The compounds described herein may be made from commercially available starting materials or synthesized using known organic, inorganic, or enzymatic processes.

5 The compounds of the present invention can be prepared in a number of ways well known to those skilled in the art of organic synthesis. By way of example, compounds of the present invention can be synthesized using the methods described in the Schemes below, together with synthetic methods known in the art of synthetic organic chemistry, or variations thereon as appreciated by those skilled in the art. These methods include but are not limited to those methods described in the Schemes below.

10

Scheme 1. General synthesis of macrocyclic esters

.OPNG

A general synthesis of macrocyclic esters is outlined in Scheme 1. An appropriately substituted aryl-3-(5-bromo-1 -ethyl-1 H-indol-3-yl)-2,2-dimethylpropan-1-ol (1) can be prepared in three steps starting

779

SUBSTITUTE SHEET (RULE 26) from protected 3-(5-bromo-2-iodo-1H-indol-3-yl)-2,2-dimethylpropan-1-ol and appropriately substituted boronic acid, including palladium mediated coupling, alkylation, and de-protection reactions. Methyl-amino-hexahydropyridazine-3-carboxylate-boronic ester (2) can be prepared in three steps, including protection, iridium catalyst mediated borylation, and coupling with methyl methyl (S)- 5 hexahydropyridazine-3-carboxylate.

An appropriately substituted acetylpyrrolidine-3-carbonyl-N-methyl-L-valine (or an alternative aminoacid derivative (4) can be made by coupling of methyl-L-valinate and protected (S)-pyrrolidine-3- carboxylic acid, followed by deprotection, coupling with a carboxylic acid containing an appropriately substituted Michael acceptor, and a hydrolysis step.

10 The final macrocyclic esters can be made by coupling of methyl-amino-hexahydropyridazine-3- carboxylate-boronic ester (2) and aryl-3-(5-bromo-1 -ethyl-1 H-indol-3-yl)-2,2-dimethylpropan-1-ol (1) in the presence of a Pd catalyst followed by hydrolysis and macrolactonization steps to result in an appropriately protected macrocyclic intermediate (5). Deprotection and coupling with an appropriately substituted intermediate 4 results in a macrocyclic product. Additional deprotection and/or 15 functionalization steps can be required to produce the final compound.

Scheme 2. Alternative general synthesis of macrocyclic esters

Alternatively, macrocyclic ester can be prepared as described in Scheme 2. An appropriately 20 protected bromo-indolyl (6) coupled in the presence of a Pd catalyst with boronic ester (3), followed by iodination, deprotection, and ester hydrolysis. Subsequent coupling with methyl (S)-hexahydropyridazine- 3-carboxylate, followed by hydrolysis and macrolactonization can result in iodo intermediate (7). Coupling in the presence of a Pd catalyst with an appropriately substituted boronic ester and alkyllation can yield fully protected macrocycle (5). Additional deprotection or functionalization steps are required to produce 25 the final compound.

In addition, compounds of the disclosure can be synthesized using the methods described in the Examples below, together with synthetic methods known in the art of synthetic organic chemistry, or variations thereon as appreciated by those skilled in the art. These methods include but are not limited to those methods described in the Examples below. For example, a person of skill in the art would be able

780

SUBSTITUTE SHEET (RULE 26) to install into a macrocyclic ester a desired -B-L-W group of a compound of Formula (I), where B, L and W are defined herein, including by using methods exemplified in the Example section herein.

Compounds of Table 1 herein were prepared using methods disclosed herein or were prepared using methods disclosed herein combined with the knowledge of one of skill in the art. Compounds of 5 Table 2 may be prepared using methods disclosed herein or may be prepared using methods disclosed herein combined with the knowledge of one of skill in the art.

Scheme 3. General synthesis of macrocyclic esters

An alternative general synthesis of macrocyclic esters is outlined in Scheme 3. An appropriately substituted indolyl boronic ester (8) can be prepared in four steps starting from protected 3-(5-bromo-2- iodo-1 H-indol-3-yl)-2,2-dimethylpropan-1 -ol and appropriately substituted boronic acid, including Palladium mediated coupling, alkylation, de-protection, and Palladium mediated borylation reactions.

15 Methyl-amino-3-(4-bromothiazol-2-yl)propanoyl)hexahydropyrid azine-3-carboxylate (10) can be prepared via coupling of (S)-2-amino-3-(4-bromothiazol-2-yl)propanoic acid (9) with methyl (S)- hexahydropyridazine-3-carboxylate.

The final macrocyclic esters can be made by coupling of Methyl-amino-3-(4-bromothiazol-2- yl)propanoyl)hexahydropyridazine-3-carboxylate (10) and an appropriately substituted indolyl boronic 20 ester (8) in the presence of Pd catalyst followed by hydrolysis and macrolactonization steps to result in an appropriately protected macrocyclic intermediate (11). Deprotection and coupling with an appropriately

781

SUBSTITUTE SHEET (RULE 26) substituted intermediate 4 can result in a macrocyclic product. Additional deprotection or functionalization steps could be required to produce a final compound 13 or 14.

Scheme 4. General synthesis of macrocyclic esters

5

An alternative general synthesis of macrocyclic esters is outlined in Scheme 4. An appropriately substituted morpholine or an alternative herecyclic intermediate (15) can be coupled with appropriately protected Intermediate 1 via Palladium mediated coupling. Subsequent ester hydrolysis, and coupling 10 with piperazoic ester results in intermediate 16.

The macrocyclic esters can be made by hydrolysis, deprotection and macrocyclization sequence. Subsequent deprotection and coupling with Intermediate 4 (or analogs) result in an appropriately substituted final macrocyclic products. Additional deprotection or functionalization steps could be required to produce a final compound 17.

15

782

SUBSTITUTE SHEET (RULE 26) Scheme 5. General synthesis of macrocyclic esters

5 An alternative general synthesis of macrocyclic esters is outlined in Scheme 5. An appropriately substituted macrocycle (20) can be prepared starting from an appropriately protected boron ic ester 18 and bromo indolyl intermediate (19), including Palladium mediated coupling, hydrolysis, coupling with piperazoic ester, hydrolysis, de-protection, and macrocyclizarion steps. Subsequent coupling with an appropriately substituted protected aminoacid followed by palladium mediated coupling yiels intermediate 10 21. Additional deprotection and derivatization steps, including alkyllation may be required at this point.

The final macrocyclic esters can be made by coupling of intermediate (22) and an appropriately substituted carboxylic acid intermediate (23). Additional deprotection or functionalization steps could be required to produce a final compound (24).

In addition, compounds of the disclosure can be synthesized using the methods described in the 15 Examples below, together with synthetic methods known in the art of synthetic organic chemistry, or variations thereon as appreciated by those skilled in the art. These methods include but are not limited to those methods described in the Examples below. For example, a person of skill in the art would be able to install into a macrocyclic ester a desired -B-L-W group of a compound of Formula (I), where B, L and W are defined herein, including by using methods exemplified in the Example section herein.

783

SUBSTITUTE SHEET (RULE 26) Pharmaceutical Compositions and Methods of Use

Pharmaceutical Compositions and Methods of Administration

The compounds with which the invention is concerned are Ras inhibitors, and are useful in the 5 treatment of cancer. Accordingly, one embodiment of the present invention provides pharmaceutical compositions containing a compound of the invention or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, as well as methods of using the compounds of the invention to prepare such compositions.

As used herein, the term “pharmaceutical composition" refers to a compound, such as a 10 compound of the present invention, or a pharmaceutically acceptable salt thereof, formulated together with a pharmaceutically acceptable excipient.

In some embodiments, a compound is present in a pharmaceutical composition in unit dose amount appropriate for administration in a therapeutic regimen that shows a statistically significant probability of achieving a predetermined therapeutic effect when administered to a relevant population. In 15 some embodiments, pharmaceutical compositions may be specially formulated for administration in solid or liquid form, including those adapted for the following: oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), tablets, e.g., those targeted for buccal, sublingual, and systemic absorption, boluses, powders, granules, pastes for application to the tongue; parenteral administration, for example, by subcutaneous, intramuscular, intravenous or epidural injection as, for 20 example, a sterile solution or suspension, or sustained-release formulation; topical application, for example, as a cream, ointment, or a controlled-release patch or spray applied to the skin, lungs, or oral cavity; intravaginally or intrarectally, for example, as a pessary, cream, or foam; sublingually; ocularly; transdermally; or nasally, pulmonary, and to other mucosal surfaces.

A “pharmaceutically acceptable excipient," as used herein, refers any inactive ingredient (for 25 example, a vehicle capable of suspending or dissolving the active compound) having the properties of being nontoxic and non-inflammatory in a subject. Typical excipients include, for example: antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspensing or dispersing agents, sweeteners, or waters 30 of hydration. Excipients include, but are not limited to: butylated optionally substituted hydroxyltoluene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmellose, crosslinked polyvinyl pyrrolidone, citric acid, crospovidone, cysteine, ethylcellulose, gelatin, optionally substituted hydroxylpropyl cellulose, optionally substituted hydroxylpropyl methylcellulose, lactose, magnesium stearate, maltitol, mannitol, methionine, methylcellulose, methyl paraben, microcrystalline cellulose,

35 polyethylene glycol, polyvinyl pyrrolidone, povidone, pregelatinized starch, propyl paraben, retinyl palmitate, shellac, silicon dioxide, sodium carboxymethyl cellulose, sodium citrate, sodium starch glycolate, sorbitol, starch (corn), stearic acid, stearic acid, sucrose, talc, titanium dioxide, vitamin A, vitamin E, vitamin C, and xylitol. Those of ordinary skill in the art are familiar with a variety of agents and materials useful as excipients. See, e.g., e.g., Ansel, et al., Ansel's Pharmaceutical Dosage Forms and 40 Drug Delivery Systems. Philadelphia: Lippincott, Williams & Wilkins, 2004; Gennaro, et al., Remington: The Science and Practice of Pharmacy. Philadelphia: Lippincott, Williams & Wilkins, 2000; and Rowe,

784

SUBSTITUTE SHEET (RULE 26) Handbook of Pharmaceutical Excipients. Chicago, Pharmaceutical Press, 2005. In some embodiments, a composition includes at least two different pharmaceutically acceptable excipients.

Compounds described herein, whether expressly stated or not, may be provided or utilized in salt form, e.g., a pharmaceutically acceptable salt form, unless expressly stated to the contrary. The term 5 “pharmaceutically acceptable salt," as use herein, refers to those salts of the compounds described herein that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and other animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, pharmaceutically acceptable salts are described in: Berge et al., J. Pharmaceutical 10 Sciences 66:1 -19, 1977 and in Pharmaceutical Salts: Properties, Selection, and Use, (Eds. P.H. Stahl and C.G. Wermuth), Wiley-VCH, 2008. The salts can be prepared in situ during the final isolation and purification of the compounds described herein or separately by reacting the free base group with a suitable organic acid.

The compounds of the invention may have ionizable groups so as to be capable of preparation as 15 pharmaceutically acceptable salts. These salts may be acid addition salts involving inorganic or organic acids or the salts may, in the case of acidic forms of the compounds of the invention, be prepared from inorganic or organic bases. In some embodiments, the compounds are prepared or used as pharmaceutically acceptable salts prepared as addition products of pharmaceutically acceptable acids or bases. Suitable pharmaceutically acceptable acids and bases are well-known in the art, such as 20 hydrochloric, sulfuric, hydrobromic, acetic, lactic, citric, or tartaric acids for forming acid addition salts, and potassium hydroxide, sodium hydroxide, ammonium hydroxide, caffeine, various amines, and the like for forming basic salts. Methods for preparation of the appropriate salts are well-established in the art.

Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate,

25 cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-optionally substituted hydroxyl-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate,

30 stearate, succinate, sulfate, tartrate, thiocyanate, toluenesulfonate, undecanoate, valerate salts and the like. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like.

35 As used herein, the term “subject" refers to any member of the animal kingdom. In some embodiments, “subject" refers to humans, at any stage of development. In some embodiments, “subject" refers to a human patient. In some embodiments, “subject" refers to non-human animals. In some embodiments, the non-human animal is a mammal (e.g., a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a sheep, cattle, a primate, or a pig). In some embodiments, subjects include, but are not 40 limited to, mammals, birds, reptiles, amphibians, fish, or worms. In some embodiments, a subject may be a transgenic animal, genetically-engineered animal, or a clone.

785

SUBSTITUTE SHEET (RULE 26) As used herein, the term “dosage form" refers to a physically discrete unit of a compound (e.g., a compound of the present invention) for administration to a subject. Each unit contains a predetermined quantity of compound. In some embodiments, such quantity is a unit dosage amount (or a whole fraction thereof) appropriate for administration in accordance with a dosing regimen that has been determined to 5 correlate with a desired or beneficial outcome when administered to a relevant population (i.e., with a therapeutic dosing regimen). Those of ordinary skill in the art appreciate that the total amount of a therapeutic composition or compound administered to a particular subject is determined by one or more attending physicians and may involve administration of multiple dosage forms.

As used herein, the term “dosing regimen" refers to a set of unit doses (typically more than one)

10 that are administered individually to a subject, typically separated by periods of time. In some embodiments, a given therapeutic compound (e.g., a compound of the present invention) has a recommended dosing regimen, which may involve one or more doses. In some embodiments, a dosing regimen comprises a plurality of doses each of which are separated from one another by a time period of the same length; in some embodiments, a dosing regimen comprises a plurality of doses and at least two 15 different time periods separating individual doses. In some embodiments, all doses within a dosing regimen are of the same unit dose amount. In some embodiments, different doses within a dosing regimen are of different amounts. In some embodiments, a dosing regimen comprises a first dose in a first dose amount, followed by one or more additional doses in a second dose amount different from the first dose amount. In some embodiments, a dosing regimen comprises a first dose in a first dose amount, 20 followed by one or more additional doses in a second dose amount same as the first dose amount. In some embodiments, a dosing regimen is correlated with a desired or beneficial outcome when administered across a relevant population (i.e., is a therapeutic dosing regimen).

A “therapeutic regimen” refers to a dosing regimen whose administration across a relevant population is correlated with a desired or beneficial therapeutic outcome.

25 The term “treatment” (also “treat" or “treating"), in its broadest sense, refers to any administration of a substance (e.g., a compound of the present invention) that partially or completely alleviates, ameliorates, relieves, inhibits, delays onset of, reduces severity of, or reduces incidence of one or more symptoms, features, or causes of a particular disease, disorder, or condition. In some embodiments, such treatment may be administered to a subject who does not exhibit signs of the relevant disease,

30 disorder or condition or of a subject who exhibits only early signs of the disease, disorder, or condition. Alternatively, or additionally, in some embodiments, treatment may be administered to a subject who exhibits one or more established signs of the relevant disease, disorder, or condition. In some embodiments, treatment may be of a subject who has been diagnosed as suffering from the relevant disease, disorder, or condition. In some embodiments, treatment may be of a subject known to have one 35 or more susceptibility factors that are statistically correlated with increased risk of development of the relevant disease, disorder, or condition.

The term “therapeutically effective amount” means an amount that is sufficient, when administered to a population suffering from or susceptible to a disease, disorder, or condition in accordance with a therapeutic dosing regimen, to treat the disease, disorder, or condition. In some 40 embodiments, a therapeutically effective amount is one that reduces the incidence or severity of, or delays onset of, one or more symptoms of the disease, disorder, or condition. Those of ordinary skill in the art will appreciate that the term “therapeutically effective amount” does not in fact require successful

786

SUBSTITUTE SHEET (RULE 26) treatment be achieved in a particular individual. Rather, a therapeutically effective amount may be that amount that provides a particular desired pharmacological response in a significant number of subjects when administered to patients in need of such treatment. It is specifically understood that particular subjects may, in fact, be “refractory" to a “therapeutically effective amount." In some embodiments,

5 reference to a therapeutically effective amount may be a reference to an amount as measured in one or more specific tissues (e.g., a tissue affected by the disease, disorder or condition) or fluids (e.g., blood, saliva, serum, sweat, tears, urine). Those of ordinary skill in the art will appreciate that, in some embodiments, a therapeutically effective amount may be formulated or administered in a single dose. In some embodiments, a therapeutically effective amount may be formulated or administered in a plurality of 10 doses, for example, as part of a dosing regimen.

For use as treatment of subjects, the compounds of the invention, or a pharmaceutically acceptable salt thereof, can be formulated as pharmaceutical or veterinary compositions. Depending on the subject to be treated, the mode of administration, and the type of treatment desired, e.g., prevention, prophylaxis, or therapy, the compounds, or a pharmaceutically acceptable salt thereof, are formulated in 15 ways consonant with these parameters. A summary of such techniques may be found in Remington: The Science and Practice of Pharmacy, 21 st Edition, Lippincott Williams & Wilkins, (2005); and Encyclopedia of Pharmaceutical Technology, eds. J. Swarbrick and J. C. Boylan, 1988-1999, Marcel Dekker, New York, each of which is incorporated herein by reference.

Compositions can be prepared according to conventional mixing, granulating or coating methods, 20 respectively, and the present pharmaceutical compositions can contain from about 0.1% to about 99%, from about 5% to about 90%, or from about 1% to about 20% of a compound of the present invention, or pharmaceutically acceptable salt thereof, by weight or volume. In some embodiments, compounds, or a pharmaceutically acceptable salt thereof, described herein may be present in amounts totaling 1-95% by weight of the total weight of a composition, such as a pharmaceutical composition.

25 The composition may be provided in a dosage form that is suitable for intraarticular, oral, parenteral (e.g., intravenous, intramuscular), rectal, cutaneous, subcutaneous, topical, transdermal, sublingual, nasal, vaginal, intravesicular, intraurethral, intrathecal, epidural, aural, or ocular administration, or by injection, inhalation, or direct contact with the nasal, genitourinary, reproductive or oral mucosa. Thus, the pharmaceutical composition may be in the form of, e.g., tablets, capsules, pills,

30 powders, granulates, suspensions, emulsions, solutions, gels including hydrogels, pastes, ointments, creams, plasters, drenches, osmotic delivery devices, suppositories, enemas, injectables, implants, sprays, preparations suitable for iontophoretic delivery, or aerosols. The compositions may be formulated according to conventional pharmaceutical practice.

As used herein, the term “administration" refers to the administration of a composition (e.g., a 35 compound, or a preparation that includes a compound as described herein) to a subject or system.

Administration to an animal subject (e.g., to a human) may be by any appropriate route. For example, in some embodiments, administration may be bronchial (including by bronchial instillation), buccal, enteral, interdermal, intra-arterial, intradermal, intragastric, intramedullary, intramuscular, intranasal, intraperitoneal, intrathecal, intravenous, intraventricular, mucosal, nasal, oral, rectal, subcutaneous,

40 sublingual, topical, tracheal (including by intratracheal instillation), transdermal, vaginal, or vitreal.

Formulations may be prepared in a manner suitable for systemic administration or topical or local administration. Systemic formulations include those designed for injection (e.g., intramuscular,

787

SUBSTITUTE SHEET (RULE 26) intravenous or subcutaneous injection) or may be prepared for transdermal, transmucosal, or oral administration. A formulation will generally include a diluent as well as, in some cases, adjuvants, buffers, preservatives and the like. Compounds, or a pharmaceutically acceptable salt thereof, can be administered also in liposomal compositions or as microemulsions.

5 For injection, formulations can be prepared in conventional forms as liquid solutions or suspensions or as solid forms suitable for solution or suspension in liquid prior to injection or as emulsions. Suitable excipients include, for example, water, saline, dextrose, glycerol and the like. Such compositions may also contain amounts of nontoxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents and the like, such as, for example, sodium acetate, sorbitan monolaurate, 10 and so forth.

Various sustained release systems for drugs have also been devised. See, for example, U.S. Patent No. 5,624,677.

Systemic administration may also include relatively noninvasive methods such as the use of suppositories, transdermal patches, transmucosal delivery and intranasal administration. Oral 15 administration is also suitable for compounds of the invention, or a pharmaceutically acceptable salt thereof. Suitable forms include syrups, capsules, and tablets, as is understood in the art.

Each compound, or a pharmaceutically acceptable salt thereof, as described herein, may be formulated in a variety of ways that are known in the art. For example, the first and second agents of the combination therapy may be formulated together or separately. Other modalities of combination therapy 20 are described herein.

The individually or separately formulated agents can be packaged together as a kit. Non-limiting examples include, but are not limited to, kits that contain, e.g., two pills, a pill and a powder, a suppository and a liquid in a vial, two topical creams, etc. The kit can include optional components that aid in the administration of the unit dose to subjects, such as vials for reconstituting powder forms, syringes for 25 injection, customized IV delivery systems, inhalers, etc. Additionally, the unit dose kit can contain instructions for preparation and administration of the compositions. The kit may be manufactured as a single use unit dose for one subject, multiple uses for a particular subject (at a constant dose or in which the individual compounds, or a pharmaceutically acceptable salt thereof, may vary in potency as therapy progresses); or the kit may contain multiple doses suitable for administration to multiple subjects (“bulk 30 packaging”). The kit components may be assembled in cartons, blister packs, bottles, tubes, and the like.

Formulations for oral use include tablets containing the active ingredient(s) in a mixture with non-toxic pharmaceutically acceptable excipients. These excipients may be, for example, inert diluents or fillers (e.g., sucrose, sorbitol, sugar, mannitol, microcrystalline cellulose, starches including potato starch, calcium carbonate, sodium chloride, lactose, calcium phosphate, calcium sulfate, or sodium phosphate); 35 granulating and disintegrating agents (e.g., cellulose derivatives including microcrystalline cellulose, starches including potato starch, croscarmellose sodium, alginates, or alginic acid); binding agents (e.g., sucrose, glucose, sorbitol, acacia, alginic acid, sodium alginate, gelatin, starch, pregelatinized starch, microcrystalline cellulose, magnesium aluminum silicate, carboxymethylcellulose sodium, methylcellulose, optionally substituted hydroxylpropyl methylcellulose, ethylcellulose, polyvinylpyrrolidone, 40 or polyethylene glycol); and lubricating agents, glidants, and antiadhesives (e.g., magnesium stearate, zinc stearate, stearic acid, silicas, hydrogenated vegetable oils, or talc). Other pharmaceutically

788

SUBSTITUTE SHEET (RULE 26) acceptable excipients can be colorants, flavoring agents, plasticizers, humectants, buffering agents, and the like.

Two or more compounds may be mixed together in a tablet, capsule, or other vehicle, or may be partitioned. In one example, the first compound is contained on the inside of the tablet, and the second 5 compound is on the outside, such that a substantial portion of the second compound is released prior to the release of the first compound.

Formulations for oral use may also be provided as chewable tablets, or as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent (e.g., potato starch, lactose, microcrystalline cellulose, calcium carbonate, calcium phosphate or kaolin), or as soft gelatin capsules 10 wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin, or olive oil. Powders, granulates, and pellets may be prepared using the ingredients mentioned above under tablets and capsules in a conventional manner using, e.g., a mixer, a fluid bed apparatus or a spray drying equipment.

Dissolution or diffusion-controlled release can be achieved by appropriate coating of a tablet,

15 capsule, pellet, or granulate formulation of compounds, or by incorporating the compound, or a pharmaceutically acceptable salt thereof, into an appropriate matrix. A controlled release coating may include one or more of the coating substances mentioned above or, e.g., shellac, beeswax, glycowax, castor wax, carnaubawax, stearyl alcohol, glyceryl monostearate, glyceryl distearate, glycerol palmitostearate, ethylcellulose, acrylic resins, dl-polylactic acid, cellulose acetate butyrate, polyvinyl 20 chloride, polyvinyl acetate, vinyl pyrrolidone, polyethylene, polymethacrylate, methylmethacrylate, 2-optionally substituted hydroxylmethacrylate, methacrylate hydrogels, 1 ,3 butylene glycol, ethylene glycol methacrylate, or polyethylene glycols. In a controlled release matrix formulation, the matrix material may also include, e.g., hydrated methylcellulose, carnauba wax and stearyl alcohol, carbopol 934, silicone, glyceryl tristearate, methyl acrylate-methyl methacrylate, polyvinyl chloride, polyethylene, or 25 halogenated fluorocarbon.

The liquid forms in which the compounds, or a pharmaceutically acceptable salt thereof, and compositions of the present invention can be incorporated for administration orally include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical 30 vehicles.

Generally, when administered to a human, the oral dosage of any of the compounds of the invention, or a pharmaceutically acceptable salt thereof, will depend on the nature of the compound, and can readily be determined by one skilled in the art. A dosage may be, for example, about 0.001 mg to about 2000 mg per day, about 1 mg to about 1000 mg per day, about 5 mg to about 500 mg per day,

35 about 100 mg to about 1500 mg per day, about 500 mg to about 1500 mg per day, about 500 mg to about 2000 mg per day, or any range derivable therein.

In some embodiments, the pharmaceutical composition may further comprise an additional compound having antiproliferative activity. Depending on the mode of administration, compounds, or a pharmaceutically acceptable salt thereof, will be formulated into suitable compositions to permit facile 40 delivery. Each compound, or a pharmaceutically acceptable salt thereof, of a combination therapy may be formulated in a variety of ways that are known in the art. For example, the first and second agents of

789

SUBSTITUTE SHEET (RULE 26) the combination therapy may be formulated together or separately. Desirably, the first and second agents are formulated together for the simultaneous or near simultaneous administration of the agents.

It will be appreciated that the compounds and pharmaceutical compositions of the present invention can be formulated and employed in combination therapies, that is, the compounds and 5 pharmaceutical compositions can be formulated with or administered concurrently with, prior to, or subsequent to, one or more other desired therapeutics or medical procedures. The particular combination of therapies (therapeutics or procedures) to employ in a combination regimen will take into account compatibility of the desired therapeutics or procedures and the desired therapeutic effect to be achieved. It will also be appreciated that the therapies employed may achieve a desired effect for the 10 same disorder, or they may achieve different effects (e.g., control of any adverse effects).

Administration of each drug in a combination therapy, as described herein, can, independently, be one to four times daily for one day to one year, and may even be for the life of the subject. Chronic, long-term administration may be indicated.

15

Numbered Embodiments

[1] A compound, or pharmaceutically acceptable salt thereof, having the structure of Formula I:

Formula I

20 wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH3)C(0)-(CH2)- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 10- membered heteroarylene;

25 B is absent, -CH(R 9 )-, >C=CR 9 R 9' , or >CR 9 R 9 ’ where the carbon is bound to the carbonyl carbon of -N(R 11 )C(0)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C1-C4 alkylene, optionally substituted C1-C4 alkenylene, optionally

30 substituted C1-C4 heteroalkylene, -C(0)0-CH(R 6 )- where C is bound to -C(R 7 R®)-, -C(0)NH-CH(R 6 )-

790

SUBSTITUTE SHEET (RULE 26) where C is bound to -C(R 7 R 8 )-, optionally substituted C1-C4 heteroalkylene, or 3 to 8-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a vinyl ketone, a vinyl sulfone, an ynone, or an alkynyl

5 sulfone;

X 1 is optionally substituted C1-C2 alkylene, NR, O, or S(0) n ;

X 2 is O or NH;

X 8 is N or CH; n is 0, 1 , or 2;

10 R is hydrogen, cyano, optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, optionally substituted C2-C4 alkynyl, C(0)R’, C(0)0R’, C(0)N(R')2, S(0)R’, S(0) 2 R\ or S(0) 2 N(R’)2; each R’ is, independently, H or optionally substituted C1-C4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

15 Y 5 is CH, CH2, or N;

Y 8 is C(O), CH, CH2, or N;

R 1 is cyano, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally 20 substituted 5 to 10-membered heteroaryl, or

R 1 and R 2 combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 2 is absent, hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally 25 substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

30 R 5 is hydrogen, C1-C4 alkyl optionally substituted with halogen, cyano, hydroxy, or C1-C4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl, or R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

35 R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), 40 C=N(0-CI-C3 alkyl), C=0, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

791

SUBSTITUTE SHEET (RULE 26) R 7a and R are, independently, hydrogen, halo, optionally substituted C1-C3 alkyl, or combine with the carbon to which they are attached to form a carbonyl;

R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted Ca-Ce 5 alkenyl, optionally substituted Cz-Ce alkyny!, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 ’ combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

10 R 9 is H, F, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl, or

R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 9' is hydrogen or optionally substituted Ci-Ce alkyl; or

15 R 9 and R 9' , combined with the atoms to which they are attached, form a 3 to 6-membered cycloalkyl or a 3 to 6-membered heterocycloalkyl;

R 10 is hydrogen, halo, hydroxy, C1-C3 alkoxy, or C1-C3 alkyl;

R i °a j s hydrogen or halo;

R 11 is hydrogen or C1-C3 alkyl; and

20 R 21 is H or C1-C3 alkyl.

[2] The compound, or pharmaceutically acceptable salt thereof, of paragraph [1], wherein G is optionally substituted C1-C4 heteroalkylene.

[3] The compound, or pharmaceutically acceptable salt thereof, of paragraph [1] or [2], wherein the compound has the structure of Formula lc:

2

Formula lc wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH3)C(0)-(CH2)- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered 30 heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene;

792

SUBSTITUTE SHEET (RULE 26) B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -N(R 11 )C(0)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

5 W is a cross-linking group comprising a vinyl ketone, a vinyl sulfone, an ynone, or an alkynyl sulfone;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2;

10 R is hydrogen, cyano, optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, optionally substituted C2-C4 alkynyl, C(0)R’, C(0)OR’, C(0)N(R')z, S(0)R’, S(0)zR\ or S(0)zN(R’)z; each R’ is, independently, H or optionally substituted C1-C4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

15 Y 5 and Y 6 are, independently, CH or N;

R 1 is cyano, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl;

20 R 2 is hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted Cz-Ce alkenyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 25 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C1-C4 alkyl optionally substituted with halogen, cyano, hydroxy, or C1-C4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl, or

30 R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted C2-C6 alkenyl, optionally substituted Ca-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl,

35 optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R e combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(0-CI-C3 alkyl), C=0, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7' is hydrogen, halogen, or optionally substituted C1-C3 alkyl; R 8' is hydrogen, halogen, hydroxy, 40 cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted Cz-Ce alkenyl, optionally substituted Cz-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally

793

SUBSTITUTE SHEET (RULE 26) substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 ’ combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

5 R 9 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 10 is hydrogen, hydroxy, C1-C3 alkoxy, or C1-C3 alkyl; and R 1 ' is hydrogen or C1-C3 alkyl.

[4] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

10 [3], wherein X 2 is NH.

[5] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[4], wherein X 3 is CH.

[6] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[5], wherein R 11 is hydrogen.

15 [7] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [5], wherein R 11 is C1-C3 alkyl.

[8] The compound, or pharmaceutically acceptable salt thereof, of paragraph [7], wherein R 11 is methyl.

[9] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to 20 [6], wherein the compound has the structure of Formula Id:

Formula Id wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH3)C(0)-(CH2)- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, 25 optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, 30 optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

794

SUBSTITUTE SHEET (RULE 26) W is a cross-linking group comprising a vinyl ketone, a vinyl sulfone, an ynone, or an alkynyl sulfone; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl,

5 optionally substituted C2-C4 alkynyl, C(0)R’, C(0)0R’, C(0)N(R’) 2 , S(0)R’, S(0) 2 R', or S(0) 2 N(R’) 2 ; each R ' is, independently, H or optionally substituted C1-C4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 and Y 6 are, independently, CH or N;

10 R 1 is cyano, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl;

R 2 is hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted C2-Ce alkenyl, optionally 15 substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

20 R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C1-C4 alkyl optionally substituted with halogen, cyano, hydroxy, or C1-C4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl, or R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally 25 substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted C 2 -Ce alkenyl, optionally substituted Cz-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

30 R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR r R 8' ; C=N(OH), C=N(0-Ci-Cs alkyl), C=0, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R T is hydrogen, halogen, or optionally substituted C1-C3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted Ca-Ce 35 alkenyl, optionally substituted Ca-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8 ’ combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

40 R 9 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; and R 10 is hydrogen, hydroxy, C1-C3 alkoxy, or C1-C3 alkyl.

795

SUBSTITUTE SHEET (RULE 26) [10] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [9] wherein X' is optionally substituted C1-C2 alkylene.

[11] The compound, or pharmaceutically acceptable salt thereof, of paragraph [10], wherein X 1 is methylene.

5 [12] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [11], wherein R 5 is hydrogen.

[13] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [11], wherein R 5 is C1-C4 alkyl optionally substituted with halogen.

10 [14] The compound, or pharmaceutically acceptable salt thereof, of paragraph [13], wherein R 5 is methyl.

[15] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [14], wherein Y 4 is C.

[16] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

15 [15], wherein R 4 is hydrogen.

[17] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[16], wherein Y 5 is CH.

[18] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[17], wherein Y® is CH.

20 [19] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[18], wherein Y 1 is C.

[20] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[19] wherein Y 2 is C.

[21] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

25 [20] wherein Y 3 is N.

[22] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[21], wherein R 3 is absent.

[23] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[22], wherein Y 7 is C.

30 [24] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [6] or [9] to [23], wherein the compound has the structure of Formula le:

796

SUBSTITUTE SHEET (RULE 26) Formula le wherein A is -N(H or CH3)C(0)-(CH2)- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 5 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

10 W is a cross-linking group comprising a vinyl ketone, a vinyl sulfone, an ynone, or an alkynyl sulfone;

R 1 is cyano, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally 15 substituted 5 to 10-membered heteroaryl;

R 2 is hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted Ca-Ce alkenyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

20 R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 5 is hydrogen, C1-C4 alkyl optionally substituted with halogen, cyano, hydroxy, or C1-C4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl, or

25 R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted Ca-Ce alkenyl, optionally substituted Ca-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl,

30 optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R® combine with the carbon atom to which they are attached to form C=CR r R 8' ; C=N(OH), C=N(0-CI-C3 alkyl), C=0, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R T is hydrogen, halogen, or optionally substituted C1-C3 alkyl; R 8' is hydrogen, halogen, hydroxy, 35 cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted Ca-Ce alkenyl, optionally substituted Ca-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8 ’ combine with the carbon atom to which they are attached to form optionally 40 substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; and

797

SUBSTITUTE SHEET (RULE 26) R 10 is hydrogen, hydroxy, C1-C3 alkoxy, or C1-C3 alkyl.

[25] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [3] to [24], wherein R 6 is hydrogen.

[26] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

5 [25], wherein R 2 is hydrogen, cyano, optionally substituted Ci-Ce alkyl, optionally substituted 3 to 6- membered cycloalkyl, or optionally substituted 3 to 6-membered heterocycloalkyl.

[27] The compound, or pharmaceutically acceptable salt thereof, of paragraph [26], wherein R 2 is optionally substituted Ci-Ce alkyl.

[28] The compound, or pharmaceutically acceptable salt thereof, of paragraph [27], wherein R 2 is

10 ethyl.

[29] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [28], wherein R 7 is optionally substituted C1-C3 alkyl.

[30] The compound, or pharmaceutically acceptable salt thereof, of paragraph 29, wherein R 7 is

C1-C3 alkyl.

15 [31] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to 30, wherein R 8 is optionally substituted C1-C3 alkyl.

[32] The compound, or pharmaceutically acceptable salt thereof, of paragraph [31], wherein R 8 is

C1-C3 alkyl.

[33] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to 20 [32], wherein the compound has the structure of Formula If:

Formula If wherein A is -N(H or CH3)C(0)-(CH2)- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- 25 membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

30 L is absent or a linker;

W is a cross-linking group comprising a vinyl ketone, a vinyl sulfone, an ynone, or an alkynyl sulfone;

798

SUBSTITUTE SHEET (RULE 26) R 1 is cyano, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl;

5 R 2 is Ci-Ce alkyl or 3 to 6-membered cycloalkyl;

R 7 is C1-C3 alkyl;

R 8 is C1-C3 alkyl; and

R 9 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl.

10 [34] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [33], wherein R 1 is optionally substituted 6 to 10-membered aryl, optionally substituted 3 to 6-membered cycloalkenyl, or optionally substituted 5 to 10-membered heteroaryl.

[35] The compound, or pharmaceutically acceptable salt thereof, of paragraph [34], wherein R 1 is optionally substituted 6-membered aryl, optionally substituted 6-membered cycloalkenyl, or optionally

15 substituted 6-membered heteroaryl.

[36] The compound, or pharmaceutically acceptable salt thereof, of paragraph [35], wherein R 1 is

[37] The compound, or pharmaceutically acceptable salt thereof, of paragraph [36], wherein R 1 is

799

SUBSTITUTE SHEET (RULE 26) [38] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [37], wherein the compound has the structure of Formula Ig:

Formula Ig

5 wherein A is -N(H or CH3)C(0)-(CH2)- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally 10 substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a vinyl ketone, a vinyl sulfone, an ynone, or an alkynyl sulfone;

15 R 2 is Ci-Ce alkyl, Ci-Ce fluoroalkyl, or 3 to 6-memberod cycloalkyl;

R 7 is C1-C3 alkyl;

R 8 is C1-C3 alkyl; and

R 9 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl

20 X ® and X* are, independently, N or CH; and

R 12 is optionally substituted Ci-Ce alkyl, optionally substituted CvCe heteroalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl.

[39] The compound, or pharmaceutically acceptable salt thereof, of paragraph [38], wherein X ® is

N and X» is CH.

25 [40] The compound, or pharmaceutically acceptable salt thereof, of paragraph [38], wherein X ® is

CH and X« is N.

[41] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [38] to [40], wherein R 12 is optionally substituted Ci-Ce heteroalkyl.

800

SUBSTITUTE SHEET (RULE 26) [42] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [38]

[43] The compound, or pharmaceutically acceptable salt thereof, of paragraph [1] or [2], wherein 5 the compound has the structure of Formula VI:

Formula VI wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

10 A is -N(H or CH3)C(0)-(CH2)- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 10- membered heteroarylene;

B is absent, -CH(R 9 )-, >C=CR 9 R 9' , or >CR 9 R 9' where the carbon is bound to the carbonyl carbon 15 of -N(R 11 )C(0)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted Ci-C* alkylene, optionally substituted C1-C4 alkenylene, optionally substituted C1-C4 heteroalkylene, -C(0)0-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(0)NH-CH(R 6 )-

20 where C is bound to -C(R 7 R 8 )-, optionally substituted C1-C4 heteroalkylene, or 3 to 8-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a vinyl ketone, a vinyl sulfone, an ynone, a haloacetal, or an alkynyl sulfone;

25 X 1 is optionally substituted C1-C2 alkylene, NR, O, or S(0) n ;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2;

801

SUBSTITUTE SHEET (RULE 26) R is hydrogen, cyano, optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, optionally substituted C2-C4 alkynyl, C(0)R’, C(0)0R', C(0)N(R’) 2 , S(0)R', S(0) 2 R', or S(0) 2 N(R') 2 ; each R ' is, independently, H or optionally substituted C1-C4 alkyl;

Y 1 is C, CH, or N;

5 Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 is CH, CH 2 , or N;

Y 6 is C(O), CH, CH 2 , or N;

R 2 is absent, hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted C 2 -Ce alkenyl, optionally substituted C 2 -Ce alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally 10 substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

15 R 5 is hydrogen, C1-C4 alkyl optionally substituted with halogen, cyano, hydroxy, or C1-C4 alkoxy, cyclopropyl, or cyclobutyl;

R® is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C1-C 3 alkyl, or R® and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

20 R® is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted C 2 -Ce alkenyl, optionally substituted C^Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R® combine with the carbon atom to which they are attached to form C=CR r R ® ’; C=N(OH), 25 C=N(0-Ci-Cs alkyl), C=0, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7a and R are, independently, hydrogen, halo, optionally substituted C1-C3 alkyl, or combine with the carbon to which they are attached to form a carbonyl;

R T is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R ®' is hydrogen, halogen, hydroxy, 30 cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted Cz-Ce alkenyl, optionally substituted Ci!-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R T and R ® ’ combine with the carbon atom to which they are attached to form optionally 35 substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is H, F, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; or

R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

40 R 9' is hydrogen or optionally substituted Ci-Ce alkyl; or

R 9 and R 9 , combined with the atoms to which they are attached, form a 3 to 6-membered cycloalkyl or a 3 to 6-membered heterocycloalkyl;

802

SUBSTITUTE SHEET (RULE 26) R 10 is hydrogen, halo, hydroxy, C1-C3 alkoxy, or C1-C3 alkyl;

R 10a is hydrogen or halo;

R 1 ' is hydrogen or Ci-Ce alkyl;

R 2 ' is hydrogen or C1-C3 alkyl (e.g., methyl); and

5 X ® and X* are, independently, N or CH.

[44] The compound, or pharmaceutically acceptable salt thereof, of paragraph [43], wherein the compound has the structure of Formula Via:

Formula Via

10 wherein A is optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, 15 optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is a cross-linking group comprising a vinyl ketone, a vinyl sulfone, an ynone, or an alkynyl sulfone;

X 1 is optionally substituted C1-C2 alkylene, NR, O, or S(0) n ;

20 X 2 is O or NH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, optionally substituted C2-C4 alkynyl, C(0)R', C(0)0R', C(0)N(R') 2 , S(0)R', S(0) 2 R\ or S(0) 2 N(R') 2 ; each R’ is, independently, H or optionally substituted C1-C4 alkyl;

25 R 2 is Ci-Ce alkyl, Ci-Ce fluoroalkyl, or 3 to 6-membered cycloalkyl;

R 7 is C1-C3 alkyl;

R 8 is C1-C3 alkyl; and

R 9 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

30 X ® and X* are, independently, N or CH;

R 1 ' is hydrogen or Ci-Ce alkyl; and R 2 ' is hydrogen or Ci-Ce alkyl.

803

SUBSTITUTE SHEET (RULE 26) [45] The compound, or pharmaceutically acceptable salt thereof, of paragraph [43] or [44], wherein the compound has the structure of Formula Vlb:

Formula Vlb

5 wherein A is optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, 10 optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

R 9 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

L is absent or a linker; and

W is a cross-linking group comprising a vinyl ketone, a vinyl sulfone, an ynone, or an alkynyl

15 sulfone.

[46] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [45], wherein A is optionally substituted 6-membered arylene.

[47] The compound, or pharmaceutically acceptable salt thereof, of paragraph [46], wherein A has the structure: wherein R 13 is hydrogen, halo, hydroxy, amino, optionally substituted Ci-Ce alkyl, or optionally substituted Ci-Ce heteroalkyl; and R 13a is hydrogen or halo.

[48] The compound, or pharmaceutically acceptable salt thereof, of paragraph [47], wherein R 13 25 and R ,3a are each hydrogen.

[49] The compound, or pharmaceutically acceptable salt thereof, of paragraph [47], wherein R 13 is hydroxy, methyl, fluoro, or difluoromethyl.

804

SUBSTITUTE SHEET (RULE 26) [50] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [45], wherein A is optionally substituted 5 to 6-membered heteroarylene.

[51] The compound, or pharmaceutically acceptable salt thereof, of paragraph [50], wherein A is:

5

10 [52] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [45], wherein A is optionally substituted Ci-C* heteroalkylene.

[53] The compound, or pharmaceutically acceptable salt thereof, of paragraph [52], wherein A is:

[54] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

15 [45], wherein A is optionally substituted 3 to 6-membered heterocycloalkylene.

[55] The compound, or pharmaceutically acceptable salt thereof, of paragraph [54], wherein A is:

805

SUBSTITUTE SHEET (RULE 26) [56] The compound, or pharmaceutically acceptable salt thereof, of paragraph [55], wherein A is

A N \

Ut

[57] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [56], wherein B is -CHR 9 -.

5 [58] The compound, or pharmaceutically acceptable salt thereof, of paragraph [57], wherein R 9 is F, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6- membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl.

[59] The compound, or pharmaceutically acceptable salt thereof, of paragraph [58], wherein R 9

10 vO

[60] The compound, or pharmaceutically acceptable salt thereof, of paragraph [59], wherein R 9 is:

[61] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

15 [56], wherein B is optionally substituted 6-membered arylene.

[62] The compound, or pharmaceutically acceptable salt thereof, of paragraph [61], wherein B is 6-membered arylene. The compound, or pharmaceutically acceptable salt thereof, of paragraph [61], wherein B is:

20

[64] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[63], wherein R 7 is methyl.

[65] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[64], wherein R® is methyl.

25 [66] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[65], wherein the linker is the structure of Formula II:

A 1 -(B 1 MC 1 )g-(B 2 )h-(D 1 MB 3 )i-(C 2 )i-(B«)k-A 2 Formula II where A 1 is a bond between the linker and B; A 2 is a bond between W and the linker; B', B 2 , B 3 ,

30 and B 4 each, independently, is selected from optionally substituted C1-C2 alkylene, optionally substituted C1-C3 heteroalky lene, O, S, and NR N ; R N is hydrogen, optionally substituted C1-C4 alkyl, optionally

806

SUBSTITUTE SHEET (RULE 26) substituted C2-C4 alkenyl, optionally substituted C2-C4 alkynyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted C1-C7 heteroalkyl; C 1 and C 2 are each, independently, selected from carbonyl, thiocarbonyl, sulphonyl, or phosphoryl; f, g, h, I, j, and k are each, independently, 0 or 1 ; and D 1 is optionally substituted C1-C10 alkylene, optionally 5 substituted C2-C10 alkenylene, optionally substituted C2-C10 alkynylene, optionally substituted 3 to 14- membered heterocycloalkylene, optionally substituted 5 to 10-membered heteroarylene, optionally substituted 3 to 8-membered cycloalkylene, optionally substituted 6 to 10-membered arylene, optionally substituted C2-C10 polyethylene glycolene, or optionally substituted C1-C10 heteroalkylene, or a chemical bond linking A 1 -(BXC')g-(B 2 )h- to -(B 3 )i-(C 2 )j-(B 4 )ic-A 2 .

10 [67] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [66], wherein the linker is acyclic.

[68] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [67], wherein the linker has the structure of Formula lla:

R 14

15 Formula lla wherein X a is absent or N;

R 14 is absent, hydrogen or optionally substituted Ci-Ce alkyl; and

L 2 is absent, -SO2-, optionally substituted C1-C4 alkylene or optionally substituted C1-C4 heteroalkylene,

20 wherein at least one of X a , R 14 , or L 2 is present.

[69] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [68], wherein the linker has the structure:

25

[70] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [66], wherein the linker is or comprises a cyclic moiety.

[71] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [70], wherein the linker has the structure of Formula Mb:

R15 l vT Xt L 3

Cy 7

O ,

30 o

Formula Mb wherein o is 0 or 1 ;

807

SUBSTITUTE SHEET (RULE 26) R 15 is hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted 3 to 8-membered cycloalkylene, or optionally substituted 3 to 8-membered heterocycloalkylene;

X 4 is absent, optionally substituted C1-C4 alkylene, O, NCHs, or optionally substituted C1-C4 heteroalkylene;

5 Cy is optionally substituted 3 to 8-membered cycloalkylene, optionally substituted 3 to 8- membered heterocycloalkylene, optionally substituted 6-10 membered arylene, or optionally substituted 5 to 10-membered heteroarylene; and

L 3 is absent, -SO2-, optionally substituted C1-C4 alkylene or optionally substituted C1-C4 heteroalkylene.

10 [72] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [71], wherein the linker has the structure:

Formula lie wherein R 15 is hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted 3 to 8- 15 membered cycloalkylene, or optionally substituted 3 to 8-membered heterocycloalkylene; and

R'sa p i » Rise Risd Rise R i s anc j Risg are independently, hydrogen, halo, hydroxy, cyano, amino, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce alkoxy, or , or R 15b and R 15d combine with the carbons to which they are attached to form an optionally substituted 3 to 8-membered cycloalkylene, or optionally substituted 3 to 8-membered heterocycloalkylene.

20 [73] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [72], wherein the linker has the structure:

CH 3

H 2 N H Q Fx f F 3 C. H 3 Q

CH 3 CH 3

N -t N~/

V Y N

25 o o O O o

808

SUBSTITUTE SHEET (RULE 26) 5 [74] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [71], wherein the linker has the structure:

809

SUBSTITUTE SHEET (RULE 26) 810

SUBSTITUTE SHEET (RULE 26)

811

SUBSTITUTE SHEET (RULE 26)

[75] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1]

5 to [74], wherein W is a cross-linking group comprising a vinyl ketone.

[76]. The compound, or a pharmaceutically acceptable salt thereof, of paragraph [75], wherein W has the structure of Formula Ilia: o R 16b

\Y R 16C

R 16a

Formula Ilia

10 wherein R 16a , R 16b , and R 16c are, independently, hydrogen, -CN, halogen, or -C1-C3 alkyl optionally substituted with one or more substituents independently selected from -OH, -O-C1-C3 alkyl, -NHa, -NH(CI-C3 alkyl), -N(CI-C3 alkyl>2, or a 4 to 7-membered saturated heterocycloalkyl.

[77] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [76], wherein W is:

O

O O o OCH 3 O CH 3

N V

15 Y CH 3

I t t o O O

[78] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1]

20 to [74], wherein W is a cross-linking group comprising an ynone.

812

SUBSTITUTE SHEET (RULE 26) [79] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [78], wherein W has the structure of Formula lllb:

O

R 17

Formula lllb

5 wherein R 17 is hydrogen, -C1-C3 alkyl optionally substituted with one or more substituents independently selected from -OH, -O-C1-C3 alkyl, -NHa, -NH(CI-C3 alkyl), -N(Ci-Cs alkyl)a, or a 4 to 7- membered saturated cycloalkyl, or a 4 to 7-membered saturated heterocycloalkyl.

[80] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [79], wherein W is:

813

SUBSTITUTE SHEET (RULE 26) o

O

5

[81] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [74], wherein W is a cross-linking group comprising a vinyl sulfone.

[82] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [81], wherein W has the structure of Formula lllc:

R 18a

/> . R’®

S

O.' 'roV P(18c

10 Formula lllc wherein R 18a , R 18b , and R ,8c are, independently, hydrogen, -CN, or -C1-C3 alkyl optionally substituted with one or more substituents independently selected from -OH, -O-C1-C3 alkyl,

-NHa, -NH(CI-C3 alkyl), -N(CI-C3 alkyl>2, or a 4 to 7-membered saturated heterocycloalkyl.

15 [83] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [82], wherein W is:

[84] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [74], wherein W is a cross-linking group comprising an alkynyl sulfone.

20 [85] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [84], wherein W has the structure of Formula Mid:

<W>

R19

Formula Mid

814

SUBSTITUTE SHEET (RULE 26) wherein R 19 is hydrogen, -C1-C3 alkyl optionally substituted with one or more substituents independently selected from -OH, -O-C1-C3 alkyl, -IMH2, -NH(Ci-C3 alkyl), -N(CI-C3 alkyl)2, or a 4 to 7- membered saturated heterocycloalkyl, or a 4 to 7-membered saturated heterocycloalkyl.

[86] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [85], wherein W

5 is:

[87] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [74], wherein W has the structure of Formula llle:

O vV R20*·

10 Formula llle wherein X s is a halogen; and

R 20 is hydrogen, -C1-C3 alkyl optionally substituted with one or more substituents independently selected from -OH, -O-C1-C3 alkyl, -NH2, -NH(CI-C3 alkyl), -N(CI-C3 alkyl>2, or a 4 to 7-membered saturated heterocycloalkyl.

15 [88] A compound, or a pharmaceutically acceptable salt thereof, selected from Table 1 or Table

2.

[89] A pharmaceutical composition comprising a compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [88], and a pharmaceutically acceptable excipient.

[90] A conjugate, or salt thereof, comprising the structure of Formula IV:

20 M-L-P Formula IV wherein L is a linker;

P is a monovalent organic moiety; and M has the structure of Formula V:

Formula V wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH3)C(0)-(CH2)- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-,

30 optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered

815

SUBSTITUTE SHEET (RULE 26) heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene;

B is absent, -CH(R 9 )-, >C=CR 9 R 9' , or >CR 9 R 9 where the carbon is bound to the carbonyl carbon of -N(R 11 )C(0)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- 5 membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C1-C4 alkylene, optionally substituted C1-C4 alkenylene, optionally substituted C1-C4 heteroalkylene, -C(0)0-CH(R®)- where C is bound to -C(R 7 R®)-, -C(0)NH-CH(R®)- where C is bound to -C(R 7 R®)-, optionally substituted C1-C4 heteroalkylene, or 3 to 8-membered

10 heteroarylene;

X 1 is optionally substituted C1-C2 alkylene, NR, O, or S(0) n ;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2;

15 R is hydrogen, cyano, optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, optionally substituted C2-C4 alkynyl, C(0)R’, C(0)0R’, C(0)N(R') 2 , S(0)R’, S(0) 2 R', or S(0) 2 N(R’) 2 ; each R’ is, independently, H or optionally substituted C1-C4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

20 Y 5 is CH, CH 2 , or N;

Y® is C(O), CH, CH 2 , or N;

R 1 is cyano, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally 25 substituted 5 to 10-membered heteroaryl, or

R 1 and R 2 combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 2 is absent, hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted C 2 -Ce alkenyl, optionally substituted C 2 -Ce alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally 30 substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

35 R 5 is hydrogen, C1-C4 alkyl optionally substituted with halogen, cyano, hydroxy, or C1-C4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl, or R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

40 R® is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted C 2 -Ce alkenyl, optionally substituted Ca-Ce alkynyl, optionally

816

SUBSTITUTE SHEET (RULE 26) substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7 R 8 ; C=N(OH), C=N(0-Ci-Ca alkyl), C=0, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally 5 substituted 3 to 7-membered heterocycloalkyl;

R 7a and R are, independently, hydrogen, halo, optionally substituted C1-C3 alkyl, or combine with the carbon to which they are attached to form a carbonyl;

R 7 is hydrogen, halogen, or optionally substituted C1-C3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C1-C3 alkoxy, optionally substituted C1-C3 alkyl, optionally substituted C2-C6 10 alkenyl, optionally substituted Cz-Ce alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 ’ combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

15 R 9 is H, F, optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl, or

R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 9' is hydrogen or optionally substituted Ci-Ce alkyl; or

20 R 9 and R 9' , combined with the atoms to which they are attached, form a 3 to 6-membered cycloalkyl or a 3 to 6-membered heterocycloalkyl;

R 10 is hydrogen, halo, hydroxy, C1-C3 alkoxy, or C1-C3 alkyl;

R 10a is hydrogen or halo;

R 11 is hydrogen or C1-C3 alkyl; and

25 R 21 is H or C1-C3 alkyl.

[91 [ The conjugate of paragraph [90], or salt thereof, wherein M has the structure of Formula Vd:

Formula Vd wherein A is optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- 30 membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

817

SUBSTITUTE SHEET (RULE 26) B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

X 1 is optionally substituted C1-C2 alkylene, NR, O, or S(0) n ;

5 X 2 is O or NH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, optionally substituted C2-C4 alkynyl, C(0)R’, C(0)0R’, C(0)N(R')2, S(0)R’, S(0) 2 R\ or S(0) 2 N(R’)2; each R’ is, independently, H or optionally substituted C1-C4 alkyl;

10 R 2 is Ci-Ce alkyl, Ci-Ce fluoroalkyl, or 3 to 6-membered cycloalkyl;

R 7 is C1-C3 alkyl;

R 8 is C1-C3 alkyl; and

R 9 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

15 X ® and X* are, independently, N or CH;

R 11 is hydrogen or C1-C3 alkyl; and

R 21 is hydrogen or C1-C3 alkyl.

[92] The conjugate of paragraph [91], or salt thereof, wherein M has the structure of Formula Ve:

20 Formula Ve wherein A is optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally 25 substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene; and

R 9 is optionally substituted Ci-Ce alkyl, optionally substituted Ci-Ce heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl.

[93] The conjugate, or salt thereof, of any one of paragraphs [90] to [92], wherein the linker has 30 the structure of Formula II:

818

SUBSTITUTE SHEET (RULE 26) A 1 -(B>(C 1 )g-(B 2 )h-(D 1 )-(B 3 )r(C 2 )j-(B 4 )k-A 2 Formula II where A' is a bond between the linker and B; A 2 is a bond between W and the linker; B 1 , B 2 , B 3 , and B 4 each, independently, is selected from optionally substituted C1-C2 alkylene, optionally substituted 5 C1-C3 heteroalkylene, O, S, and NR N ; R N is hydrogen, optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, optionally substituted C2-C4 alkynyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted C1-C7 heteroalkyl; C 1 and C 2 are each, independently, selected from carbonyl, thiocarbonyl, sulphonyl, or phosphoryl; f, g, h, I, j, and k are each, independently, 0 or 1 ; and D 1 is optionally substituted C1-C10 alkylene, optionally 10 substituted C2-C10 alkenylene, optionally substituted C2-C10 alkynylene, optionally substituted 3 to 14- membered heterocycloalkylene, optionally substituted 5 to 10-membered heteroarylene, optionally substituted 3 to 8-membered cycloalkylene, optionally substituted 6 to 10-membered arylene, optionally substituted C2-C10 polyethylene glycolene, or optionally substituted C1-C10 heteroalkylene, or a chemical bond linking A 1 -(B>(C 1 ) g -(B 2 ) h - to -(B 3 )i-(C 2 )j-(B 4 )ic-A 2 .

15 [94] The conjugate, or salt thereof, of any one of paragraphs [90] to [93], wherein the monovalent organic moiety is a protein.

[95] The conjugate, or salt thereof, of paragraph [94], wherein the protein is a Ras protein.

[96] The conjugate, or salt thereof, of paragraph [95], wherein the Ras protein is K-Ras G12C, K- Ras G13C, H-Ras G12C, H-Ras G13C, N-Ras G12C, or N-Ras G13C.

20 [97] The conjugate, or salt thereof, of any one of paragraphs [93] to [96], wherein the linker is bound to the monovalent organic moiety through a bond to a sulfhydryl group of an amino acid residue of the monovalent organic moiety.

[98] A method of treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound, or a pharmaceutically

25 acceptable salt thereof, of any one of paragraphs [1 ] to [88] or a pharmaceutical composition of paragraph [89].

[99] The method of paragraph [98], wherein the cancer is pancreatic cancer, colorectal cancer, non-small cell lung cancer, or endometrial cancer.

[100] The method of paragraph [98] or [99], wherein the cancer comprises a Ras mutation.

30 [101] The method of paragraph [100], wherein the Ras mutation is K-Ras G12C, K-Ras G13C, H-Ras G12C, H-Ras G13C, N-Ras G12C, or N-Ras G13C.

[102] A method of treating a Ras protein-related disorder in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1 ] to [88] or a pharmaceutical

35 composition of paragraph [89].

[103] A method of inhibiting a Ras protein in a cell, the method comprising contacting the cell with an effective amount of a compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [88] or a pharmaceutical composition of paragraph [89],

[104] The method of paragraph [102] or [103], wherein the Ras protein is K-Ras G12C, K-Ras

40 G13C, H-Ras G12C, H-Ras G13C, N-Ras G12C, or N-Ras G13C.

[105] The method of paragraph [103] or [104], wherein the cell is a cancer cell.

819

SUBSTITUTE SHEET (RULE 26) [106] The method of paragraph [105], wherein the cancer cell is a pancreatic cancer cell, a colorectal cancer cell, a non-small cell lung cancer cell, or an endometrial cancer cell.

Examples

5 The disclosure is further illustrated by the following examples and synthesis examples, which are not to be construed as limiting this disclosure in scope or spirit to the specific procedures herein described. It is to be understood that the examples are provided to illustrate certain embodiments and that no limitation to the scope of the disclosure is intended thereby. It is to be further understood that resort may be had to various other embodiments, modifications, and equivalents thereof which may 10 suggest themselves to those skilled in the art without departing from the spirit of the present disclosure or scope of the appended claims.

Chemical Syntheses

Definitions used in the following examples and elsewhere herein are:

CHaClz, DCM Methylene chloride, Dichloromethane CHsCN, MeCN Acetonitrile

Cul Copper (I) iodide

DIPEA Diisopropylethyl amine

DMF N.N-Dimethylformamide

EtOAc Ethyl acetate h hour H 2 O Water

HCI Hydrochloric acid

K3PO4 Potassium phosphate (tribasic)

MeOH Methanol Na 2 SO 4 Sodium sulfate

NMR N-methyl pyrrolidone

Pd(dppf)Cla [1 ,1 '-Bis(diphenylphosphino)ferrocene]dichloropalladium(ll)

15

Instrumentation

Mass spectrometry data collection took place with a Shimadzu LCMS-2020, an Agilent 1260LC- 6120/6125MSD, a Shimadzu LCMS-2010EV, or a Waters Acquity UPLC, with either a QDa detector or SQ Detector 2. Samples were injected in their liquid phase onto a C-18 reverse phase. The compounds 20 were eluted from the column using an acetonitrile gradient and fed into the mass analyzer. Initial data analysis took place with either Agilent ChemStation, Shimadzu LabSolutions, or Waters MassLynx. NMR data was collected with either a Bruker AVANCE III HD 400MHz, a Bruker Ascend 500MHz instrument, or a Varian 400MHz, and the raw data was analyzed with either TopSpin or Mestrelab Mnova.

820

SUBSTITUTE SHEET (RULE 26) Synthesis of Intermediates

Intermediate 1. Synthesis of 3-(5-bromo-1 -ethyl-2-[2-[(1 S)-1 -methoxyethyQpyridln-3- yl]indol-3-yl)-2,2-dimethylpropan-1-ol

5

Step 1. To a mixture of 3-((fert-butyldiphenylsilyl)oxy)-2,2-dimethylpropanoyl chloride (65 g, 137 mmol, crude) in DCM (120 mL) at 0 °C under an atmosphere of Na was added 1 M SnCU in DCM (137 mL, 137 mmol) slowly. The mixture was stirred at 0 °C for 30 min, then a solution of 5-bromo-1 H- indole (26.8 g, 137 mmol) in DCM (40 mL) was added dropwise. The mixture was stirred at 0 °C for 45 min, then

10 diluted with EtOAc (300 mL), washed with brine (100 mL x 4), dried over NaaSCU, and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 1-(5-bromo-1 H- indol-3-yl)-3-((tert-butyldiphenylsilyl)oxy)-2,2-dimethylpro pan-1- one (55 g, 75% yield). LCMS (ESI): m/z [M+Na] calc’d for CzsHaaBrNOaSiNa 556.1 ; found 556.3.

Step 2. To a mixture of 1-(5-bromo-1 H-indol-3-yl)-3-((tert-butyldiphenylsilyl)oxy)-2,2-

15 dimethylpropan-1 -one (50 g, 93.6 mmol) in THF (100 mL) at 0 °C under an atmosphere of N 2 was added UBH4 (6.1 g, 281 mmol). The mixture was heated to 60 °C and stirred for 20 h, then MeOH (10 mL) and EtOAc (100 mL) were added and the mixture washed with brine (50 mL), dried over Na 2 SO 4 , filtered, and the filtrate concentrated under reduced pressure. The residue was diluted with DCM (50 mL), cooled to 10 °C and diludine (9.5 g, 37.4 mmol) and TSOH.H 2 O (890 mg, 4.7 mmol) added. The mixture was stirred

20 at 10 °C for 2 h, filtered, the filtrate concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 1-(5-bromo-1 H-indol-3-yl)-3-((tert-butyldiphenylsilyl)oxy)-2,2- dimethylpropan-1 -one (41 g, 84% yield). LCMS (ESI): m/z [M+H] calc’d for C2gH34BrNOSi 519.2; found 520.1 ; 1 H NMR (400 MHz, CDCh) 67.96 (s, 1H), 7.75 - 7.68 (m, 5H), 7.46 - 7.35 (m, 6H), 7.23 - 7.19 (m, 2H), 6.87 (d, J= 2.1 Hz, 1H), 3.40 (s, 2H), 2.72 (s, 2H), 1.14 (s, 9H), 0.89 (s, 6H).

25 Step 3. To a mixture of 1-(5-bromo-1 H-indol-3-yl)-3-((terf-butyldiphenylsilyl)oxy)-2,2- dimethylpropan-1 -one (1.5 g, 2.9 mmol) and I2 (731 mg, 2.9 mmol) in THF (15 mL) at rt was added AgOTf (888 mg, 3.5 mmol). The mixture was stirred at rt for 2 h, then diluted with EtOAc (200 mL) and washed with saturated NaaSaOa (100 mL), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography

821

SUBSTITUTE SHEET (RULE 26) to give 5-bromo-3-(3-((Zert-butyldiphenylsilyl)oxy)-2,2-dimethylprop yl)-2-iodo-1 H- indole (900 mg, 72% yield) as a solid. 1 H NMR (400 MHz, DMSO-dfe) 6 11.70 (s, 1 H), 7.68 (d, J = 1.3 Hz, 1 H), 7.64 - 7.62 (m, 4H), 7.46 - 7.43 (m, 6H), 7.24 - 7.22 (d, 1H), 7.14 - 7.12 (dd, J= 8.6, 1 .6 Hz, 1H), 3.48 (s, 2H), 2.63 (s, 2H), 1.08 (s, 9H), 0.88 (s, 6H).

5 Step 4. To a stirred mixture of HCOOH (66.3 g, 1.44 mol) in TEA (728 g, 7.2 mol) at 0 °C under an atmosphere of Ar was added (4S,5S)-2-chloro-2-methyl-1 -(4-methylbenzenesulfonyl)-4,5-diphenyl-1 ,3- diaza-2-ruthenacyclopentane cymene (3.9 g, 6.0 mmol) portion-wise. The mixture was heated to 40 °C and stirred for 15 min, then cooled to rt and 1-(3-bromopyridin-2-yl)ethanone (120 g, 600 mmol) added in portions. The mixture was heated to 40 °C and stirred for an additional 2 h, then the solvent was

10 concentrated under reduced pressure. Brine (2 L) was added to the residue, the mixture was extracted with EtOAc (4 x 700 mL), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give (1 S)-1-(3- bromopyridin-2-yl)ethanol (100 g, 74% yield) a an oil. LCMS (ESI): m/z [M+H] calc'd for CyHeBrNO 201.1 ; found 201.9.

15 Step 5. To a stirred mixture of (1 S)-1-(3-bromopyridin-2-yl)ethanol (100 g, 495 mmol) in DMF (1 L) at 0 °C was added NaH, 60% dispersion in oil (14.25 g, 594 mmol) in portions. The mixture was stirred at 0 °C for 1 h. Mel (140.5 g, 990 mmol) was added dropwise at 0 °C and the mixture was allowed to warm to rt and stirred for 2 h. The mixture was cooled to 0 °C and saturated NH4CI (5 L) was added. The mixture was extracted with EtOAc (3 x 1.5 L), dried over anhydrous Na 2 SO 4 and filtered. The filtrate was 20 concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 3-bromo-2-[(1 S)-1 -methoxyethyljpyridine (90 g, 75% yield) as an oil. LCMS (ESI): m/z [M+H] calc'd for CsHioBrNO 215.0; found 215.9.

Step 6. To a stirred mixture of 3-bromo-2-[(1 S)-1 -methoxyethyljpyridine (90 g, 417 mmol) and Pd(dppf)Cl 2 (30.5 g, 41.7 mmol) in toluene (900 mL) at rt under an atmosphere of Ar was added 25 bis(pinacolato)diboron (127 g, 500 mmol) and KOAc (81.8 g, 833 mmol) in portions. The mixture was heated to 100 °C and stirred for 3 h. The filtrate was concentrated under reduced pressure and the residue was purified by AI2O3 column chromatography to give 2-[(1 S)-1 -methoxyethyl]-3-(4, 4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridine (100 g, 63% yield) as a semi-solid. LCMS (ESI): m/z [M+H] calc’d for C14H22BNO3263.2; found 264.1.

30 Step 7. To a stirred mixture of 5-bromo-3-[3-[(tert-butyldiphenylsilyl)oxy]-2,2-dimethylprop yl]-2- iodo-1 H- indole (140 g, 217 mmol) and 2-[(1 S)-1-methoxyethyl]-3-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan- 2-yl)pyridine (100 g, 380 mmol) in 1 ,4-dioxane (1.4 L) at rt under an atmosphere of Ar was added K2CO3 (74.8 g, 541 mmol), Pd(dppf)Cl 2 (15.9 g, 21.7 mmol), and H 2 O (280 mL) in portions. The mixture was heated to 85 °C and stirred for 4 h, then cooled, H 2 O (5 L) added, and the mixture extracted with EtOAc 35 (3 x 2 L). The combined organic layers were washed with brine (2 x 1 L), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 5-bromo-3-[3-[(tert-butyldiphenylsilyl)oxy]-2,2-dimethylprop yl]-2-[2- [(1 S)-1 -methoxyethyl]pyridin-3-yl]-1 H-indole (71 g, 45% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 3 7H43BrN 2 02Si 654.2; found 655.1.

40 Step 8. To a stirred mixture of 5-bromo-3-[3-[(tert-butyldiphenylsilyl)oxy]-2,2-dimethylprop yl]-2- [2-[(1 S)-1-methoxyethyl]pyridin-3-yl]-1 H- indole (71 g, 108 mmol) in DMF (0.8 L) at 0 °C under an atmosphere of N 2 was added CS2CO3 (70.6 g, 217 mmol) and Etl (33.8 g, 217 mmol) in portions. The

822

SUBSTITUTE SHEET (RULE 26) mixture was warmed to rt and stirred for 16 h then H 2 O (4 L) added and the mixture extracted with EtOAc (3 x 1.5 L). The combined organic layers were washed with brine (2 x 1 L), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 5-bromo-3-[3-[(te/t-butyldiphenylsilyl)oxy]-2,2-dimethylprop yl]-1-ethyl- 5 2-[2-[(1 S)-1 -methoxyethyl]pyridin-3-yl]indole (66 g, 80% yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for

Csel-UyBrNaOzSi 682.3; found 683.3.

Step 9. To a stirred mixture of TBAF (172.6 g, 660 mmol) in THF (660 mL) at rt under an atmosphere of N 2 was added 5-bromo-3-[3-[(te/i-butyldiphenylsilyl)oxy]-2,2-dimethylprop yl]-1-ethyl-2-[2- [(1 S)-1-methoxyethyl]pyridin-3-yl]indole (66 g, 97 mmol) in portions. The mixture was heated to 50 °C and 10 stirred for 16 h, cooled, diluted with HzO (5 L), and extracted with EtOAc (3 x 1.5 L). The combined organic layers were washed with brine (2 x 1 L), dried over anhydrous Na 2 SO 4 , and filtered. After filtration, the filtrate was concentrated under reduced pressure. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 3-(5-bromo-1 - ethyl-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]indol-3-yl)-2,2-dimethylpropa n-1-ol (30 g, 62% yield) as a 15 solid. LCMS (ESI): m/z [M+H] calc’d for CaaHzgBrNzOa 444.1 ; found 445.1.

Intermediate 1. Alternative Synthesis through Fisher Indole Route. k Intermediate 1

Step 1. To a mixture of APrMgCI (2M in in THF, 0.5 L) at -10 °C under an atmosphere of Na was 20 added />BuLi, 2.5 M in hexane (333 mL, 833 mmol) dropwise over 15 min. The mixture was stirred for 30 min at -10 °C then 3-bromo-2-[(1 S)-1 -methoxyethyljpyridine (180 g, 833 mmol) in THF (0.5 L) added dropwise over 30 min at -10 °C. The resulting mixture was warmed to -5 °C and stirred for 1 h, then 3,3- dimethyloxane-2,6-dione (118 g, 833 mmol) in THF (1.2 L) was added dropwise over 30 min at -5 °C. The mixture was warmed to 0 °C and stirred for 1.5 h, then quenched with the addition of pre-cooled 4M HCI 25 in 1 ,4-dioxane (0.6 L) at 0 °C to adjust pH ~5. The mixture was diluted with ice-water (3 L) and extracted with EtOAc (3 x 2.5 L). The combined organic layers were dried over anhydrous Na 2 SO 4 , filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography to give 5-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]-2,2-dimethyl-5-oxopentanoic acid (87 g, 34% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C15H21NO4279.2; found 280.1.

30 Step 2. To a mixture of 5-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]-2,2-dimethyl-5-oxopentanoic acid

(78 g, 279 mmol) in EtOH (0.78 L) at rt under an atmosphere of Na was added (4-bromophenyl)hydrazine

823

SUBSTITUTE SHEET (RULE 26) HCI salt (68.7 g, 307 mmol) in portions. The mixture was heated to 85 °C and stirred for 2 h, cooled to rt, then 4M HCI in 1 ,4-dioxane (69.8 mL, 279 mmol) added dropwise. The mixture was heated to 85 °C and stirred for an additional 3 h, then concentrated under reduced pressure, and the residue was dissolved in TFA (0.78 L). The mixture was heated to 60 °C and stirred for 1.5 h, concentrated under reduced 5 pressure, and the residue adjusted to pH ~5 with saturated NaHCOs, then extracted with EtOAc (3 x 1.5 L). The combined organic layers were dried over anhydrous Na 2 SO 4 , filtered, the filtrate concentrated under reduced pressure, and the residue was purified by silica gel column chromatography to give 3-(5- bromo-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]-1 /+indol-3-yl)-2,2-dimethylpropanoic acid and ethyl (S)-3- (5-bromo-2-(2-(1-methoxyethyl)pyridin-3-yl)-1 H- indol-3-yl)-2,2-dimethylpropanoate (78 g, crude). LCMS 10 (ESI): m/z [M+H] calc’d for CaiHaaBrNaOa 430.1 and CasHayBrNaOa 458.1 ; found 431.1 and 459.1.

Step 3. To a mixture of 3-(5-bromo-2-[2-[(1 S)-1 -methoxyethyl]pyridin-3-yl]-1 H- indol-3-yl)-2,2- dimethylpropanoic acid and ethyl (S)-3-(5-bromo-2-(2-(1-methoxyethyl)pyridin-3-yl)-1 H-indol-3-yl)-2,2- dimethylpropanoate (198 g, 459 mmol) in DMF (1.8 L) at 0 °C under an atmosphere of Na was added CsaCOa (449 g, 1.38 mol) in portions. Etl (215 g, 1.38 mmol) in DMF (200 mL) was then added dropwise 15 at 0 °C. The mixture was warmed to rt and stirred for 4 h then diluted with brine (5 L) and extracted with EtOAc (3 x 2.5 L). The combined organic layers were washed with brine (2 x 1.5 L), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give ethyl 3-(5-bromo-1 -ethyl-2-[2-[(1 S)-1 -methoxyethyl]pyridin-3- yl]indol-3-yl)-2,2-dimethylpropanoate (160 g, 57% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for 20 CasHsiBrNaOs 486.2; found 487.2.

Step 4. To a mixture of ethyl 3-(5-bromo-1 -ethyl-2-[2-[(1 S)-1 -methoxyethyl]pyridin-3-yl]indol-3- yl)-2,2-dimethylpropanoate (160 g, 328 mmol) in THF (1.6 L) at 0 °C under an atmosphere of Na was added LiBH* (28.6 g, 1.3 mol). The mixture was heated to 60 °C for 16 h, cooled, and quenched with precooled (0 °C) aqueous NH4CI (5 L). The mixture was extracted with EtOAc (3 x 2 L) and the combined 25 organic layers were washed with brine (2 x 1 L), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give to two atropisomers (as single atropisomers) of 3-(5-bromo-1 -ethyl-2-(2-((S)-1 - methoxyethyl)pyridin-3-yl)-1 H-indol-3-yl)-2,2-dimethylpropan-1-ol (60 g, 38% yield) and (40 g, 26% yield) both as solids. LCMS (ESI): m/z [M+H] calc'd for CaaHagBrNaOa 444.1 ; found 445.2.

30

824

SUBSTITUTE SHEET (RULE 26) Intermediate 2 and Intermediate 4. Synthesis of (S)-1-((S)-2-((tert-butoxycarbonyl)amlno)-3- (3-(4,4,5,5-tetramethyl-1 f 3,2-dk>xaborolan-2-yl)-5-

((trllsopropylsllyl)oxy)phenyl)propanoyl)hexahydropyrldaz lne-3-carboxylate

Intermediate 2.

5 Step 1. To a mixture of (S)-methyl 2-(fert-butoxycarbonylamino)-3-(3-hydroxyphenyl)propanoate (10.0 g, 33.9 mmol) in DCM (100 mL) was added imidazole (4.6 g, 67.8 mmol) and TIPSCI (7.8 g, 40.7 mmol). The mixture was stirred at rt overnight then diluted with DCM (200 mL) and washed with H 2 O (150 mL x 3). The organic layer was dried over anhydrous NaaSO*, filtered, concentrated under reduced pressure, and the residue was purified by silica gel column chromatography to give (S)-methyl 2 -{tert- 10 butoxycarbonylamino)-3-(3-(triisopropylsilyloxy)phenyl)-prop anoate (15 g, 98% yield) as an oil. LCMS (ESI): m/z [M+Na] calc’d for Ca+H+iNOsSiNa 474.3; found 474.2.

Step 2. A mixture of (S)-methyl 2-(tert-butoxycarbonylamino)-3-(3-(triisopropylsilyloxy)phen yl)- propanoate (7.5 g, 16.6 mmol), PinBa (6.3 g, 24.9 mmol), [lr(OMe)(COD)]a(1.1 g, 1.7 mmol), and 4 -tert- butyl-2-(4-tert-butyl-2-pyridyl)pyridine (1.3 g, 5.0 mmol) was purged with Ar ( x3), then THF (75 mL) was 15 added and the mixture placed under an atmosphere of Ar and sealed. The mixture was heated to 80 °C and stirred for 16 h, concentrated under reduced pressure, and the residue was purified by silica gel column chromatography to give (S)-methyl 2-(tert-butoxycarbonylamino)-3-(3-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)-5-(triisopropylsilyloxy)phenyl)-propanoat e (7.5 g, 78% yield) as a solid. LCMS (ESI): m/z [M+Na] calc'd for CsoHszBNOSiNa 600.4; found 600.4; Ή NMR (300 MHz, CDaOD) δ 7.18 (s, 1 H), 20 7.11 (s, 1 H), 6.85 (s, 1 H), 4.34 (m, 1 H), 3.68 (s, 3H), 3.08 (m, 1 H), 2.86 (m, 1 H), 1.41 - 1.20 (m, 26H),

1.20 - 1.01 (m, 22H), 0.98 - 0.79 (m, 4H).

Step 3. To a mixture of triisopropylsilyl (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(4, 4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)-5-((triisopropylsilyl)oxy)phenyl)pro panoate (4.95 g, 6.9 mmol) in MeOH (53 mL) at 0 °C was added LiOH (840 mg, 34.4 mmol) in H 2 O (35 mL). The mixture was stirred at 25 0 °C for 2 h, then acidified to pH ~5 with 1 M HCI and extracted with EtOAc (250 mL x 2). The combined organic layers were washed with brine (100 mL x 3), dried over anhydrous NaaSOA, filtered, and the filtrate concentrated under reduced pressure to give (S)-2-((fert-butoxycarbonyl)amino)-3-(3-(4,4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)-5-((triisopropylsilyl)oxy)phenyl)pro panoic acid (3.7 g, 95% yield),

825

SUBSTITUTE SHEET (RULE 26) which was used directly in the next step without further purification. LCMS (ESI): m/z [M+NhU] calc’d for C29HsoBN07SiNH4581.4; found 581.4.

Step 4. To a mixture of methyl (S)-hexahydropyridazine-3-carboxylate (6.48 g, 45.0 mmol) in DCM (200 mL) at 0 °C was added NMM (41.0 g, 405 mmol), (S)-2-((tert-butoxycarbonyl)amino)-3-(3-

5 (4,4,5,5-tetramethyM ,3,2-dioxaborolan-2-yl)-5-((triisopropylsilyl)oxy)phenyl)pro panoic acid (24 g,

42.6 mmol) in DCM (50 mL) then HOBt (1.21 g, 9.0 mmol) and EDCI HCI salt (12.9 g, 67.6 mmol). The mixture was warmed to rt and stirred for 16 h, then diluted with DCM (200 mL) and washed with H 2 O (3 x 150 mL). The organic layer was dried over anhydrous NaaSO, filtered, the filtrate concentrated under reduced pressure, and the residue was purified by silica gel column chromatography to give methyl (S)-1 -

10 ((S)-2-((fert-butoxycarbonyl)amino)-3-(3-(4,4,5,5-tetramethy l-1,3,2-dioxaborolan-2-yl)-5- ((triisopropylsilyl)oxy)phenyl)propanoyl)hexahydropyridazine -3-carboxylate (22 g, 71% yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for CasHeoBNsOsSi 689.4; found 690.5.

Intermediate 3. Synthesis of N-((S)-1-acryloylpyrrolldine-3-carbonyl)-N-methyl-L-valine

TFAZDCM (1/10)

20°C, 3h Ηο^γΐΡ ' ΐΓ

15 Intemwdlst· 3

Step 1. To a mixture of (S)-1-(tert-butoxycarbonyl)pyrrolidine-3-carboxylic acid (2.2 g, 10.2 mmol) in DMF (10 mL) at rt was added HATU (7.8 g, 20.4 mmol) and DIPEA (5 mL). After stirring at rt for 10 min, fert-butyl methyl-L-valinate (3.8g, 20.4 mmol) in DMF (10 mL) was added. The mixture was stirred at rt for 3 h, then diluted with DCM (40 mL) and H 2 O (30 mL). The aqueous and organic layers were

20 separated and the organic layer was washed with H 2 O (3 χ 30 mL), brine (30 mL), dried over anhydrous NaaSO*, and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give (S)-fe/t- butyl 3-(((S)-1 -(fert-butoxy)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamoyl)pyrrolidine-1-carboxylate (3.2 g, 82% yield) as an oil. LCMS (ESI): m/z [M+Na] calc'd for CaoHaeNaOsNa 407.3; found 407.2.

25 Step 2. A mixture of (S)-tert-butyl 3-(((S)-1 -(te/t-butoxy)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamoyl)pyrrolidine-1-carboxylate (3.2 g, 8.4 mmol) in DCM (13 mL) and TFA (1.05 g, 9.2 mmol) was stirred at rt for 5 h. The mixture was concentrated under reduced pressure to give ( S)-tert - butyl 3-methyl-2-((S)-N-methylpyrrolidine-3-carboxamido)butanoate (2.0 g, 84% yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for CisHaeNaOs 284.2; found 285.2.

30 Step 3. To a mixture of (S)-tert- butyl 3-methyl-2-((S)-N-methylpyrrolidine-3- carboxamido)butanoate (600 mg, 2.1 mmol) in DCM (6 mL) at 0 °C was added TEA (342 mg, 3.36 mmol). After stirring at 0 °C for 10 mins, acryloyl chloride (284 mg, 3.2 mmol) in DCM (10 mL) was added. The mixture was warmed to rt and stirred for 24 h, then diluted with DCM (30 mL) and H 2 O (30 mL). The aqueous and organic layers were separated and the organic layer was washed with H 2 O (3 χ 30 mL),

35 brine (30 mL), dried over anhydrous NaaSO.», and filtered. The filtrate was concentrated under reduced

826

SUBSTITUTE SHEET (RULE 26) pressure and the residue was purified by silica gel column chromatography to give fert-butyl N-((S)-1- acryloylpyrrolidine-3-carbonyl)-/\Amethyl-L-valinate (500 mg, 70% yield) as an oil.

Step 4. To a mixture of fert-butyl N-((S)-1-acryloylpyrrolidine-3-carbonyl)-N-methyl-L-valinate (100 mg, 0.29 mmol) in DCM (3.0 mL) at 15 °C was added TFA (0.3 mL). The mixture was warmed to rt 5 and stirred for 5 h, then the mixture was concentrated under reduced pressure to give N-((S)-1 - acryloylpyrrolidine-3-carbonyl)-N-methyl-L-valine (150 mg) as a solid. The crude product was used directly in the next step without further purification. LCMS (ESI): m/z [M+H] calc’d for C14H22N 2 O4282.2; found 283.2.

10 Intermediate 5. Synthesis of tert-butyl ((63S,4S)-11 -ethyl-12-(2-((S)-1 -methoxyethyl)pyridin- 3-yl)-10,10-dlmethyl-5,7-dloxo-25-((trllsopropylsllyl)oxy)-6 1 ,62,63, 64, 65,66-hexahydro-l 1 H-8-oxa- 1 (5,3)-lndola-6(1 ,3)-pyrldazina-2(1 ,3)-benzenacyck>undecaphane-4-yl)carbamate.

Step 1. To a stirred mixture of 3-(5-bromo-1 -ethyl-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]indol-3-

15 yl)-2,2-dimethylpropan-1 -ol (30 g, 67 mmol) and methyl (3S)-1-[(2S)-2-[(fert-butoxycarbonyl)amino]-3-[3- (4,4,5 I 5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-5-[(triisopropylsilyl)oxy]phenyl]pro panoyl]-1 I 2-diazinane-3- carboxylate (55.8 g, 80.8 mmol) in 1 ,4-dioxane (750 mL) at rt under an atmosphere of Ar was added NaaCOs (17.9 g, 168.4 mmol), Pd(DtBPF)Clz (4.39 g, 6.7 mmol), and H 2 O (150.00 mL) in portions. The mixture was heated to 85 °C and stirred for 3 h, cooled, diluted with H 2 O (2 L), and extracted with EtOAc

20 (3 x 1 L). The combined organic layers were washed with brine (2 x 500 mL), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give methyl (3S)-1 -[(2S)-2-[(terf-butoxycarbonyl)amino]-3-[3-[1 - ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]indol-5-yl]-5- [(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3-carboxylate (50 g, 72% yield) as a solid. LCMS

25 (ESI): m/z [M+H] calc’d for CsaHyrNsOsSi 927.6; found 928.8.

Step 2. To a stirred mixture of methyl (3S)-1 -[(2S)-2-[(fert-butoxycarbonyl)amino]-3-[3-[1 -ethyl-3- (3-hydroxy-2,2-dimethylpropyl)-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]indol-5-yl]-5-

827

SUBSTITUTE SHEET (RULE 26) [(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3-carboxylate (50 g, 54 mmol) in DCE (500 mL) at rt was added trimethyltin hydroxide (48.7 g, 269 mmol) in portion. The mixture was heated to 65 °C and stirred for 16 h, then filtered and the filter cake washed with DCM (3 x 150 mL). The filtrate was concentrated under reduced pressure to give (3S)-1-[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[1-ethyl-3- 5 (S-hydroxy^^-dimethylpropyll^-p-Iil Si-l-methoxyethyllpyridin-S-yllindol-S-yll-S-

[(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3-carboxylic acid (70 g t crude), which was used directly in the next step without further purification. LCMS (ESI): m/z [M+H] calc'd for CsiHTsNsOaSi 913.5; found 914.6.

Step 3. To a stirred mixture of (3S)-1 -[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[1 -ethyl-3-(3-

10 hydroxy-2,2-dimethylpropyl)-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]indol-5-yl]-5- [(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3-carboxylic acid (70 g) in DCM (5 L) at 0 °C under an atmosphere of N 2 was added DIPEA (297 g, 2.3 mol), HOBT (51.7 g, 383 mmol) and EDCI (411 g, 2.1 mol) in portions. The mixture was warmed to rt and stirred for 16 h, then diluted with DCM (1 L), washed with brine (3 x 1 L), dried over anhydrous NaaSO*, and filtered. The filtrate was concentrated under

15 reduced pressure and the residue was purified by silica gel column chromatography to give fert-butyl ((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5 ,7-dioxo-2 5 - ((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 Ή-8-oxa-l (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate (36 g, 42% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for CsiHreNsOySi 895.5; found 896.5.

20

Intermediate 6. Synthesis of tert-butyl N-[(8S, 14S)-21-iod o-18,18-dimethy 1-9,15-dioxo-4- [(triisopropylsilyl)oxy]-16-oxa-10,22,28- triazapentacyck>[18.5.2.1 A [2,6].1 A [10,14l.0 A [23,27]]nonacosa-1(26), 2,4, 6(29), 20, 23(27), 24-heptaen- 8-ylJcarbamate.

TBA5

.Sr .Br A¾0 Br

N N N

H h H

25

Step 1. This reaction was undertaken on 5-batches in parallel on the scale illustrated below. Into a 2L round-bottom flasks each were added 5-bromo-3-[3-[(tert-butyldiphenylsilyl)oxy]-2,2- dimethylpropyl]-1 H- indole (100 g, 192 mmol) and TBAF (301.4 g, 1.15 mol) in THF (1.15 L) at rt. The resulting mixture was heated to 50 °C and stirred for 16 h, then the mixture was concentrated under

828

SUBSTITUTE SHEET (RULE 26) reduced pressure. The combined residues were diluted with H 2 O (5 L) and extracted with EtOAc (3 x 2 L). The combined organic layers were washed with brine (2 x 1.5 L), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 3-(5-bromo-1 H- indol-3-yl)-2,2-dimethylpropan-1-ol (310 g, crude) as a 5 solid. LCMS (ESI): m/z [M+H] calc’d for CiaHieBrNO 281.0 and 283.0; found 282.1 and 284.1.

Step 2. This reaction was undertaken on two batches in parallel on the scale illustrated below.

To a stirred mixture of 3-(5-bromo-1 H- indol-3-yl)-2,2-dimethylpropan-1 -ol (135 g, 478 mmol) and TEA (145.2 g, 1.44 mol) in DCM (1.3 L) at 0 °C under an atmosphere of Nzwas added AcaO (73.3 g, 718 mmol) and DMAP (4.68 g, 38.3 mmol) in portions. The resulting mixture was stirred for 10 min at 0 °C,

10 then washed with H 2 O (3 x 2 L). The organic layers from each experiment were combined and washed with brine (2 x 1 L), dried over anhydrous NaaSO*, and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by column chromatography to give 3-(5-bromo-1 H-indol-3- yl)-2,2-dimethylpropyl acetate (304 g, 88% yield) as a solid. 1 H NMR (400 MHz, DMSO-db) 6 11.16 - 11.11 (m, 1H), 7.69 (d, J= 2.0 Hz, 1H), 7.32 (d, J= 8.6 Hz, 1H), 7.19 - 7.12 (m, 2H), 3.69 (s, 2H), 2.64 (s, 15 2H), 2.09 (s, 3H), 0.90 (s, 6H).

Step 3. This reaction was undertaken on four batches in parallel on the scale illustrated below.

Into a 2L round-bottom flasks were added methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-(3- (4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-5-[(triisopropylsilyl)oxy]phenyl]pro panoate (125 g, 216 mmol), 1 ,4-dioxane (1 L), H 2 O (200 mL), 3-(5-bromo-1 H-indol-3-yl)-2,2-dimethylpropyl acetate (73.7 g,

20 227 mmol), K2CO3 (59.8 g, 433 mmol), and Pd(DtBPF)Cla (7.05 g, 10.8 mmol) at rt under an atmosphere of Ar. The resulting mixture was heated to 65 °C and stirred for 2 h, then diluted with H 2 O (10 L) and extracted with EtOAc (3 x 3 L). The combined organic layers were washed with brine (2 x 2 L), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by column chromatography to give methyl (2S)-3-(3-[3-[3-(acetyloxy)-2,2-dimethylpropyl]-1 H- 25 indol-5-yl]-5-[(triisopropylsilyl)oxy]phenyl)-2-[(tert-butox ycarbonyl)amino]propanoate (500 g, 74% yield) as an oil. LCMS (ESI): m/z [M+Na] calc'd for CsgHseNaOrSINa 717.4; found 717.3.

Step 4. This reaction was undertaken on three batches in parallel on the scale illustrated below.

To a stirred mixture of methyl (2S)-3-(3-[3-[3-(acetyloxy)-2,2-dimethylpropyl]-1 H- indol-5-yl]-5- [(triisopropylsilyl)oxy]phenyl)-2-[(te/t-butoxycarbonyl)amin o]propanoate (150 g, 216 mmol) and NaHCOs 30 (21.76 g, 259 mmol) in THF (1.5 L) was added AgOTf (66.5 g, 259 mmol) in THF dropwise at 0 °C under an atmosphere of nitrogen. I2 (49.3 g, 194 mmol) in THF was added dropwise over 1 h at 0 °C and the resulting mixture was stirred for an additional 10 min at 0 °C. The combined experiments were diluted with aqueous NaaSpOa (5 L) and extracted with EtOAc (3 x 3 L). The combined organic layers were washed with brine (2 x 1.5 L), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated 35 under reduced pressure and the residue was purified by column chromatography to give methyl (2S)-3- (3-[3-[3-(acetyloxy)-2,2-dimethylpropyl]-2-iodo-1 H-indol-5-yl]-5-[(triisopropylsilyl)oxy]phenyl)-2-[(tert- butoxycarbonyl)amino]propanoate (420 g, 71% yield) as an oil. LCMS (ESI): m/z [M+Na] calc’d for C 3 9H57lN 2 07SiNa, 843.3; found 842.9.

Step 5. This reaction was undertaken on three batches in parallel on the scale illustrated below.

40 To a 2L round-bottom flask were added methyl (2S)-3-(3-[3-[3-(acetyloxy)-2,2-dimethylpropyl]-2- iodo-1 H-indol-5-yl]-5-[(triisopropylsilyl)oxy]phenyl)-2-[(tert-but oxycarbonyl)amino]propanoate (140 g, 171 mmol), MeOH (1.4 L) and K3PO4 (108.6 g, 512 mmol) at 0 °C. The mixture was warmed to rt and stirred

829

SUBSTITUTE SHEET (RULE 26) for 1 h, then the combined experiments were diluted with H 2 O (9 L) and extracted with EtOAc (3 x 3 L). The combined organic layers were washed with brine (2 x 2 L), dried over anhydrous Na 2 SO 4 , filtered, and the filtrate was concentrated under reduced pressure to give methyl (2S)-2-[(tert- butoxycarbonyl)amino]-3-[3-[3-(3-hydroxy-2,2-dimethylpropyl) -2-iodo-1 H- indol-5-yl]-5- 5 [(triisopropylsilyl)oxy]phenyl]propanoate (438g, crude) as a solid. LCMS (ESI): m/z [M+Na] calc’d for CsyHssINzOeSiNa 801.3; found 801.6.

Step 6. This reaction was undertaken on three batches in parallel on the scale illustrated below.

To a stirred mixture of methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[3-(3-hydroxy-2,2- dimethylpropyl)-2-iodo-1 H- indol-5-yl]-5-[(triisopropylsilyl)oxy]phenyl]propanoate (146 g, 188 mmol) in THF 10 (1.46 L) was added LiOH (22.45 g, 937 mmol) in H 2 O (937 mL) dropwise at 0 °C. The resulting mixture was warmed to rt and stirred for 1.5 h [note: LCMS showed 15% de-TIPS product]. The mixture was acidified to pH 5 with 1 M HCI (1 M) and the combined experiments were extracted with EtOAc (3 x 3 L). The combined organic layers were washed with brine (2 x 2 L), dried over anhydrous Na 2 SO 4 , filtered, and the filtrate was concentrated under reduced pressure to give (2S)-2-[(fert-butoxycarbonyl)amino]-3- 15 [3-[3-(3-hydroxy-2,2-dimethylpropyl)-2-iodo-1 H-indol-5-yl]-5-[(triisopropylsilyl)oxy]phenyl]propanoic acid (402 g, crude) as a solid. LCMS (ESI): m/z [M+Na] calc'd for CseHsaINzOeSiNa 787.3; found 787.6.

Step 7. To a stirred mixture of (2S)-2-[(fert-butoxycarbonyl)amino]-3-[3-[3-(3-hydroxy-2,2- dimethylpropyl)-2-iodo-1 H-indol-5-yl]-5-[(triisopropylsilyl)oxy]phenyl]propanoic acid (340 g, 445 mmol) and methyl (3S)-1 ,2-diazinane-3-carboxylate (96.1 g, 667 mmol) in DCM (3.5 L) was added NMM (225 g, 2.2 20 mol), EDCI (170 g, 889 mmol), and HOST (12.0 g, 88.9 mmol) portionwise at 0 °C. The mixture was warmed to rt and stirred for 16 h, then washed with H 2 O (3 x 2.5 L), brine (2 x 1 L), dried over anhydrous NazS04 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by column chromatography to give methyl (3S)-1-[(2S)-2-[(te/t-butoxycarbonyl)amino]-3-[3-[3-(3-hydro xy- 2,2-dimethylpropyl)-2-iodo-1 H- indol-5-yl]-5-[(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3- 25 carboxylate (310 g, 62% yield) as an oil. LCMS (ESI): m/z [M+H] calc'd for C42H63IN4O7S1890.4; found 890.8.

Step 8. This reaction was undertaken on three batches in parallel on the scale illustrated below.

To a stirred mixture of methyl (3S)-1-[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[3-(3-hydro xy-2,2- dimethylpropyl)-2-iodo-1 H- indol-5-yl]-5-[(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3-

30 carboxylate (85.0 g, 95.4 mmol) in THF (850 mL) each added LiOH (6.85 g, 286 mmol) in H 2 O (410 mL) dropwise at 0 °C under an atmosphere of N 2 . The mixture was stirred at 0 °C for 1.5 h [note: LCMS showed 15% de-TIPS product], then acidified to pH 5 with 1 M HCI and the combined experiments extracted with EtOAc (3 x 2 L). The combined organic layers were washed with brine (2 x 1.5 L), dried over anhydrous NaaSO*, filtered, and the filtrate was concentrated under reduced pressure to give (3S)-1-

35 [(2S)-2-[(fert-butoxycarbonyl)amino]-3-[3-[3-(3-hydroxy-2,2- dimethylpropyl)-2-iodo-1 H-indol-5-yl]-5- [(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3-carboxylic acid (240 g, crude) as a solid. LCMS (ESI): m/z [M+H] calc’d for C41H61IN4O7S1 876.3; found 877.6.

Step 9.

This reaction was undertaken on two batches in parallel on the scale illustrated below.

40 To a stirred mixture of (3S)-1 -[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[3-(3-hydroxy-2,2 - dimethylpropyl)-2-iodo-1 H-indol-5-yl]-5-[(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3-carboxylic acid (120 g, 137 mmol) in DCM (6 L) was added DIPEA (265 g, 2.05 mol), EDCI (394 g, 2.05 mol), and

830

SUBSTITUTE SHEET (RULE 26) HOBT (37 g, 274 mmol) in portions at 0 °C under an atmosphere of Na. The mixture was warmed to rt and stirred overnight, then the combined experiments were washed with H 2 O (3 x 6 L), brine (2 x 6 L), dried over anhydrous NaaSO*, and filtered. After filtration, the filtrate was concentrated under reduced pressure. The filtrate was concentrated under reduced pressure and the residue was purified by column 5 chromatography to give fert-butyl N-[(8S,14S)-21 -iodo-18,18-dimethyl-9,15-dioxo-4-[(triisopropylsilyl)oxy]-

16-oxa-10,22,28-triazapentacyclo[18.5.2.1 A [2,6].1 A [10,14].0 A [23,27]]nonacosa- 1 (26),2,4,6(29),20,23(27),24-heptaen-8-yl]carbamate (140 g, 50% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C^htalN-tOeSi 858.9; found 858.3.

10 Intermediate 7. Synthesis of (S)-methyl 2-(( tert-butoxycarbonyl)amino)-3-(4-(4, 4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)pyrldin-2-yl)propanoate c 29 % yield

Step 1. Zn dust (28 g, 428 mmol) was added to a 1 L, three necked, round bottomed flask, purged with Na, and heated with a heat gun for 10 min under vacuum. The mixture was cooled to rt, and a 15 solution of 1 ,2-dibromoethane (1.85 mL, 21.5 mmol) in DMF (90 mL) was added dropwise over 10 min. The mixture was heated at 90 °C for 30 min and re-cooled to rt. TMSCI (0.55 mL, 4.3 mmol) was added, and the mixture was stirred for 30 min at rt, then a mixture of (fl)-methyl 2-((tert-butoxycarbonyl)amino)-3- iodopropanoate (22.5g, 71.4 mmol) in DMF (200 mL) was added dropwise over a period of 10 min. The mixture was heated at 35 °C and stirred for 2 h, then cooled to rt, and 2,4-dichloropyridine (16 g, 109 20 mmol) and Pd(PPh3)2Cl 2 (4 g, 5.7 mmol) added. The mixture was heated at 45 °C and stirred for 2 h, cooled, and filtered, then H 2 O (1 L) and EtOAc (0.5 L) were added to the filtrate. The organic and aqueous layers were separated, and the aqueous layer was extracted with EtOAc (2 x 500 mL). The organic layers were dried over anhydrous NaaSO*, filtered, and concentrated under reduced pressure. The crude residue was purified by silica gel column chromatography to give (S)-methyl 2 -((tert- 25 butoxycarbonyl)amino)-3-(4-chloropyridin-2-yl)propanoate (6.5 g, 29% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C14H19CIN 2 O4314.1 ; found 315.1.

Step 2. To a mixture of (S)-methyl 2-((terf-butoxycarbonyl)amino)-3-(4-chloropyridin-2- yl)propanoate (6.5 g, 20.6 mmol) in 1 ,4-dioxane (80 mL) at rt under an atmosphere of N 2 was added bis(pinacolato)diboron (6.3 g, 24.7 mmol), KOAc (8.1 g, 82.4 mmol), and Pd(PCya)2Cl 2 (1.9 g, 2.5 mmol). 30 The mixture was heated to 100 °C and stirred for 3 h, then H 2 O (100 mL) added and the mixture extracted with EtOAc (3 x 200 mL). The organic layers were combined, washed with brine (2 x 100 mL), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give (S)-methyl 2 -{(tert-

831

SUBSTITUTE SHEET (RULE 26) butoxycarbonyl)amino)-3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxab orolan-2-yl)pyridin-2-yl)propanoate (6 g, 72% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for CaoHsiBNaOe 406.2; found 407.3.

Synthesis of Intermediate 8.

Step 1. To a mixture of 4-(dimethylamino)but-2-ynoic acid (900 mg, 7.0 mmol) in DMF (20 mL) at -5 °C was added fert-butyl N-methyl-N-((S)-pyrrolidine-3-carbonyl)-L-valinate (1.0 g, 3.5 mmol), DIPEA (2.2 g, 17.6 mmol) and HATU (2.7 g, 7.0 mmol) in portions. The mixture was stirred between -5 to 5 °C for 1 h, then diluted with EtOAc (100 mL) and ice-H 2 O (100 mL). The aqueous and organic layers were

10 separated and the organic layer was washed with H 2 O (3 χ 100 mL), brine (100 mL), dried over anhydrous NaaSO*. and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give fert-butyl N-((S)-1 -(4-(dimethylamino)but-2- ynoyl)pyrrolidine-3-carbonyl)-N-methyl-L-valinate (900 mg, 55% yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for C21H35N3O4393.5; found 394.3.

15 Step 2. To a mixture of fert-butyl N-((S)-1-(4-(dimethylamino)but-2-ynoyl)pyrrolidine-3-carbony l)- W-methyl-L-valinate (260 mg, 0.66 mmol) in DCM (6 mL) was added TEA (3 mL) at rt. The mixture was stirred at rt for 2 h, then the solvent was concentrated under reduced pressure to give (2S)-2-{1 -[(3S)-1 - [4-(dimethylamino)but-2-ynoyl]pyrrolidin-3-yl]-N-methylforma mido}-3-methylbutanoic acid (280 mg) as an impure oil. The crude product was used directly in the next step without further purification. LCMS (ESI):

20 m/z [M+H] calc’d for C17H27N3O4337.2; found 338.3.

Synthesis of Intermediate 9.

9 ζ Ρ

Q

Step 1. To a mixture of fert-butyl N-methyl-N-((S)-pyrrolidine-3-carbonyl)-L-valinate (500 mg, 1.8

25 mmol) in DCM (8 mL) at 5 °C was added TEA (533 mg, 5.3 mmol) followed by dropwise addition of 2- chloroethane-1 -sulfonyl chloride (574 mg, 3.5 mmol) in DCM (2 mL) The mixture was stirred at 5 °C for 1 h, then diluted with H 2 O (20 mL) and extracted with EtOAC (3 x 10 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous NaaSO*, and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give tert-

30 butyl yV-methyl-AA-((S)-1-(vinylsulfonyl)pyrrolidine-3-carbonyl)-L -valinate (300 mg, 45% yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for CvHsoNaOsS 374.2; found 375.2.

Step 2. To a mixture of fert-butyl N-methyl-N-((S)-1-(vinylsulfonyl)pyrrolidine-3-carbonyl)-L- valinate (123 mg, 0.33 mmol) in DCM (3 mL) at rt was added TEA (1 mL). The mixture was stirred at rt for 1 h, then concentrated under reduced pressure to give N-methyl-N-((S)-1-(vinylsulfonyl)pyrrolidine-3-

832

SUBSTITUTE SHEET (RULE 26) carbonyl)-L-valine (130 mg, crude) as a solid, which was used directly in the next step without further purification. LCMS (ESI): m/z [M+H] calc’d for C13H22N 2 O5S 318.1 ; found 319.1 .

Synthesis of Intermediate 10.

5

Step 1. A mixture of 5-chloro-1 H-pyrrolo[3,2-b]pyridine-3-carbaldehyde (8.5 g, 47.1 mmol) and ethyl 2-(triphenylphosphoranylidene)propionate (2.56 g, 70.7 mmol) in 1 ,4-dioxane (120 mL) was stirred at reflux for 4 h, then concentrated under reduced pressure. EtOAc (200 mL) was added and the mixture was washed with brine, dried over NazSO*. and filtered. The filtrate was concentrated under reduced 10 pressure and the residue was purified by silica gel column chromatography to give ethyl (E)-3-(5-chloro- 1 /+pyrrolo[3,2-b]pyridin-3-yl)-2-methylacrylate (7.5 g, 60% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for C13H13CIN 2 O2 264.1 ; found 265.1 .

Step 2. To a mixture of ethyl (E)-3-(5-chloro-1 H- pyrrolo[3,2-b]pyridin-3-yl)-2-methylacrylate (7.5 g, 28.3 mmol) and N1CI2 (4.8 g, 28.3 mmol) in 1 :1 THF/MeOH (300 mL) was added NaBhU (21 .5 g, 566 15 mmol) in 20 portions every 25 minutes. After complete addition, the mixture was stirred at rt for 30 min, then diluted with EtOAc (500 mL) and washed with brine, dried over NaaSOA, and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give ethyl 3-(5-chloro-1 H- pyrrolo[3,2-b]pyridin-3-yl)-2-methylpropanoate (3.4 g, 45% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C13H15CIN 2 O2 266.1 ; found 267.1 .

20 Step 3. To a mixture of ethyl 3-(5-chloro-1 H-pyrrolo[3,2-b]pyridin-3-yl)-2-methylpropanoate (7.0 g, 26.2 mmol) and AgOTf (6.7 g, 26.2 mmol) in THE (50 mL) at 0 °C was added I2 (6.65 g, 26.2 mol).

The mixture was stirred at 0 °C for 30 min then diluted with EtOAc (100 mL), washed with NaaSOs (50 mL), brine (50 mL), dried over Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give ethyl 3-(5-chloro-2-iodo-1 H- 25 pyrrolo[3,2-b]pyridin-3-yl)-2-methylpropanoate (6 g, 58% yield) as white solid. LCMS (ESI): m/z [M+H] calc’d for C13H14CIIN 2 O2392.0; found 393.0.

Step 4. To a mixture of ethyl 3-(5-chloro-2-iodo-1 H- pyrrolo[3,2-b]pyridin-3-yl)-2- methylpropanoate (6.0 g, 15.3 mmol) and 2-(2-(2-methoxyethyl)phenyl)-4,4,5,5-tetramethyl-1 ,3,2- dioxaborolane (5.6 g, 21 .4 mmol) and K2CO3 (6.3 g, 45.9 mmol) in 1 ,4-dioxane (150 mL) and H 2 O (30 30 mL) under an atmosphere of N 2 was added Pd(dppf)Cl 2 .DCM (1 .3 g, 3.1 mmol). The mixture was heated to 80 °C and stirred for 4 h, then diluted with EtOAc (500 mL), washed with brine, dried over Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 3-(5-chloro-2-(2-(2-methoxyethyl)phenyl)-1 H- pyrrolo[3,2-b]pyridin-3-yl)-2-

833

SUBSTITUTE SHEET (RULE 26) methylpropanoate (5.5 g, 50% yield) as a viscous oil. LCMS (ESI): m/z [M+H] calc'd for C22H25CIN 2 O3 400.2; found 401.2.

Step 5. A mixture of ethyl 3-(5-chloro-2-(2-(2-methoxyethyl)phenyl)-1 H- pyrrolo[3,2-b]pyridin-3- yl)-2-methylpropanoate (5.5 g, 13.8 mmol), CS2CO3 (8.9 g, 27.5 mmol), and Etl (3.5 g, 27.5 mmol) in

5 DMF (30 mL) at rt was stirred for 10 h. The mixture was diluted with EtOAc (100 mL), washed with brine (20 mL x 4), dried over NaaSO*, filtered, and concentrated in vacuo. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give ethyl 3-(5- chloro-1 -ethyl-2-(2-(2-methoxyethyl)phenyl)-1 H-pyrrolo[3,2-b]pyridin-3-yl)-2-methylpropanoate (5.6 g, 95% yield) as a viscous oil. LCMS (ESI): m/z [M+H] calc’d for C25H31CIN 2 O3428.2; found 429.2.

10 Step 6. To a mixture of ethyl 3-(5-chloro-1-ethyl-2-(2-(2-methoxyethyl)phenyl)-1 H-pyrrolo[3,2- b]pyridin-3-yl)-2-methylpropanoate (5.4 g, 12.6 mmol) in THE (50 mL) at -65 °C was added 2M LDA (25 mL, 50 mmol) and stirred at -65 °C for 1 h. Mel (3.6 g, 25 mmol) was added and the mixture was stirred at -65 °C for 2.5 h, then aqueous NH4CI and EtOAc (50 mL) were added. The aqueous and organic layers were separated and the organic layer was washed with brine (30 mL), dried over NaaSO*, and filtered.

15 The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give ethyl 3-(5-chloro-1-ethyl-2-(2-(2-methoxyethyl)phenyl)-1 H-pyrrolo[3,2-b]pyridin-3- yl)-2,2-dimethylpropanoate (3.2 g, 57% yield) as a viscous oil. LCMS (ESI): m/z [M+H] calc'd for C25H31CIN 2 O3442.2; found 443.2.

Step 7. To a mixture of ethyl 3-(5-chloro-1-ethyl-2-(2-(2-methoxyethyl)phenyl)-1 H-pyrrolo[3,2-

20 b]pyridin-3-yl)-2,2-dimethylpropanoate (1.0 g, 2.3 mmol) in THE (10 mL) at 5 °C was added LiBH* (196 mg, 9.0 mmol). The mixture was heated to 65 °C and stirred for 2 h then aqueous NH4CI and EtOAc (50 mL) added. The aqueous and organic layers were separated and the organic layer was washed with brine (30 mL), dried over Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 3-(5-chloro-1-ethyl-2-(2-(2-

25 methoxyethyl)phenyl)-1 W-pyrrolo[3,2-b]pyridin-3-yl)-2,2-dimethylpropan-1-ol (0.75 g, 82% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C23H29CIN 2 O2400.2; found 401.2.

Intermediate 11: Methyl (3S)-1-{(2S)-2-(tert-butoxycait>onyl)amlno-3-[3-fluoro-5- (4,4,5,5- tetramethyH,3,2-dloxaborolan-2-yl)phenyl]propanoyl}-1,2-dlaz lnane-3-carboxylate

Br 1 F

°vC Hv°

Bzpinz 'NHBoc

Pd(dppf)Cl 2 . KOAc, Dioxane

'F

30

Step 1. To a stirred solution of methyl (2R)-2-{[(tert-butoxy)carbonyl]amino}-3- (iodozincio)propanoate (12 g, 30 mmol, 1.2 eq) in DMF (100 mL) was added 1 -bromo-3-fluoro-5-

834

SUBSTITUTE SHEET (RULE 26) iodobenzene (7.5 g, 25 mmol, 1 eq) and Pd(PPfi3)2Cl 2 (1.7 g, 2.5 mmol, 0.1 equiv) at 20°C under N 2 atmosphere. The resulting mixture was stirred for 2 hrs at 65°C under N 2 atmosphere. The reaction mixture was quenched with water and extracted with EA (200 mL x 2). The organic phase was washed with water (200 mL x 1) and brine (100 mL x 1) and concentrated to dryness to give a residue. The 5 residue was purified by prep-TLC (PE/EA=10/1 ) to afford methyl 3-(3-bromo-5-fluorophenyl)-2-{[(tert- butoxy)carbonyl]amino}propanoate (6 g, 58% yield) as a colorless oil. LCMS (ESI) m/z = 398.1 [M+Na] + , calculated for CisHi9BrFN04: 375.0

Step 2. To a solution of methyl 3-(3-bromo-5-fluorophenyl)-2-{[(tert- butoxy)carbonyl]amino}propanoate(3.2 g, 8.5 mmol, 1 eq) in THF (50 mL) was added Lithium 10 hydroxide(610.7 mg, 25.5 mmol, 3 eq) in H 2 O (10 mL). Then the reaction mixture was stirred at 20 °C for 1 h. The mixture was adjusted to pH = 5.0 with 1 M HCI aqueous solution. The mixture was quenched with H 2 O (150 mL) and extracted with EA (200 mL x 3). The combined organic layers was washed bine (50 mL), dried over Na 2 SO 4 and concentrated to afford 3-(3-bromo-5-fluorophenyl)-2-{[(tert- butoxy)carbonyl]amino}propanoic acid (2.65 g, 68% yield) as a white solid. LCMS (ESI) m/z = 384.1 15 [M+Na] + , calculated for CuHisBrFNCX MW: 361 .0

Step 3. To a mixture of 3-(3-bromo-5-fluorophenyl)-2-{[(tert-butoxy)carbonyl]amino}p ropanoic acid(2.3 g, 6.4 mmol, 1 eq) and methyl (3S)-1 ,2-diazinane-3-carboxylate(1.66 g, 11.5 mmol, 1.8 eq) in DMF(150 mL) was added HATU(4.9 g, 12.8 mmol, 2 eq) and DIEA(16.5 g, 128 mmol, 20 eq) in DMF(50 mL) at 0 °C. Then the reaction mixture was stirred at 0 °C for 1 h. The mixture was quenched with H 2 O 20 (100 mL) and extracted with EA (300 mL x 3). The combined organic layers was washed bine (50 mL), dried over Na 2 SO 4 and concentrated to give the residue, which was purified by Pre-HPLC eluting with acetonitrile in water (0.1 %FA) from 60% to 70% in 10 minutes to give methyl (3S)-1 -[(2S)-3-(3-bromo-5- fluorophenyl)-2-{[(tert-butoxy)carbonyl]amino}propanoyl]-1 ,2-diazinane-3-carboxylate(2.7 g, 78% yield) as a pale yellow solid. LCMS (ESI) m/z = 510.1 [M+Na]*, calculated for C2oH27BrFNOs: 487.1.

25 Step 4. A mixture of methyl (S)-1 -((S)-3-(3-bromo-5-fluorophenyl)-2-((tert- butoxycarbonyl)amino)propanoyl)hexahydropyridazine-3-carboxy late (3 g, 6.16 mmol, 1 eq), 4, 4,5,5- tetramethyl-2-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-1 ,3,2-dioxaborolane (1.9 g, 7.4 mmol, 1.2 eq), KOAc (900 mg, 9.24 mmol, 1.5 eq) and Pd(dppf)Cl 2 DCM (0.3 g, 0.37 mmol, 0.05 eq) in dioxane (50 mL) was heated at 100 °C for 17 h under N 2 atmosphere. The mixture was concentrated and purified by 30 column chromatography (DCM/MeOH=100/1 to 40/1 ) to give methyl (3S)-1 -(2S)-2-{(tert- butoxycarbonyl)amino-3-[3-fluoro-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl]propanoyl}-1 ,2- diazinane-3-carboxylate (2.6 g, 79% yield) as a yellow oil. LCMS (ESI) m/z = 536.2 [M+HJ + , calculated for C26H39BFNO7: 535.3.

Compounds A341 and A342 may be prepared using methods disclosed herein via Intermediate

35 11.

835

SUBSTITUTE SHEET (RULE 26) Example A75. Synthesis of two atropisomers of (2S)-W-[(8S,14S,20M)-22-ethyl-4-hydroxy- 21 -{2-[(1 S)-1 -methoxyethyl]pyrldln-3-yl}-18,18-dlmethyF9,15-dloxo-16-oxa- 10,22,28- trlazapentacyclo[18.5.2.1 2 ,*.1 10 , 14 .0 23 , 3T ]nonacosa-1(26),2,4,6(29),20,23(27),24-heptaen-8-yl]-3 - methyl-2-{A#-methyl-1-[(3S)-1-(prop-2-enoyl)pyrrolldln-3-yl] formamldo}butanamlde.

5

Step 1. To a stirred mixture of tertbutyl ((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3- yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (18.0 g, 20.1 mmol) in THF (180 mL) at 0 °C was added a 1 M solution of TBAF in THF (24.1 mL, 24.1 mmol). The mixture was stirred at 0 °C

10 for 1 h, then diluted with brine (1.5 L) and extracted with EtOAc (3 x 1 L). The combined organic layers were washed with brine (2 x 500 mL), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give tertbutyl ((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7- dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-

15 benzenacycloundecaphane-4-yl)carbamate (11.5 g, 69% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C42H53N5O7739.4; found 740.4.

Step 2. To a stirred mixture of fert-butyl ((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (11.5 g, 15.5 mmol) in DCM (120

20 mL) at 0 °C was added TFA (60 mL, 808 mmol). The mixture was stirred at 0 °C for 1 h, then concentrated under reduced pressure and the residue again concentrated under reduced pressure with toluene (20 mL; repeated x3) to give (6 3 S,4S)-4-amino-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-5,7-dione (12 g, crude), which was used directly in the next

25 step without further purification. LCMS (ESI): m/z [M+H] calc'd for C37H45N5O5639.3; found 640.6.

Step 3. To a stirred mixture of (6 3 S,4S)-4-amino-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-5,7-dione (11.9 g, 18.6 mmol) in DMF (240 mL) at 0 °C under an atmosphere of N 2 was added DIPEA (48.1 g, 372 mmol), (2S)-3-methyl-2-[N-methyl-1 -[(3S)-1 -

30 (prop-2-enoyl)pyrrolidin-3-yl]formamido]butanoic acid (9.45 g, 33.5 mmol) and COMU (11.95 g, 27.9 mmol) in portions. The mixture was stirred ay 0 °C for 90 min, then diluted with brine (1.5 L) and extracted with EtOAc (3 x 1 L). The combined organic layers were washed with brine (2 x 500 mL), dried over

836

SUBSTITUTE SHEET (RULE 26) anhydrous Na 2 SO 4 , and filtered. After filtration, the filtrate was concentrated under reduced pressure. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography (x 2) to give two atropisomers of (2S)-N-[(8S,14S,20M)-22-ethyl-4-hydroxy-21-{2-[(1 S)-1- methoxyethyl]pyridin-3-yl}-18,18-dimethyl-9,15-dioxo-16-oxa- 10,22,28- 5 triazapentacyclo[18.5.2.1 Z ,M ^.^.O^.^nonacosa-I (26),2,4,6(29),20,23(27),24-heptaen-8-yl]-3-methyl-2- {N-methyl-1-[(3S)-1-(prop-2-enoyl)pyrrolidin-3-yl]formamido} butanamide (2.7 g, 15.5%, yield) and (4.2 g, 24.7% yield) both as solids. LCMS (ESI): m/z [M+H] calc'd for CsiHesNyOe 903.5; found 904.7; 1 H NMR (400 MHz, DMSO-db) 69.35 - 9.27 (m, 1H), 8.77 (dd, J= 4.7, 1.7 Hz, 1 H), 7.95 (dq, J= 6.2, 2.0 Hz, 2H), 7.55 (ddd, J= 28.0, 8.2, 4.3 Hz, 3H), 7.08 (dd, J= 37.9, 6.2 Hz, 2H), 6.69 - 6.48 (m, 2H), 6.17 (ddt, J =

10 16.7, 7.2, 2.3 Hz, 1 H), 5.74 - 5.62 (m, 1 H), 5.43 - 5.34 (m, 1 H), 5.12 - 5.00 (m, 1 H), 4.25 (d, J = 12.3 Hz,

1 H), 4.17 - 3.99 (m, 3H), 3.89 - 3.65 (m, 4H), 3.66 - 3.45 (m, 3H), 3.12 (s, 4H), 2.95 - 2.70 (m, 6H), 2.41 - 2.06 (m, 5H), 1.99 - 1.88 (m, 1 H), 1.82 (d, J = 12.1 Hz, 2H), 1.54 (t, J = 12.0 Hz, 1 H), 1.21 (dd, J = 6.3,

2.5 Hz, 3H), 1.11 (t, J= 7.1 Hz, 3H), 0.99 - 0.88 (m, 6H), 0.79 (ddd, J= 27.8, 6.7, 2.1 Hz, 3H), 0.48 (d, J = 3.7 Hz, 3H) and LCMS (ESI): m/z [M+H] calc'd for CSIHMNTOS 903.5; found 904.7; 1 H NMR (400 MHz,

15 DMSO-db) 69.34 - 9.27 (m, 1H), 8.77 (dd, J= 4.7, 1.7 Hz, 1 H), 8.17 - 7.77 (m, 3H), 7.64 - 7.43 (m, 3H), 7.33 (d, J= 13.7 Hz, 1H), 7.05 - 6.94 (m, 1H), 6.69 - 6.41 (m, 2H), 6.26 - 5.94 (m, 1H), 5.73 - 5.63 (m,

1H), 5.50 - 5.20 (m, 2H), 4.40 - 4.15 (m, 3H), 4.00 - 3.40 (m, 9H), 3.11 (d, J= 4.4 Hz, 3H), 2.93 - 2.60 (m, 8H), 2.29 - 2.01 (m, 3H), 1.99 (s, 1 H), 1.87 - 1.75 (m, 2H), 1.73 - 1.47 (m, 2H), 1.40 (d, J = 6.0 Hz, 3H),

1.01 - 0.88 (m, 6H), 0.85 - 0.65 (m, 7H), 0.56 (s, 3H).

20

Example A89. Synthesis of (25)-Ν-[(85,145)-22-βΙΙιγΙ-4-Ιιγ<ΐΓθχγ-18, 18-<1ίιηβΙΙιγΙ-21-[6-(4- methylpiperazin-1 -yl)pyridin-3-yl]-9,15-dk>xo-16-oxa-10,22,28- trlazapentacyclo[18.5.2.1 2 ,M 10 , 14 .0”, 2 lnonacosa-1(26),2,4,6(29),20,23(27),24-heptaen-8-yq-3- methyl-2-{N-methyl-1-[(3S)-1-(prop-2-enoyl)pyrrolldln-3-yl]f onnamlclo}butanamlde

Step 1. To a mixture of tert-butyl ((6 3 S,4S)-1 2 -iodo-10,10-dimethyl-5,7-dioxo-2 5 - ((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate (240.00 mg, 0.279 mmol, 1.00 equiv) and CsaCOs (182 mg,

837

SUBSTITUTE SHEET (RULE 26) 0.558 mmol, 2 equiv) in DMF (5.00 mL) was added ethyl iodide (113.45 mg, 0.727 mmol, 2.60 equiv) dropwise at 0 °C. The reaction was stirred for 16 h at 25 °C. The resulting mixture was diluted with water (10 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (3 x 10 mL), and dried over anhydrous NazSO*. The filtrate was concentrated under reduced pressure and the 5 remaining residue was purified by silica gel column chromatography to afford tert-butyl ((6 3 S,4S)-1 1 -ethyl- 1 2 -iodo-10,10-dimethyl-S.T-dioxo^^iitriisopropylsilyOoxy l-e 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (190 mg, 77%yield) as a yellow solid.

Step 2. A mixture of fertbutyl ((6 3 S,4S)-1 ’-ethyl-1 2 -iodo-10,10-dimethyl-5,7-dioxo-2 s - 10 ((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 ® -hexahydro-1 ' H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate (500 mg, 0.54 mmol), 1-methyl-4-[5-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)pyridin-2-yl]piperazine (257 mg, 0.8 mmol), Pd(dppf)Cl 2 (83 mg, 0.11 mmol) and K2CO3 (156 mg, 1 .1 mmol) in 1 ,4-dioxane (25 mL) and HzO (5 mL) under an atmosphere of Ar was stirred at 80 °C for 2 h. The mixture was concentrated under reduced pressure and the residue was purified by 15 prep-TLC to afford fert-butyl ((6 3 S,4S)-1 1 -ethyl-10,10-dimethyl-1 2 -(6-(4-methylpiperazin-1 -yl)pyridin-3-yl)-

5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6’ ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (400 mg, 76% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for CssHTyNyOeSi 935.6; found 936.6.

Step 3. A mixture of fertbutyl ((6 3 S,4S)-1 1 -ethyl-10,10-dimethyl-1 2 -(6-(4-methylpiperazin-1 - 20 yl)pyridin-3-yl)-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-

6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (350 mg, 0.36 mmol) and 1M TBAF in THF (0.4 mL, 0.4 mmol) in THF (5 mL) was stirred at 0 °C for 1 h. The mixture was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give fertbutyl ((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-10,10-dimethyl-1 2 -(6-(4-methylpiperazin-1 -yl)pyridin-3-yl)-5,7-dioxo- 25 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-

4-yl)carbamate (290 mg, 100% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for C44H57N7O6779.4; found 780.4.

Step 4. A mixture of fertbutyl ((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-10,10-dimethyl-1 2 -(6-(4- methylpiperazin-1 -yl)pyridin-3-yl)-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 ' H- 8-oxa-1 (5,3)-indola-6(1 ,3)- 30 pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (300 mg, 0.37 mmol) in TFA (5 mL) and DCM (5 mL) was stirred at rt for 1 h. The mixture was concentrated under reduced pressure to give (6 3 S,4S)-4-amino-1 1 -ethyl-2 5 -hydroxy-10,10-dimethyl-1 2 -(6-(4-methylpiperazin-1 -yl)pyridin-3-yl)- 6',6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane- 5,7-dione (300 mg, crude) as a solid. LCMS (ESI): m/z [M+H] calc'd for C39H49N7O4679.4; found 680.3.

35 Step 5. To a mixture of (6 3 S,4S)-4-amino-1 1 -ethyl-2 5 -hydroxy-10,10-dimethyl-1 2 -(6-(4- methylpiperazin-1 -yl)pyridin-3-yl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 'H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-5,7-dione (300 mg, 0.36 mmol) in DMF (3 mL) at 0 °C under an atmosphere of Nz was added DIPEA (0.96 mL, 5.4 mmol) and (2S)-3-methyl-2-[N-methyl-1-[(3S)-1-(prop- 2-enoyl)pyrrolidin-3-yl]formamido]butanoic acid (213 mg, 0.72 mmol), followed by dropwise addition of 40 COMU (243 mg, 0.56 mmol). HzO was added at 0 °C and the mixture was extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the crude residue was purified by

838

SUBSTITUTE SHEET (RULE 26) Prep-HPLC to give (2S)-N-[(8S,14S)-22-ethyl-4-hydroxy-18,18-dimethyl-21 -[6-(4-methylpiperazin-1 - yl)pyridin-3-yl]-9,15-dioxo-16-oxa-10,22,28-triazapentacyclo [18.5.2.1 Z ,M lO , l4 .0 23 , 27 ]nonacosa- 1(26),2,4,6(29),20,23(27),24-heptaen-8-yl]-3-methyl-2-{N-met hyl-1-[(3S)-1-(prop-2-enoyl)pyrrolidin-3- yl]formamido}butanamide (45 mg, 13.2% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for CsaHmNeO?

5 943.5; found 944.8; Ή NMR (400 MHz, DMSO-db) δ 9.39 - 9.23 (m, 1H), 8.64 - 8.60 (m, 1H), 8.19 - 8.16

(m, 1 H), 8.15 (d, J = 6.2 Hz, 1 H), 7.86 (s, 1 H), 7.66 - 7.62 (m, 1 H), 7.56-7.54 (m, 1 H), 7.50 - 7.43 (m, 1 H), 7.13 - 7.11 (m, 1H), 7.03 - 6.95 (m, 1H), 6.70 - 6.47 (m, 2H), 6.17 (ddt, J= 16.8, 6.4, 2.8 Hz, 1H), 5.76 - 5.63 (m, 1 H), 5.45 - 5.33 (m, 1 H), 5.11 (m, 1 H), 4.75 - 4.72 (m, 1 H), 4.28 - 4.24 (m, 1 H), 4.11 - 3.98 (m, 4H), 3.91 - 3.76 (m, 1 H), 3.73 - 3.71 (m, 1 H), 3.59 - 3.56 (m, 7H), 3.51 - 3.40 (m, 2H), 3.08 - 2.94 (m, 1 H), 10 2.94 - 2.92 (m, 2H), 2.92 - 2.87 (m, 2H), 2.86 - 2.83 (m, 2H), 2.80 - 2.65 (m, 2H), 2.83 - 2.82 (m, 3H), 2.28

- 2.25 (m, 3H), 2.08 - 2.05 (m, 2H), 2.02 - 1.96 (m, 1 H), 1.87 - 1.78 (m, 1 H), 1.74 - 1.66 (m, 1 H), 1.56 - 1.48 (m, 1 H), 1.11 - 1.08 (m, 4H), 0.99 - 0.92 (m, 2H), 0.89 - 0.87 (m, 5H), 0.82 - 0.73 (m, 2H).

Example A115. Synthesis of two atropisomers of (2S)-N-[(8S,14S,20/Y22-ethyl-21 -{4-[(1 Sy

15 1-ιη β Ι1ιοχγ β ΙΙιγΙ]ργΓΐ<1Ιη-3-γΙ}-18,18-<1Ιπ' β ΙΙ'γΜ,15-<1Ιοχο-16-οχ β -10,22 ι 28- triazapentacyclo[18.5.2.1 2 , , .1 10 , 14 .0 23 , 27 ]nonacosa-1(26),2,4,6(29),20,23(27),24-heptaen-8-yl]-3 - methyl-2-{W-methyl-1-[(3S)-1-(prop-2-enoyl)pyrrolidin-3-yl]f ormamido}butanamide

20 Step 1. A 1 L round-bottom flask was charged with tert-butyl ((6 3 S,4S)-1 2 -iodo-10,10-dimethyl- 5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate (22.00 g, 32.042 mmol, 1.00 equiv), toluene (300.00 mL), Pd2(dba)3 (3.52 g, 3.845 mmol, 0.12 equiv), S-Phos (3.95 g, 9.613 mmol, 0.30 equiv), and KOAc (9.43 g, 96.127 mmol, 3.00 equiv) at room temperature. To the mixture was added 4,4,5,5-tetramethyM ,3,2-

25 dioxaborolane (26.66 g, 208.275 mmol, 6.50 equiv) dropwise with stirring at room temperature. The resulting solution was stirred for 3 h at 60 °C. The resulting mixture was filtered, and the filter cake was washed with EtOAc. The filtrate was concentrated under reduced pressure and the remaining residue

839

SUBSTITUTE SHEET (RULE 26) was purified by silica gel column chromatography to afford tert-butyl ((6 3 S,4S)-10,10-dimethyl-5,7-dioxo- 12-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (22 g, 90 %) as a light yellow solid. ESI-MS m/z = 687.3 [M+H] + ; Calculated MW: 686.4

5 Step 2. A mixture of tert-butyl ((6 3 S,4S)-10,10-dimethyl-5,7-dioxo-1 2 -(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)-6 , ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate (2.0 g, 2.8 mmol), 3-bromo-2-[(1 S)-1 -methoxyethyl]pyridine (0.60 g, 2.8 mmol), Pd(dppf)Cl 2 (0.39 g, 0.5 mmol), and K3PO4 (1.2 g, 6.0 mmol) in 1 ,4-dioxane (50 mL) and HzO (10 mL) under an atmosphere of Nz was heated to 70 °C and stirred for 2 h. The mixture was

10 diluted with HzO (50 mL) and extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (3 x 50 mL), dried over anhydrous NazSO*. and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give tert- butyl ((6 3 S,4S)-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate

15 (1.5 g, 74% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for C40H49N5O6695.4; found 696.5.

Step 3. A mixture of tert-butyl ((6 3 S,4S)-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl- 5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate (1.5 g, 2.1 mmol), CszCOs (2.1 g, 6.3 mmol), and ethyl iodide (0.43 mL, 5.1 mmol) in DMF (50 mL) was stirred at 0 °C for 16 h. The mixture was quenched at 0 °C with

20 HzO and extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (3 x 50 mL), dried over anhydrous NazSO*. and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give tert-butyl ((6 3 S,4S)-1 1 -ethyl-1 2 - (2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa- 1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (1.5 g, 99% yield) as a

25 solid. LCMS (ESI): m/z [M+H] calc’d for C42H53N5O6723.4; found 724.6.

Step 4. A mixture of tert-butyl ((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 FF8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate (1.3 g, 1.7 mmol) in TFA (10 mL) and DCM (20 mL) was stirred at 0 °C for 2 h. The mixture was concentrated under reduced pressure to afford (6 3 S,4S)-4-amino-

30 1 '-ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 ' H-8-oxa- 1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-5,7-dione (1.30 g, crude) as a solid. LCMS (ESI): m/z [M+H] calc’d for C37H45N5O4623.3; found 624.4.

Step 5. Into a 40-mL vial purged and maintained with an inert atmosphere of Ar, was placed (6 3 S,4S)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -

35 hexahydro-1 Ή-8-oxa-l (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-5,7-dione (250 mg, 0.4 mmol), (2S)-3-methyl-2-[N-methyl-1-[(3S)-1-(prop-2-enoyl)pyrrolidin -3-yl]formamido]butanoic acid (226 mg, 0.8 mmol), DIPEA (774 mg, 6.0 mmol), and DMF (3 mL). A solution of COMU (257 mg, 0.6 mmol) in DMF (2 mL) was added at 0 °C and the resulting mixture was stirred at 0 °C for 1 h. The mixture was filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by prep-

40 HPLC to give two atropisomers of (2S)-N-[(8S,14S,20P)-22-ethyl-21-{4-[(1 S)-1-methoxyethyl]pyridin-3-yl}- 18,18-dimethyl-9,15-dioxo-16-oxa-10,22,28-triazapentacyclo[1 8.5.2.1 Z ,M 10 , 14 .0 23 , 27 ]nonacosa- 1(26),2,4,6(29),20,23(27),24-heptaen-8-yl]-3-methyl-2-{N-met hyl-1-[(3S)-1-(prop-2-enoyl)pyrrolidin-3-

840

SUBSTITUTE SHEET (RULE 26) yl]formamido}butanamide (56 mg, 15% yield) and (46 mg, 13% yield) both as a solid. LCMS (ESI): rn/z [M+H] calc'd for CsiHesNyOy 887.5; found 888.4; 1 H NMR (400 MHz, DMSO-db) 68.81 (s, 1H), 8.07 (s,1H), 8.05 - 7.96(m, 1H), 7.78 - 7.45 (m, 5H), 7.41 - 7.08(m, 2H), 6.66 -6.58 (m, 1H), 6.18 (d, J= 17.0 Hz, 1 H), 5.75 - 5.67 (m, 1 H), 5.46 - 5.31 (m, 1 H), 5.16 - 5.04 (m, 1 H), 4.75 (dd, J = 10.9, 4.5 Hz, 1 H), 4.31

5 - 4.21 (m, 2H), 4.11 - 3.95 (m, 3H), 3.87 - 3.71 (m, 5H), 3.74 - 3.54 (m, 3H), 3.11 (s, 4H), 2.95 (d, J= 9.7 Hz, 2H), 2.85 - 2.72 (m, 3H), 2.31 - 2.04 (m, 3H), 1.88 - 1.47(m, 2H), 1.24 - 1.21 (m, 3H), 1.16 - 1.08 (m, 3H), 1.03 - 0.91 (m, 6H), 0.85 -0.74 (m, 3H), 0.51 - 0.46 (m, 3H) and LCMS (ESI): m/z [M+H] calc’d for C51H65N7O7887.5; found 888.4; Ή NMR (400 MHz, DMSO-db) δ 8.77 (s, 1H), 8.71 - 8.63 (m, 0.5H), 8.23

- 8.17 (m, 0.5H), 8.00 (s, 1H), 7.85 (t, J= 9.9 Hz, 2H), 7.77 - 7.62 (m, 3H), 7.57 - 7.50 (m, 1H), 7.33 - 7.22

10 (m, 1 H), 7.15 - 7.06 (m, 1 H), 6.73 - 6.56 (m, 1 H), 6.17 (ddd, J = 16.7, 6.1 , 2.7 Hz, 1 H), 5.76 - 5.64 (m,

1H), 5.49 - 5.29 (m, 2H), 4.70 (dd, J= 10.8, 3.5 Hz, 1H), 4.33 - 4.22 (m, 3H), 4.14 - 3.95 (m, 2H), 3.86 - 3.77 (m, 1 H), 3.72 - 3.65 (m, 2H), 3.61 (t, ,1 = 10.6 Hz, 3H), 3.46 - 3.42 (m, 1 H), 3.13 (d, J= 4.8 Hz, 3H), 2.99 (d, J= 14.4 Hz, 1H), 2.95 - 2.70 (m, 6H), 2.24 - 1.99 (m, 4H), 1.95 - 1.44 (m, 4H), 1.40 (d, J= 6.1 Hz, 3H), 0.98 - 0.87 (m, 6H), 0.86 - 0.64 (m, 6H), 0.64 - 0.54 (m, 3H).

15

841

SUBSTITUTE SHEET (RULE 26) Example A2. Synthesis of 4S)-4-amlno-22-ethyl-21 -[2-(2- methoxyethyl)phenyl]-18,18-dlmethyl-9,15-dk>xo-16-oxa-10, 22,28- triazapentacyclo[18.5.2.1 2 , t .1 10 , 14 .0 23 , 3T ]nonacosa-1(26),2,4,6(29),20,23(27),24-heptaen-8-yl]-3 - methyl-2-{N-methyl-1-[(3S)-1-(prop-2-enoyl)pyrrolldln-3-yllf ormamldo}butanamlde

5

Step 1. Into a 25 mL sealed tube were added 3-[1-ethyl-2-[2-(methoxymethyl)phenyl]-5-(4, 4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)indol-3-yl]-2,2-dimethylpropan-1-ol (590 mg, 1.2 mmol), methyl (2S)- 3-(3-bromo-5-nitrophenyl)-2-[(tert-butoxycarbonyl)amino]prop anoate (747 mg, 1.9 mmol), XPhos Pd G3 (105 mg, 0.12 mmol), XPhos (71 mg, 0.15 mmol), K2CO3 (427 mg, 3.1 mmol), and 1 ,4-dioxane (2 mL)

10 under an atmosphere of N 2 at rt. The mixture was heated to 60 °C and stirred overnight, then cooled and H 2 O added. The mixture was extracted with EtOAc (3 x 20 mL) and the combined organic layers were washed with brine (1 x 20 mL), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give (2S)-2- [(fert-butoxycarbonyl)amino]-3-[3-[1 -ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-[2-

15 (methoxymethyl)phenyl]indol-5-yl]-5-nitrophenyl]propanoic acid (500 mg, 61% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C37H45N3O8659.3; found 660.4.

Step 2. A mixture of (2S)-2-[(fert-butoxycarbonyl)amino]-3-[3-[1 -ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-[2-(methoxymethyl)phenyl]indol-5-yl]-5-nit rophenyl]propanoic acid (500 mg, 0.79 mmol), methyl (3S)-1 ,2-diazinane-3-carboxylate (164 mg, 1.1 mmol), DCM (6 mL), DIPEA (294 mg, 2.3

20 mmol) and HATU (432 mg, 1.1 mmol) was stirred at 0 °C for 1 h under an atmosphere of air. H 2 O was added and the mixture was extracted with DCM (3 x 20 mL), then the combined organic layers were dried

842

SUBSTITUTE SHEET (RULE 26) over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give methyl (3S)-1-[(2S)-2-[(tert- butoxycarbonyl)amino]-3-[3-[1-ethyl-3-(3-hydroxy-2,2-dimethy lpropyl)-2-[2-(methoxymethyl)phenyl]indol- 5-yl]-5-nitrophenyl]propanoyl]-1 ,2-diazinane-3-carboxylate (520 mg, 87% yield) as a solid. LCMS (ESI):

5 m/z [M+H] calc’d for C43H55N5O9785.4; found 786.8

Step 3. Into a 40 mL sealed tube were added methyl (3S)-1-[(2S)-2-[(ferf-butoxycarbonyl)amino]- 3-[3-[1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-[2-(methoxy methyl)phenyl]indol-5-yl]-5- nitrophenyl]propanoyl]-1 ,2-diazinane-3-carboxylate (510 mg, 0.65 mmol), DCE (5 mL) and trimethyltin hydroxide (587 mg, 3.3 mmol) at rt under an atmosphere of air. The mixture was heated to 60 °C and 10 stirred overnight, cooled, and diluted with DCM (20 mL). The mixture was washed with 0.1 N KHSO4 (3 x 20 mL), dried over anhydrous NaaSO*, filtered, and the filtrate was concentrated under reduced pressure to give (3S)-1 -[(2S)-2-[(te/1-butoxycarbonyl)amino]-3-[3-[1 -ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-[2- (methoxymethyl)phenyl]indol-5-yl]-5-nitrophenyl]propanoyl]-1 ,2-diazinane-3-carboxylic acid (500 mg, 100%) as a solid. LCMS (ESI): m/z [M+H] calc’d for C42H53N5O9771.4; found 772.7.

15 Step 4. A mixture of (3S)-1-[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[1-ethyl-3- (3-hydroxy-2,2- dimethylpropyl)-2-[2-(methoxymethyl)phenyl]indol-5-yl]-5-nit rophenyl]propanoyl]-1 ,2-diazinane-3- carboxylic acid (490 mg, 0.64 mmol), DCM (100 mL), DIPEA (2.5 g, 19.0 mmol), HOST (429 mg, 3.2 mmol), and EDCI (3.65 g, 19.0 mmol) at room temperature was stirred at rt overnight under an atmosphere of air. H 2 O was added and the mixture was extracted with DCM (3 x 60 mL), dried over 20 anhydrous NaaS04, and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give fert-butyl ((6 3 S,4S)-1 1 -ethyl-1 2 -(2- (methoxymethyl)phenyl)-10,10-dimethyl-2 5 -nitro-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (350 mg, 73% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C42H51N5O8753.4; found 754.2.

25 Step 5. A mixture of ferf-butyl ((6 3 S,4S)-1 1 -ethyl-1 2 -(2-(methoxymethyl)phenyl)-10,10-dimethyl- 2 5 -nitro-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 e -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate (200 mg, 0.27 mmol), MeOH (4 mL), and Pd on carbon (20 mg) was stirred at rt for 2 h under an atmosphere of Ha. The mixture was filtered, the filter cake was washed with MeOH (3 x 5 mL), and the filtrate was concentrated under reduced pressure to give fert-butyl 30 ((6 3 S,4S)-2 5 -amino-1 1 -ethyl-1 2 -(2-(methoxymethyl)phenyl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 ' H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (60 mg, 31% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C42H53N5O6723.4; found 724.4.

Step 6. Into an 8 mL vial were added fert-butyl ((6 3 S,4S)-2 5 -amino-1 1 -ethyl-1 2 -(2- (methoxymethyl)phenyl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 ' H- 8-oxa-1 (5,3)-indola- 35 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (50 mg, 0.07 mmol), DCM (1 mL), and TFA (158 mg, 1.4 mmol) at 0 °C under an atmosphere of air. The mixture was stirred for at 0 °C for 2 h then concentrated under reduced pressure to give (6 3 S,4S)-2 5 ,4-diamino-1 '-ethyl-1 2 -(2- (methoxymethyl)phenyl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 ' H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-5,7-dione (45 mg) as a solid, which was used directly in the

843

SUBSTITUTE SHEET (RULE 26) next step directly without further purification. LCMS (ESI): m/z [M+H] calc’d for CS/HASNSO* 623.3; found 624.4.

Step 7. Into an 8 mL vial were added (6 3 S,4S)-2 5 ,4-diamino-1 1 -ethyl-1 2 -(2- (methoxymethyl)phenyl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-

5 pyridazina-2(1 ,3)-benzenacycloundecaphane-5,7-dione (40 mg, 0.06 mmol), DMF (1 mL), DIPEA (75 mg, 0.58 mmol), and COMU (41 mg, 0.1 mmol) at 0 °C under an atmosphere of air. The mixture was stirred at 0 °C for 1 h, then H 2 O added. The mixture was extracted with EtOAc (3 x 30 mL), the combined organic layers were concentrated under reduced pressure, and purified by prep-HPLC to give (2S)-N-[(8S,14S)-4- amino-22-ethyl-21 -[2-(2-methoxyethyl)phenyl]-18,18-dimethyl-9,15-dioxo-16-oxa -10,22,28-

10 triazapentacyclo[18.5.2.1 Z ,M ^.^.O^.^nonacosa-I (26),2,4,6(29),20,23(27),24-heptaen-8-yl]-3-methyl-2- {N-methyl-1 -[(3S)-1 -(prop-2-enoyl)pyrrolidin-3-yl]formamido}butanamide (2.5 mg, 4.4% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for CSIHBSNTO? 887.5; found 888.6; 1 H NMR (400 MHz, DMSO-db) 68.74 - 8.55 (m, 1H), 7.89 (d, J= 9.6 Hz, 1H), 7.66 - 7.53 (m, 1H), 7.57 - 7.47 (m, 6H), 7.32 (t, J= 6.4 Hz, 1H), 6.85 (d, J= 8.4 Hz, 2H), 6.70 - 6.55 (m, 1H), 6.24 - 6.12 (m, 1H), 5.69 (ddd, J= 14.8, 8.0, 3.9 Hz, 1H),

15 5.41 (s, 1H), 5.09 - 4.80 (m, 2H), 4.26 (d, J = 10.1 Hz, 2H), 4.19 (s, 2H), 4.17 - 4.06 (m, 1H), 4.02 (dd, J = 12.0, 3.9 Hz, 1H), 3.92 (d, J= 8.0 Hz, 3H), 3.78 (d, J= 8.7 Hz, 5H), 3.29 (s, 2H), 3.14 (d, J= 1.9 Hz, 1H), 2.98 - 2.92 (m, 1 H), 2.87 - 2.68 (m, 3H), 2.62 (d, J = 12.5 Hz, 3H), 2.15 - 1.99 (m, 4H), 1.80 (s, 1 H), 1.68 - 1.53 (m, 2H), 1.08 (t, J= 7.1 Hz, 1H), 0.98 - 0.88 (m, 6H), 0.82 (dd, J= 23.3, 16.4 Hz, 3H), 0.74 (t, J = 7.2 Hz, 3H), 0.44 (s, 2H), 0.43 (s, 3H).

20

844

SUBSTITUTE SHEET (RULE 26) Example A118. Synthesis of (2S)-N-[(7S,13S)-21-ethyl·20-[2-(methoxymethyl)pyrldln-3-yl ]- 17,17-dlmethyl-8,14-dloxo-15-oxa-3-thia-9,21 ,27,28- tetraazapentacyclo[17.5.2.1 1 , 6 .1 • f 13 .0 Z2 ,*]octacosa-1 (25), 2(28), 4,19^2(26)^23-hexaen-7-yl]-3-methyl- 2-{N-methyl-1-[(3S)-1-(prop-2-enoyl)pyrrolidin-3-yl]formamid o}butanamide

Step 1. A mixture of methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-(1 ,3-thiazol-4-yl)propanoate (2.08 g, 7.26 mmol) and mCPBA (1.88 g, 10.9 mmol) in DCE (15 mL) at 0 °C under an atmosphere of Nz was diluted with DCM (100 mL). The mixture was allowed to warm to rt and stirred for 16 h, then diluted with DCM, washed with HzO (1 x 30 mL), dried over anhydrous NazSO*, and filtered. The filtrate was

10 concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 4-[(2S)-2-[(tert-butoxycarbonyl)amino]-3-methoxy-3-oxopropyl ]-1 ,3-thiazol-3-ium-3-olate (1.15 g, 47% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C12H18N 2 O5S 302.1 ; found 303.2.

Step 2. To a mixture of 4-[(2S)-2-[(tert-butoxycarbonyl)amino]-3-methoxy-3-oxopropyl ]-1 ,3- thiazol-3-ium-3-olate (1.15 g, 3.8 mmol) in THF at 0 °C under an atmosphere of N 2 was added NBS (0.74

15 g, 4.2 mmol) dropwise. The mixture was allowed to warm to rt and stirred for 2 h, then diluted with H 2 O (500mL) and extracted with EtOAc (3 x 500 mL). The combined organic layers were washed with water (2 x 30 mL), dried over anhydrous NazSO*. and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 2-bromo-4-[(2S)-2- [(te/t-butoxycarbonyl)amino]-3-methoxy-3-oxopropyl]-1 ,3-thiazol-3-ium-3-olate (1.2 g, 74% yield) as a

20 solid. LCMS (ESI): m/z [M+H] calc’d for CizHiyBrNzOsS 380.0; found 381 .0.

Step 3. To a stirred mixture of 2-bromo-4-[(2S)-2-[(tert-butoxycarbonyl)amino]-3-methoxy-3- oxopropyl]-1 ,3-thiazol-3-ium-3-olate (1.2 g, 3.2 mmol) and 4,4,5,5-tetramethyl-2-(tetramethyl-1 ,3,2- dioxaborolan-2-yl)-1 ,3,2-dioxaborolane (1.04 g, 4.1 mmol) in MeCN at 70 °C under an atmosphere of N 2 was added ethane-1 ,2-diamine (1.89 g, 31.5 mmol) in portions. The mixture was cooled to 60 °C and the

25 mixture was stirred overnight, then diluted with water (500 mL) and extracted with EtOAc (3 x 400 mL).

845

SUBSTITUTE SHEET (RULE 26) The combined organic layers were washed with brine (1 x 50 mL), dried over anhydrous NaaSO^ and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give methyl (2S)-3-(2-bromo-1 ,3-thiazol-4-yl)-2-[(tert- butoxycarbonyl)amino]propanoate (653 mg, 54% yield) as a solid.

5 Step 4. A 50 mL sealed tube was charged with 3-[1 -ethyl-2-[2-(methoxymethyl)pyridin-3-yl]-5- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)indol-3-yl]-2,2 -dimethylpropan-1-ol (1.00 g, 2.1 mmol), K2CO3 (727 mg, 5.2 mmol), Pd(dppf)Cl 2 (153 mg, 0.21 mmol), and 2,4-dibromo-1 ,3-thiazole (1.0 g, 4.2 mmol) at rt under an atmosphere of N 2 , then 1 ,4-dioxane (1.0 mL) and H 2 O (0.20 mL) were added. The mixture was heated to 70 °C and stirred for 4 h, then cooled, diluted with H 2 O (100 mL) and extracted with EtOAc

10 (3 x 100 mL). The combined organic layers were washed with brine (3 x 100 mL), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 3-[5-(4-bromo-1 ,3-thiazol-2-yl)-1 -ethyl-2-[2- (methoxymethyl)pyridin-3-yl]indol-3-yl]-2,2-dimethylpropan-1 -ol (727 mg, 67% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C34H44N4O6S 636.3; found 637.3.

15 Step 5. To a stirred mixture of methyl (2S)-2-[(fert-butoxycarbonyl)amino]-3-[2-[1 -ethyl-3-(3- hydroxy-2,2-dimethylpropyl)-2-[2-(methoxymethyl)pyridin-3-yl ]indol-5-yl]-1 ,3-th iazol-4-yl]propanoate (636 mg, 1.0 mmol) and UOH.H 2 O (126 mg, 3.0 mmol) in THE at 0 °C under an atmosphere of N 2 was added H 2 O (1.24 mL) portionwise. The mixture was allowed to warm to rt and stirred for 1 h, then diluted with water (300 mL) and extracted with EtOAc (3 x 300 mL). The combined organic layers were washed with

20 brine (1 x 100 mL), dried over anhydrous Na 2 SO 4 , filtered, and the filtrate concentrated under reduced pressure to give (2S)-2-[(tert-butoxycarbonyl)amino]-3-[2-[1 -ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-[2- (methoxymethyl)pyridin-3-yl]indol-5-yl]-1 ,3-th iazol-4-y IJpropanoic acid (622 mg, crude), which was used in the next step directly without further purification. LCMS (ESI): m/z [M+H] calc'd for C33H42N4O6S 622.3; found 623.2.

25 Step 6. To a stirred mixture of (2S)-2-[(fert-butoxycarbonyl)amino]-3-[2-[1-ethyl-3-(3-hydro xy-2,2- dimethylpropyl)-2-[2-(methoxymethyl)pyridin-3-yl]indol-5-yl] -1 ,3-thiazol-4-yl]propanoic acid (622 mg, 1.0 mmol) and methyl (3S)-1 ,2-diazinane-3-carboxylate (288 mg, 2.0 mmol) in DMF at 0 °C under an atmosphere of N 2 was added HATU (570 mg, 1.5 mmol). The mixture was stirred at 0 °C for 1 h, then diluted with EtOAc and washed with H 2 O (1 x 10 mL), dried over anhydrous NaaSO*, filtered, and the 30 filtrate concentrated under reduced pressure to give methyl (3S)-1 -[(2S)-2-[(tert-butoxycarbonyl)amino]-3- [2-[1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-[2-(methoxyme thyl)pyridin-3-yl]indol-5-yl]-1,3-thiazol-4- yl]propanoyl]-1 ,2-diazinane-3-carboxylate (550 mg, 62% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for CseHszNeOS 748.4; found 749.6.

Step 7. (3S)-1 -[(2S)-2-[(fert-butoxycarbonyl)amino]-3-[2-[1 -ethyl-3-(3-hydroxy-2,2- 35 dimethylpropyl)-2-[2-(methoxymethyl)pyridin-3-yl]indol-5-yl] -1,3-thiazol-4-yl]propanoyl]-1,2-diazinane-3- carboxylic acid was synthesized in a manner similar to (3S)-1-[(2S)-2-[(fert-butoxycarbonyl)amino]-3-[3- [3-(3-hydroxy-2,2-dimethylpropyl)-2-iodo-1H-indol-5-yl]-5-[( triisopropylsilyl)oxy]phenyl]propanoyl]-1,2- diazinane-3-carboxylic acid except methyl (3S)-1-[(2S)-2-[(fert-butoxycarbonyl)amino]-3-[3-[3-(3-hydro xy- 2,2-dimethylpropyl)-2-iodo-1 H- indol-5-yl]-5-[(triisopropylsilyl)oxy]phenyl]propanoyl]-1,2- diazinane-3- 40 carboxylate was substituted with methyl (3S)-1 -[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[2-[1 -ethyl-3-(3-

846

SUBSTITUTE SHEET (RULE 26) hydroxy-2,2-dimethylpropyl)-2-[2-(methoxymethyl)pyridin-3-yl ]indol-5-yl]-1 ,3-thiazol-4-yl]propanoyl]-1 ,2- diazinane-3-carboxylate. LCMS (ESI): m/z [M+H] calc'd for CaeHsoNeOyS 734.3; found 735.3.

Step 8. fert-butyl ((6 3 S,4S,2)-1 1 -ethyl-1 2 -(2-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7- dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-2(2,4)-thiazola-1 (5,3)-indola-6(1 ,3)- 5 pyridazinacycloundecaphane-4-yl)carbamate was synthesized in a manner similar to fert-butyl ((6 3 S,4S)- 1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane- 4-yl)carbamate except (3S)-1 -[(2S)-2-[(fert-butoxycarbonyl)amino]-3-[3-[1 -ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]indol-5-yl]-5- 10 [(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3-carboxylic acid was substituted with (3S)-1 -[(2S)-

2-[(fert-butoxycarbonyl)amino]-3-[2-[1-ethyl-3-(3-hydroxy -2,2-dimethylpropyl)-2-[2- (methoxymethyl)pyridin-3-yl]indol-5-yl]-1 ,3-thiazol-4-yl]propanoyl]-1 ,2-diazinane-3-carboxylic acid. LCMS (ESI): m/z [M+H] calc’d for CaeHwNeOeS 716.3; found 717.4.

Step 9. To a stirred mixture of fert-butyl ((6 3 S,4S,2)-1 1 -ethyl-1 2 -(2-(methoxymethyl)pyridin-3-yl)- 15 10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-2(2,4)-thiazola-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-4-yl)carbamate (253 mg) in DCM at 0 °C under an atmosphere of Na was added TEA (1.0 mL) dropwise. The mixture was stirred at 0 °C for 1 h, then concentrated under reduced pressure and then repeated using toluene (20 mL x 3) to give (6 3 S,4S,2)-4-amino-1 1 -ethyl-1 2 -(2- (methoxymethyl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-2(2,4)-thiazola-1 (5,3)- 20 indola-6(1 ,3)-pyridazinacycloundecaphane-5,7-dione (253 mg, crude) as a solid. LCMS (ESI): m/z [M+H] calc’d for C33H40N6O4S 616.3; found 617.3.

Step 10. (2S)-N-[(7S,13S)-21 -ethyl-20-[2-(methoxymethyl)pyridin-3-yl]-17,17-dimethyl-8,1 4- dioxo-15-oxa-3-thia-9,21 ,27,28-tetraazapentacyclo[17.5.2.1 z , e .1 e , 13 .0 22 , 2e ]octacosa- 1 (25),2(28),4,19,22(26),23-hexaen-7-yl]-3-methyl-2-{N-methyl- 1 -[(3S)-1 -(prop-2-enoyl)pyrrolidin-3- 25 yl]formamido}butanamide was synthesized in a manner similar to (2S)-N-[(8S,14S)-4-amino-22-ethyl-21 - [2-(2-methoxyethyl)phenyl]-18,18-dimethyl-9,15-dioxo-16-oxa- 10,22,28- triazapentacyclo[18.5.2.1 2 ,M «" O^nonacosa-I (26),2,4,6(29),20,23(27),24-heptaen-8-yl]-3-methyl-2- [N-methyl-1 -[(3S)-1 -(prop-2-enoyl)pyrrolidin-3-yl]formamido}butanamide except (6 3 S,4S)-4-amino-1 1 - ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8- 30 oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-5,7-dione was substituted with (6 3 S,4S,Z)-4-amino-1 1 -ethyl-1 2 -(2-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-6' ,β^,β^,β 6 - hexahydro-1 ' H- 8-oxa-2(2,4)-thiazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-5,7-dione. LCMS (ESI): m/z [M+H] calc’d for C47H60NBOS 880.4; found 881.6; 1 H NMR (400 MHz, DMSO-flfe) 68.75 (m, 1H), 8.55 (d, J= 6.7 Hz, 1H), 8.32 (d, J= 8.3 Hz, 1H), 7.99 (d, J= 7.7 Hz, 1H), 7.65 - 7.51 (m, 3H), 7.11 - 35 6.92 (m, 1H), 6.72 - 6.56 (m, 1H), 6.18 (dd, J= 16.8, 2.9 Hz, 1H), 5.82 - 5.65 (m, 1H), 5.61 - 5.46 (m, 1H),

5.02 (dd, J= 24.2, 12.2 Hz, 1H), 4.69 (d, J = 10.9 Hz, 1 H), 4.37 - 4.11 (m, 5H), 4.05 - 3.79 (m, 4H), 3.76 - 3.50 (m, 6H), 3.47 (s, 2H), 3.08 (s, 3H), 3.04 (s, 1H), 2.98 (d, J= 1.9 Hz, 1H), 2.95 (d, J= 3.6 Hz, 2H),

847

SUBSTITUTE SHEET (RULE 26) 2.83 (d, J = 2.0 Hz, 2H), 2.24 - 2.03 (m, 4H), 1.81 (s, 2H), 1.56 (s, 1 H), 1.11 (t , J= 7.0 Hz, 2H), 1.02 - 0.87 (m, 8H), 0.80 (dd, J= 24.6, 6.6 Hz, 3H), 0.41 (s, 2H), 0.31 (s, 1H).

Example A194. Synthesis of (25)-Ν-[(78,135)-21-βΙΙιγΙ-20-[2-(πιβΙΙιοχγ ηθΙΙιγΙ)ργΓΐάΙη-3-γΙ]- 5 17,17-dimethyl-8,14-dioxo-15-oxa-4-thia-9,21 ,27,28- tetraazapentacyclo[17.5.2.1 2 , 6 .1 •, 13 .0 22 , 2 *]octacosa-1 (25), 2,5(28), 19^2(26)^3-hexaen-7-yl]-3-methyl- 2-{A/-methyl-1 -[(3S)-1 -(prop-2-enoyl)pyrrolidin-3-yl]formamido}butanamide

Step 1. A mixture of Zn (1.2 g, 182 mmol) and 1 ,2-dibromoethane (1.71 g, 9.1 mmol) and DMF

10 (50 mL) was stirred for 30 min at 90 °C under an atmosphere of Ar. The mixture was allowed to rt, then TMSCI (198 mg, 1.8 mmol) was added dropwise over 30 min at rt. Methyl (2fl)-2-[(tert-butoxycarbonyl) amino]-3-iodopropanoate (10.0 g, 30.4 mmol) in DMF (100 mL) was added dropwise over 10 min at rt. The mixture was heated to 35 °C and stirred for 2 h, then a mixture of 2,5-dibromo-1 ,3-thiazole (1.48 g, 60.8 mmol) and Pd(PPha)2Cl 2 (2.1 g, 3.0 mmol) in DMF (100 mL) was added dropwise .The mixture was

15 heated to 70 °C and stirred for 2h, then filtered and the filtrate diluted with EtOAc (1 L) and washed with H 2 O (3 x 1 L), dried with anhydrous NaaSO*, and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give methyl (2S)-3-(5- bromo-1 ,3-thiazol-2-yl)-2-[(tert-butoxycarbonyl)amino]propanoate (3 g, 27 % yield) as a semi-solid. LCMS (ESI): m/z [M+H] calc'd for CiaHvBrNaOtS 364.0; found 365.1.

20 Step 2. Into a 20mL sealed tube were added 3-[1 -ethyl-2-[2-(methoxymethyl)pyridin-3-yl]-5- (4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)indol-3-yl]-2,2-dimethylpropan-1-ol (100 mg, 0.21 mmol), K3PO4 (111 mg, 0.52 mmol), Pd(dppf)Cl 2 (15 mg, 0.02 mmol), methyl (2S)-3-(4-bromo-1 ,3-thiazol-2-yl)-2- [(fert-butoxycarbonyl)amino]propanoate (153 mg, 0.42 mmol), toluene (1 mL), and H 2 O (0.2 mL) at rt under an atmosphere of N 2 . The mixture was heated to 60 °C and stirred for 3 h, cooled, diluted with H 2 O

25 (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give methyl (2S)-2-[(tert- butoxycarbonyl)amino]-3-[4-[1-ethyl-3-(3-hydroxy-2,2-dimethy lpropyl)-2-[2-(methoxymethyl)pyridin-3-

848

SUBSTITUTE SHEET (RULE 26) yl]indol-5-yl]-1 ,3-thiazol-2-yl]propanoate (72 mg, 54% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for C34H44N4O6S 636.3; found 637.2.

Step 3. A mixture of methyl (2S)-2-[(terf-butoxycarbonyl)amino]-3-[4-[1 -ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-[2-(methoxymethyl)pyridin-3-yl]indol-5-yl] -1 ,3-thiazol-2-yl]propanoate (40 mg, 0.06 5 mmol) and UOH.H 2 O (unspecified) in THE (1 mL) and H 2 O (0.2 mL) was stirred at rt under an atmosphere of N 2 for 2 h. The mixture was acidified to pH 5 with aqueous NaHSC>4 and extracted with EtOAc (3 x 10mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure to give (2S)-2-((fert- butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3-hydroxy-2,2-dimethy lpropyl)-2-(2-(methoxymethyl)pyridin-3-yl)- 10 1 H- indol-5-yl)thiazol-2-yl)propanoic acid. The crude product was used in the next step directly without further purification. LCMS (ESI): m/z [M+H] calc’d for C33H42N4O6S 622.3; found 623.3.

Step 4. Methyl (3S)-1-((2S)-2-((fert-butoxycarbonyl)amino)-3-(4-(1-ethyl-3- (3-hydroxy-2,2- dimethylpropyl)-2-(2-(methoxymethyl)pyridin-3-yl)-1 H- indol-5-yl)thiazol-2- yl)propanoyl)hexahydropyridazine-3-carboxylate was synthesized in a manner similar to methyl (3S)-1- 15 [(2S)-2-[(fe/1-butoxycarbonyl)amino]-3-[2-[1 -ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-[2-

(methoxymethyl)pyridin-3-yl]indol-5-yl]-1 ,3-thiazol-4-yl]propanoyl]-1 ,2-diazinane-3-carboxylate except (2S)-2-[(tert-butoxycarbonyl)amino]-3-[2-[1-ethyl-3-(3-hydro xy-2,2-dimethylpropyl)-2-[2- (methoxymethyl)pyridin-3-yl]indol-5-yl]-1 ,3-thiazol-4-yl]propanoic acid was substituted with (2S)-2-((tert- butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3-hydroxy-2,2-dimethy lpropyl)-2-(2-(methoxymethyl)pyridin-3-yl)- 20 1 H-indol-5-yl)thiazol-2-yl)propanoic acid. LCMS (ESI): m/z [M+H] calc'd for C39H52N6O7S 748.4; found

749.4.

Step 5. (3S)-1 -[(2S)-2-[(fert-butoxycarbonyl)amino]-3-[4-[1 -ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-[2-(methoxymethyl)pyridin-3-yl]indol-5-yl] -1 ,3-thiazol-2-yl]propanoyl]-1 ,2-diazinane-3- carboxylic acid was synthesized in a manner similar to (3S)-1-[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[3- 25 [3-(3-hydroxy-2,2-dimethylpropyl)-2-iodo-1 H-indol-5-yl]-5-[(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2- diazinane-3-carboxylic acid except methyl (3S)-1-[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[3-(3-hydro xy- 2,2-dimethylpropyl)-2-iodo-1 H- indol-5-yl]-5-[(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3- carboxylate was substituted with methyl (3S)-1-((2S)-2-((terf-butoxycarbonyl)amino)-3-(4-(1-ethyl-3- (3- hydroxy-2,2-dimethylpropyl)-2-(2-(methoxymethyl)pyridin-3-yl )-1 H-indol-5-yl)thiazol-2- 30 yl)propanoyl)hexahydropyridazine-3-carboxylate. LCMS (ESI): m/z [M+H] calc’d for CaeHsoNeOyS 734.3; found 735.4.

Step 6. 7ert-butyl ((6 3 S,4S,2)-1 1 -ethyl-12-(2-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7 - dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 8 -hexahydro-1 , H-8-oxa-2(4,2)-thiazola-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-4-yl)carbamate was synthesized in a manner similar to tert-butyl ((6 3 S,4S)-

35 1 '-ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)- 6',6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane- 4-yl)carbamate except (3S)-1 -[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[1 -ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]indol-5-yl]-5-

[(triisopropylsilyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3-carboxylic acid was substituted with (3S)-1-[(2S)-

40 2-[(tert-butoxycarbonyl)amino]-3-[4-[1-ethyl-3-(3-hydroxy-2, 2-dimethylpropyl)-2-[2-

849

SUBSTITUTE SHEET (RULE 26) (methoxymethyl)pyridin-3-yl]indol-5-yl]-1 ,3-thiazol-2-yl]propanoyl]-1 ,2-diazinane-3-carboxylic acid. LCMS (ESI): m/z [M+H] calc’d for CaeHteNeOeS 716.3; found 717.3.

Step 7. (6 3 S,4S,2)-4-amino-1 1 -ethyl-1 2 -(2-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-2(4,2)-thiazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane- 5 5,7-dione was synthesized in a manner similar to (6 3 S,4S,2)-4-amino-1 ’-ethyl-1 2 -(2-

(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-2(2,4)-thiazola-1 (5,3)- indola-6(1 ,3)-pyridazinacycloundecaphane-5,7-dione except tert-butyl ((6 3 S,4S,2)-1’-ethyl-1 2 -(2- (methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-2(2,4)- thiazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)carbamate was substituted with fert-butyl 10 ((6 3 S,4S,Z)-1 1 -ethyl-12-(2-(methoxymethyl)pyridin-3-yl)-10,10-dimeth yl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 S ,6 6 - hexahydro-1 1 H-8-oxa-2(4,2)-thiazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)carbamate. LCMS (ESI): m/z [M+Na] calc'd for C33H4oNe04SNa 639.3; found 640.3.

Step 8. (2S)-N-[(7S, 13S)-21 -ethyl-20-[2-(methoxymethyl)pyridin-3-yl]-17, 17-dimethyl-8, 14-dioxo- 15-oxa-4-thia-9,21 ,27,28-tetraazapentacyclo[17.5.2.1 2 , e .1 e , 13 .0 2Z , 2e loctacosa-1(25),2,5(28),19,22(26),23- 15 hexaen-7-yl]-3-methyl-2-{N- methyl-1 -[(3S)-1 -(prop-2-enoyl)pyrrolidin-3-yl]formamido}butanamide was synthesized in a manner similar to (2S)-N-[(7S,13S)-21-ethyl-20-[2-(methoxymethyl)pyridin-3-yl] -17,17- dimethyl-8,14-dioxo-15-oxa-3-thia-9,21 ,27,28-tetraazapentacyclo[17.5.2.1 2 , e .1 e , 13 .0 22 , 26 ]octacosa- 1 (25),2(28),4,19,22(26),23-hexaen-7-yl]-3-methyl-2-{N-methyl- 1 -[(3S)-1 -(prop-2-enoyl)pyrrolidin-3- yl]formamido}butanamide except (6 3 S,4S)-4-amino-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-((S)-1 -methoxyethyl)pyridin- 20 3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione was substituted with (6 3 S,4S,Z)-4-amino-1 1 -ethyl-1 2 -(2- (methoxymethyl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-2(4,2)-thiazola-1 (5,3)- indola-6(1 ,3)-pyridazinacycloundecaphane-5,7-dione. LCMS (ESI): m/z [M+H] calc’d for C47H60N8O7S 880.4; found 881.5; Ή NMR (400 MHz, DMSO-dfe) δ 8.70 (dt, J= 16.2, 8.1 Hz, 1H), 8.54 (ddd, J= 6.6,

25 4.7, 1.7 Hz, 1 H), 8.50 (m, 1 H), 7.96 (d, J = 7.8 Hz, 1 H), 7.88 (t, J = 2.1 Hz, 2H), 6.70 - 6.57 (m, 2H), 6.24 -

6.13 (m, 2H), 5.75 (m, 1H), 5.55 (t, J= 7.3 Hz, 1H), 5.46 (d, J= 8.5 Hz, 1H), 5.14 (d, J= 13.0 Hz, 1H), 4.84 - 4.75 (m, 1H), 4.35 (d, J= 10.7 Hz, 1H), 4.28 - 4.19 (m, 4H), 3.91 (s, 3H), 3.87 (dd, J= 10.4, 8.1 Hz, 1 H), 3.78 - 3.70 (m, 2H), 3.63 (t, J = 8.8 Hz, 2H), 3.61 - 3.49 (m, 2H), 2.87 (d, J = 1.1 Hz, 2H), 2.79 (s,

1 H), 2.38 (s, 1 H), 2.18 (s, 1 H), 2.13 (d, J = 10.7 Hz, 4H), 1.96 (s, 2H), 1 .81 (s, 1 H), 1 .53 (s,2H), 1.11 (t, J 30 = 7.1 Hz, 2H), 0.99 - 0.89 (m, 7H), 0.93 - 0.81 (m, 2H), 0.78 (d, J= 6.6 Hz, 2H), 0.28 (s, 3H).

850

SUBSTITUTE SHEET (RULE 26) Example A71. Synthesis of (2S)-2-(1-{1-[(2E)-4-(dlmethylamlno)but-2-enoyl]azetldln-3-y l}-/V· methylformamido)-W-[(8S,14S)-22-ethyl-4-hydroxy-21-[2-(metho xymethyl)pyridin-3-yl]-18,18- dlmethyl-9,15-dloxo-16-oxa-10,22,28-trlazapentacyclo[18.5.2. 1 2 ,*.1 10 , 14 .0 23 , 27 ]nonacosa- 5 1 (26)^,4,6(29),20, 23(27), 24-heptaen-8-yl]-3-methylbutanamide

Step 1. To a mixture of tert-butyl AF(azetidine-3-carbonyl)-N-methyl-L-valinate (350 mg, 1.3 mmol) and (2E)-4-(dimethylamino)but-2-enoic acid (201 mg, 1.56 mmol) in DCM (8 mL) at 5 °C was added a solution of T3P, 50% in EtOAc (827 mg, 2.6 mmol) and DIPEA (1.7 g, 13 mmol) in DCM (2 mL).

10 The mixture was stirred for 1 h, then diluted with EtOAc (20 mL) and H 2 O (20 mL). The aqueous and organic layers were separated and the organic layer was washed with H 2 O (3 x 10 mL), brine (10 mL), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by prep-HPLC to give tert-butyl (£)-N-(1-(4-(dimethylamino)but-2-enoyl)azetidine-3- carbonyl)-N-methyl-L-valinate (200 mg, 39% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for

15 C20H35N3O4381 .3; found 382.3.

Step 2. To a mixture of tert-butyl (E)-N-(1-(4-(dimethylamino)but-2-enoyl)azetidine-3-carbonyl) - AA-methyl-L-valinate (190 mg, 0.32 mmol) in DCM (3 mL) at rt was added TFA (1 mL). The mixture was stirred at rt for 1 h, then concentrated under reduced pressure to give (E)-N-(1 -(4-(dimethylamino)but-2- enoyl)azetidine-3-carbonyl)-N-methyl-L-valine (190 mg, 90%) as a solid, which was used directly in the

20 next step without further purification. LCMS (ESI): m/z [M+H] calc’d for C16H27N3O4325.2; found 326.2.

Step 3. To a mixture of (6 3 S,4S)-4-amino-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-(methoxymethyl)pyridin-3-yl)- 10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 ® -hexahydro-1 ' H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione (172 mg, 0.27 mmol) and (E)-N-(1 -(4-(dimethylamino)but-2- enoyl)azetidine-3-carbonyl)-N-methyl-L-valine (105 mg, 0.32 mmol) in DMF (2 mL) at 5 °C was added a

25 mixture of HATU (133 mg, 0.297 mmol) and DIPEA (348 mg, 2.7 mmol) in DMF (1 mL). The mixture was stirred for 1 h, then diluted with EtOAc (20 mL) and H 2 O (20 mL). The aqueous and organic layers were separated, and the organic layer was washed with H 2 O (3 x 10 mL), brine (10 mL), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by prep-TLC to give (2S)-2-(1-{1-[(2E)-4-(dimethylamino)but-2-enoyl]azetidin-3-y l}-N-methylformamido)-

30 AF[(8S,14S)-22-ethyl-4-hydroxy-21 -[2-(methoxymethyl)py ridi n-3-y I]- 18,18-dimethyl-9,15-dioxo-16-oxa- 10,22,28-triazapentacyclo[18.5.2.1 2 ,".1 10 , 14 .0 23 , 2 nnonacosa-1 (26),2,4,6(29),20,23(27),24-heptaen-8-yl]-3- methylbutanamide (4.8 mg, 2% yield over 2 steps) as a solid. LCMS (ESI): m/z [M+H] calc’d for CsaHeeNeOe 932.5; found 933.5; Ή NMR (400 MHz, CD3OD) δ 8.71 (d, J= 3.2 Hz, 1H), 8.50 (s, 1.5H), 8.08 - 7.85 (m, 2H), 7.65 - 7.44 (m, 3H), 7.32 - 7.14 (m, 1H), 7.07 - 6.95 (m, 1H), 6.80 (dt, J= 22.1 , 6.8

851

SUBSTITUTE SHEET (RULE 26) Hz, 1H), 6.55 (d, J= 35.8 Hz, 1H), 6.30 (d, J= 15.4 Hz, 1H), 5.56 (dd, J= 13.8, 6.7 Hz, 1H), 4.76 (dd, J = 19.8, 10.5 Hz, 1 H), 4.54 (dd, J= 15.9, 7.5 Hz, 2H), 4.48 - 4.38 (m, 2H), 4.36 - 4.23 (m, 3H), 4.22 - 4.14 (m, 1H), 3.96 (qd, J= 15.6, 7.9 Hz, 3H), 3.77 (ddd, J= 25.8, 23.4, 11.9 Hz, 2H), 3.58 (dd, J= 17.2, 8.3 Hz, 2H), 3.38 (s, 2H), 3.25 - 3.11 (m, 3H), 3.05 - 2.94 (m, 1 H), 2.94 - 2.81 (m, 4H), 2.73 (dd, J= 20.9,

5 11.0 Hz, 1 H), 2.45 (d, J = 6.9 Hz, 5H), 2.32 - 2.07 (m, 3H), 1 .92 (d, J = 13.2 Hz, 1 H), 1.72 (s, 1 H), 1.64 - 1.51 (m, 1H), 1.18 (t, J= 7.0 Hz, 2H), 1.00 (ddd, J= 14.6, 11.8, 8.5 Hz, 6H), 0.92 - 0.81 (m, 4H), 0.55 - 0.41 (m, 3H).

Example A67. Synthesis of (2E)-4-(dlmethylamlno)-N-(6-{[(1 S)-1 -{[(8S,14S)-22-ethyl-4- 10 hydroxy-21 -[2-(methoxymethyl)pyrldin-3-yl]-18,18-dlmethyl-9,15-dloxo-1 6-oxa-10,22,28- triazapentacyclo[18.5.2.1 2 , e .1 10 , 14 .0 23 , 2T ]nonacosa-1(26),2,4,6(29),20,23(27),24-heptaen-8- yl]carbamoyl}-2-methylpropyl](methyl)carbamoyl}pyridin-3-yl) but-2-enamide

Step 1. To a mixture of (6 3 S,4S)-4-amino-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-(methoxymethyl)pyridin-3-yl)-

15 10,1 O-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione TFA salt (225 mg, 0.28 mmol) and (E)-N-(5-(4-(dimethylamino)but- 2-enamido)picolinoyl)-yV-methyl-L-valine TFA salt (260 mg crude, 0.56 mmol) in DMF (5 mL) at 0 °C were added DIPEA (0.46 mL, 2.8 mmol) followed by HATU (140 mg, 0.36 mmol). The mixture was stirred at 0- 10 °C for 1 h, then concentrated under reduced pressure and the residue was purified by prep-HPLC to

20 give (2E)-4-(dimethylamino)-N-(6-{[(1 S)-1 -{[(8S,14S)-22-ethyl-4-hydroxy-21 -[2-(methoxymethyl)pyridin-3- yl]-18,18-dimethyl-9,15-dioxo-16-oxa-10,22,28-triazapentacyc lo[18.5.2.1 2 ,M ^.^ O^ nnonacosa- 1(26), 2,4, 6(29),20, 23(27), 24-heptaen-8-yl]carbamoyl}-2-methylpropyl](methyl)carbamoyl} pyridin-3-yl)but- 2-enamide TFA salt (23.3 mg, 8% yield over 2 steps) as a solid. LCMS (ESI): m/z [M+Na] calc’d for CwHezNgOeNa 992.5; found 992.4; 1 H NMR (400 MHz, CDsOD) 69.05 (d, J= 2.5 Hz, 1H), 8.85 - 8.71 (m,

25 1H), 8.43 (ddd, J= 33.3, 18.0, 2.6 Hz, 2H), 8.01 - 7.87 (m, 2H), 7.83 - 7.70 (m, 1H), 7.60 - 7.47 (m, 2H), 7.31 - 7.19 (m, 1H), 7.07 - 6.90 (m, 2H), 6.70 - 6.36 (m, 3H), 5.81 - 5.61 (m, 1H), 4.50 - 4.20 (m, 4H), 4.01 - 3.68 (m, 3H), 3.64 - 3.35 (m, 5H), 3.27 - 3.08 (m, 3H), 3.04 - 2.44 (m, 11 H), 2.36 - 2.10 (m, 3H), 1.93 (d, J = 13.0 Hz, 1 H), 1.61 (dd, J = 34.3, 21.6 Hz, 3H), 1.39 - 1.16 (m, 3H), 1.12 - 0.81 (m, 6H), 0.78 - 0.45 (m,

6H).

852

SUBSTITUTE SHEET (RULE 26) Example A54. Synthesis of (2S)-2-{1-[(3S)-1-[(2£)-4-(dlmethylamlno)but-2-enoyl]pyrrol ldln- 3-yl]-N-methylformamldo}-N-[(8S,14S)-22-ethyl-4-hydroxy-21-[ 2-(methoxymethyl)pyrldln-3-yl]- 18,18-dimethyl-9,15-dioxo-16-oxa-10,22,28-trlazapentacyclo[1 8.5.2.1 2 ,M 10 , 14 .0 23 , 27 ]nonacosa- 5 1 (26)^,4,6(29),20,23(27),24-heptaen-8-yl]-3-methylbutanamlde o

NH TFA/DCM

.N. = k ¾°<_

Step 1. To a mixture of fert-butyl W-methyl-W-((S)-pyrrolidine-3-carbonyl)-L-valinate (210 mg,

0.73 mmol) in DMF (4 mL) at rt were added 4-(dimethylamino)-4-methylpent-2-ynoic acid (450 mg, 2.9 mmol), DIPEA (1.2 mL, 7.3 mmol), and HATU (332 mg, 0.88 mmol). The mixture was stirred at rt for 1 h

10 then diluted with EtOAc, and the mixture washed with H 2 O, brine, dried over NaaSOA, and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give fert-butyl N-((S)-1-(4-(dimethylamino)-4-methylpent-2-ynoyl)pyrrolidine -3- carbonyl)-N-methyl-L-valinate (140 mg, 45% yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for C23H39N3O4421 .3; found 422.3.

15 Step 2. A mixture of fert-butyl N-((S)-1-(4-(dimethylamino)-4-methylpent-2-ynoyl)pyrrolidine -3- carbonyl)-N-methyl-L-valinate (130 mg, 0.31 mmol) in DCM (2 mL) and TEA (1 mL) was stirred at rt for 90 min. The mixture was concentrated under reduced pressure to give N-((S)-1-(4-(dimethylamino)-4- methylpent-2-ynoyl)pyrrolidine-3-carbonyl)-N- methyl-L-valine TEA salt (150 mg) as an oil, which was used directly in the next step without further purification. LCMS (ESI): m/z [M+H] calc'd for C19H31N3O4

20 365.2; found 366.2.

Step 3. (3S)-1 -(4-(dimethylamino)-4-methylpent-2-ynoyl)-N-((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-2 5 - hydroxy-1 2 -(2-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-di oxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8- oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan- 2-yl)-N-methylpyrrolidine-3-carboxamide TEA salt was synthesized in a manner similar to 1 -acryloyl-AE

25 ((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-(methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-di oxo- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 W-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane- 4-yl)amino)-3-methyl-1 -oxobutan-2-yl)-N- methylazetidine-3-carboxamide except (2S)-2-{1 -[(3S)-1 -[(2E)-4- (dimethylamino)but-2-enoyl]pyrrolidin-3-yl]-N-methylformamid o}-N-[(8S,14S)-22-ethyl-4-hydroxy-21-[2- (methoxymethyl)pyridin-3-yl]-18,18-dimethyl-9,15-dioxo-16-ox a-10,22,28-

30 triazapentacyclo[18.5.2.1 2 ,».1 «".O^nonacosa-I (26),2,4,6(29),20,23(27),24-heptaen-8-yl]-3- methylbutanamide TEA salt. (120 mg, 54% yield over 2 steps) as a solid. 1 H-NMR (400 MHz, CD3OD) 6 8.76 - 8.68 (m, 1 H), 8.44 (s, 1 H), 8.02 - 7.94 (m, 1 H), 7.94 - 7.84 (m, 1 H), 7.65 - 7.43 (m, 3H), 7.27 - 7.14 (m, 1 H), 7.06 - 6.96 (m, 1 H), 6.65 - 6.48 (m, 1 H), 5.62 - 5.46 (m, 1 H), 4.81 - 4.57 (m, 1 H), 4.46 - 4.22 (m, 3H), 4.10 - 3.35 (m, 11 H), 3.26 - 2.93 (m, 6H), 2.91 - 2.51 (m, 4H), 2.42 - 2.09 (m, 9H), 1.95 - 1.87 (m,

853

SUBSTITUTE SHEET (RULE 26) 1 H), 1.85 - 1.40 (m, 6H), 1.38 - 1.10 (m, 6H), 1.07 - 0.81 (m, 9H), 0.56 - 0.38 (m, 3H). LCMS (ESI): m/z [M+H] CsaHeeNeOe found 947.7.

Example A95. Synthesis of (2S)-N-[(8S,14S)-22-ethyl-4-hydroxy-21 -[2- 5 (methoxymethyl)pyridln-3-yl]-18,18-dlmethyl-9,15-dioxo-16-ox a-10,22,28- triazapentacyclo[18^.2.1 2 , t .1 10 , 14 .0 23 , 27 ]nonacosa-1(26),2,4,6(29),20,23(27),24-heptaen-8-yl]-3 · methyl-2-{N-methyl-1-[(3S)-1-[4-(moipholin-4-yl)but-2-ynoyl] pyrrolldin-3-yl]formamldo}butanamlde

Step 1. A mixture of fert-butyl (2S)-3-methyl-2-[N-methyl-1-(3S)-pyrrolidin-3- 10 ylformamido]butanoate (500 mg, 1.8 mmol), 4-(morpholin-4-yl)but-2-ynoic acid (1 .49 g, 8.8 mmol), DIPEA (682 mg, 5.3 mmol) and GIF (635 mg, 2.3 mmol) in DMF (5 mL) was stirred at 0 °C for 2 h.. The mixture was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give fert-butyl N-methyl-N-((S)-1-(4-morpholinobut-2-ynoyl)pyrrolidine-3-car bonyl)-L- valinate (150 mg, 19% yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for C23H37N3O5435.3; found 436.5.

15 Step 2. A mixture of fert-butyl fV-methyl-AA((S)-1-(4-morpholinobut-2-ynoyl)pyrrolidine-3- carbonyl)-L-valinate (250 mg, 0.57 mmol) in DCM (5 mL) and TFA (2.5 mL) was stirred at rt for 2h. The mixture was concentrated under reduced pressure to give (2S)-3-methyl-2-[N-methyl-1 -[(3S)-1 -[4- (morpholin-4-yl)but-2-ynoyl]pyrrolidin-3-yl]formamido]butano ic acid (310mg, crude) as an oil, which was used directly in the next step without further purification. LCMS (ESI): m/z [M+H] calc’d for C19H29N3O5 20 379.2; found 380.2.

Step 3. A mixture of (6 3 S,4S)-4-amino-1 1 -ethyl-2 5 -hydroxy-1 2 -(2-(methoxymethyl)pyridin-3-yl)- 10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione (250 mg, 0.4 mmol), DIPEA (516 mg, 4.0 mmol), (2S)-3-methyl-2- [N-methyl-1-[(3S)-1-[4-(morpholin-4-yl)but-2-ynoyl]pyrrolidi n-3-yl]formamido]butanoic acid (182 mg, 0.48

25 mmol), and COMU (205 mg, 0.48 mmol) in DMF (3 mL) was stirred at -20 °C for 2 h. The mixture was diluted with H 2 O (10 mL), then extracted with EtOAc (3 x 10 mL) and the combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na 2 SO 4 , and filtered. The mixture was concentrated under reduced pressure and the residue was purified by reverse-phase silica gel column chromatography to give (2S)-N-[(8S,14S)-22-ethyl-4-hydroxy-21 -[2-(methoxymethyl)pyridin-3-yl]-18,18-dimethyl-9,15-

30 dioxo-16-oxa-10,22,28-triazapentacyclo[18.5.2.1 2 , e .1 ^.^.O^.^nonacosa-I (26), 2, 4, 6(29), 20, 23(27), 24- heptaen-8-yl]-3-methyl-2-{N-methyl-1-[(3S)-1-[4-(morpholin-4 -yl)but-2-ynoyl]pyrrolidin-3- yl]formamido}butanamide (207 mg, 53% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for CssH/oNeOg 986.5; found 987.8; 1 H NMR (400 MHz, DMSO-db) 69.39 - 9.28 (m, 1H), 8.74 (t, J= 4.8, 1H), 8.70 - 8.04 (m, 1 H), 7.98 - 7.90 (m, 1 ,5H), 7.82 (d, J = 7.7 Hz, 0.5H), 7.63 - 7.46 (m, 3H), 7.26 - 7.10 (m, 1 H), 7.03

854

SUBSTITUTE SHEET (RULE 26) (s, 1 H), 6.58 - 6.43 (m, 1 H), 5.44 - 5.30 (m, 1 H), 5.06 (q, 0.5H), 4.72 (t, J = 11.0 , 0.5H), 4.39 - 4.20 (m, 3H), 4.15 (d, J = 11.1 Hz, 1 H), 4.09 - 3.85 (m, 4H), 3.66 (s, 2H), 3.65 - 3.58 (m, 4H), 3.58 - 3.55 (m, 2H), 3.55 - 3.48 (m, 3H), 3.47 - 3.41 (m, 3H), 3.31 (s, 2H), 3.10 (s, 2H), 2.92 (s, 1H), 2.89 - 2.65 (m, 5H), 2.68 (s, 1 H), 2.45 - 2.38 (m , 1 H), 2.29 - 2.24 (m, 1 H), 2.23 - 1.99 (m, 3H), 1.82 (d, J = 12.1 Hz, 1 H), 1.76 -

5 1.62 (m, 1 H), 1.61 - 1.45 (m, 1H), 1.14 - 1.04 (m, 2H), 1.02 - 0.92 (m, 3H), 0.91 - 0.86 (m, 3H), 0.83 - 0.77 (m, 3H), 0.77 - 0.70 (m, 2H), 0.50 - 0.35 (m, 3H).

Example A145. Synthesis of two atropisomers of (2S)-2-{1 -[(3S)-1 -(but-2-ynoyl)pyrrolldln-3- yl]-N-methylformamldo}-N-[(8S,14S,20Af)-22-ethyl-4-hydroxy-2 1-{2-[(1 S)-1 -methoxyethyl]pyrldin-3- 10 ylM 8,18-dim ethyl-9,15-dloxo-16-oxa-10,22,28-trlazapentacyclop 8.5.2.1V-1 ’V'-iF.^nonacosa-

1(26)^, 4, 6(29), 20, 23(27), 24-heptaen-8-yl]-3-methylbutanamide

Step 1. To a mixture of but-2-ynoic acid (222 mg) and CIP (588 mg) in ACN (8 mL) at 0 °C under an atmosphere of Ar was added DIPEA (681 mg). The mixture was stirred at 0 °C then fert-butyl N-

15 methyl-N-((S)-pyrrolidine-3-carbonyl)-L-valinate (500 mg) in ACN (3 mL) was added dropwise and the mixture stirred at 0 °C for 2 h. EtOAc was added and the mixture was washed with brine (3 x 20 mL), dried over anhydrous NaaSO*, and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give fert-butyl N-((S)-1 -(but-2- ynoyl)pyrrolidine-3-carbonyl)-N-methyl-L-valinate as a solid. LCMS (ESI): m/z [M+H] calc’d for

20 C19H30N 2 O4350.2; found 352.1.

Step 2. A mixture of fert-butyl N-((S)-1 -(but-2-ynoyl)pyrrolidine-3-carbonyl)-N-methyl-L-valinate (200 mg) in DCM (4 mL) and TFA (2 mL) was stirred at 0 °C for 2 h. The mixture was concentrated under reduced pressure with azeotropic removal of H 2 O using toluene (4 mL x 2) to give N-((S)-1 -(but-2- ynoyl)pyrrolidine-3-carbonyl)-N-methyl-L-valinate as a solid. LCMS (ESI): m/z [M+H] calc’d for

25 C15H22N 2 O4294.2; found 295.2.

Step 3. Two atropisomers of (2S)-2-{1 -[(3S)-1 -(but-2-ynoyl)pyrrolidin-3-yl]-N-methylformamido}- N-[(8S,14S,20M)-22-ethyl-4-hydroxy-21 -{2-[(1 S)-1 -methoxyethyl]pyridin-3-yl}-18,18-dimethyl-9,15-dioxo- 16-oxa-10,22,28-triazapentacyclo[18.5.2.1 2 , e .1 ^.^.O^.^nonacosa-I (26), 2, 4,6(29), 20, 23(27), 24-heptaen- 8-yl]-3-methylbutanamide was synthesized in a manner similar to (2S)-N-{(8S,14S)-22-ethyl-4-hydroxy-

30 21 -[2-(methoxymethyl)pyridin-3-yl]-18,18-dimethyl-9,15-dioxo-1 6-oxa-10,22,28- triazapentacyclo[18.5.2.1 a , e .1 " ’«.O^nonacosa-I (26), 2,4,6(29), 20,23(27), 24-heptaen-8-yl]-3-methyl-2- {N-methyl-1 -[(3S)-1 -[4-(morpholin-4-yl)but-2-ynoyl]pyrrolidin-3-yl]formamido}bu tanamide except (2S)-3- methyl-2-[A/-methyl-1 -[(3S)-1 -[4-(morpholin-4-yl)but-2-ynoyl]pyrrolidin-3-yl]formamido]bu tanoic acid was substituted with N-((S)-1-(but-2-ynoyl)pyrrolidine-3-carbonyl)-N- methyl-L-valinate. (43.3 mg, 12% yield)

855

SUBSTITUTE SHEET (RULE 26) and (33 mg, 9% yield) both as solids. LCMS (ESI): m/z [M+H] calc’d for CsaHesN/Oe 915.5; found 916.7; 1H NMR (400 MHz, DMSO-db) δ 9.34 - 9.27 (m, 1 H), 8.78 (t, J= 2.5 Hz, 1 H), 8.68 (t, J= 8.5 Hz, 0.5H), 8.20 - 8.11 (m, 0.6H), 7.95 (ddt, J= 5.4, 3.5, 1.7 Hz, 2H), 7.63 - 7.60(m, 1H), 7.61 - 7.49 (m, 2H), 7.13 (s, 1H), 7.03 (d, J= 6.2 Hz, 1H), 6.60 - 6.49 (d, J= 35.5 Hz, 1H), 5.43 - 5.39 (m, 1H), 5.12 - 5.00 (m, 0.7H),

5 4.74 (d, J = 10.6 Hz, 0.4H), 4.32 - 4.25 (m, 1 H), 4.18 - 3.85 (m, 5H), 3.81 - 3.45 (m, 8H), 3.18 - 3.02 (m,

5H), 2.93 - 2.80 (m, 4H), 2.80 - 2.70 (m, 2H), 2.42 - 2.36 (m, 1 H), 2.31 - 2.20 (m, 1 H), 2.18 - 1.96 (m, 6H), 1.85 - 1.74 (m, 1 H), 1.74 - 1.63 (m, 1 H), 1.62- 1.42 (m, 1 H), 1.32 - 1.16 (m, 4H), 1.15-1.05 (t, J = 6.3 Hz, 4H), 1.04 - 0.95 (m, 2H), 0.95 - 0.85 (m, 5H), 0.68 - 0.52(m, 4H), 0.52 - 0.37 (m, 4H). and LCMS (ESI): m/z [M+H] calc’d for CSZHWNTOB 915.5; found 916.7; 1 H NMR (400 MHz, DMSO-db) δ 9.36 - 9.28 (m, 1 H), 10 8.77 (dd, J = 4.7, 1.8 Hz, 1 H), 8.62 - 8.57 (m, 0.5H), 8.15 - 8.07 (m, 0.5H), 7.95 (s, 1 H), 7.87 - 7.81 (m,

1H), 7.65 - 7.51 (m, 3H), 7.37 - 7.25 (m, 1H), 7.10 - 7.03 (m, 1H), 6.54 (d, J= 35.5Hz, 1H), 5.52 - 5.21 (m, 2H), 4.78 - 4.66 (m, 0.5H), 4.34 - 4.20 (m, 3H), 4.15 - 3.85(m, 4H), 3.85 - 3.42 (m, 7H), 3.22 - 3.11 (m,

3H), 2.97- 2.72 (m, 7H), 2.62 - 2.54 (m, 1 H), 2.28 - 1.96 (m, 7H), 1.95 - 1.74 (m, 2H), 1.73 - 1.44 (m, 2H),

1.42 - 1.37 (m, 3H), 1.28 - 1.14 (m, 1 H), 1.03 - 0.85 (m, 6H), 0.83 - 0.72 (m, 7H), 0.71 - 0.55 (m, 3H).

15

856

SUBSTITUTE SHEET (RULE 26) Example A28. Synthesis of (2S)-N-[(8S,14S)-22-ethyl-4-hydroxy-21 -[4- (methoxymethyl)pyrldln-3-yl]-18,18-dimethyl-9,15-dloxo-16-ox a-10,22,28- trlazapentacyck>[18.5.2.1 A [2,6].1 A [10,14].0 A [23,27l]nonacosa-1(26), 2, 4,6(29), 20, 23(27), 24-heptaen- 8-yl]-3-methyl-2-[N-methyl-1-[(3S)-1-(prop-2-enoyl)pyrrolidi n-3-yl]formamido]butanamide

Step 1. To a mixture of 3-bromo-4-(methoxymethyl)pyridine (1.00 g, 5.0 mmol), 4,4, 5,5- tetramethy l-2-(tetramethyl- 1 ,3,2-dioxaborolan-2-yl)-1 ,3,2-dioxaborolane (1.51 g, 5.9 mmol) and KOAc (1.21 g, 12.3 mmol) in toluene (10 mL) at rt under an atmosphere of Ar was added Pd(dppf)Cl 2 (362 mg, 0.5 mmol). The mixture was heated to 110 °C and stirred overnight, then concentrated under reduced

10 pressure to give 4-(methoxymethyl)-3-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)pyridine, which was used directly in the next step directly without further purification. LCMS (ESI): m/z [M+H] calc’d for C13H20BNO3249.2; found 250.3.

Step 2. To a mixture of 4-(methoxymethyl)-3-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)pyridine (290 mg, 1.16 mmol), K3PO4 (371 mg, 1.75 mmol) and tert-butyl N-[(8S,14S)-21 -iodo-18,18-

15 dimethyl-9,15-dioxo-4-[(triisopropylsilyl)oxy]-16-oxa-10,22, 28- triazapentacyclo[18.5.2.1 Λ [2,6].1 A [10,14].0 A [23,27]]nonacosa-1 (26), 2, 4, 6(29), 20,23(27), 24-heptaen-8- yljcarbamate (500 mg, 0.58 mmol) in 1 ,4-dioxane (5 mL) and H 2 O (1 mL) at rt under an atmosphere of Ar was added Pd(dppf)Cl 2 (43 mg, 0.06 mmol). The mixture was heated to 70 °C and stirred for 2 h, then H 2 O added and the mixture extracted with EtOAc (2 x 10 mL). The combined organic layers were washed

857

SUBSTITUTE SHEET (RULE 26) with brine (10 mL), dried over anhydrous NaaSO*, and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give fert-butyl N- [(8S,14S)-21 -[4-(methoxymethyl)pyridin-3-yl]-18,18-dimethyl-9,15-dioxo-4 -[(triisopropylsilyl)oxy]-16-oxa- 10,22,28-triazapentacyclo[18.5.2.1 A [2,6].1 A [10,14].0 A [23,27]]nonacosa-1 (26), 2, 4, 6(29), 20 ,23(27), 24- 5 heptaen-8-yl]carbamate (370 mg, 74% yield) as a foam. LCMS (ESI): m/z [M+H] calc’d for CwHe/NsOSi 853.6; found 854.6.

Step 3. A mixture of fert-butyl AA[(8S,14S)-21 -[4-(methoxymethyl)pyridin-3-yl]-18,18-dimethyl-

9,15-dioxo-4-[(triisopropylsilyl)oxy]-16-oxa-10,22,28- triazapentacyclo[18.5.2.1 A [2,6].1 A [10,14].0 A [23,27]]nonacosa-1 (26),2,4,6(29),20,23(27),24-heptaen-8- 10 yljcarbamate (350 mg, 0.41 mmol), CS2CO3 (267 mg, 0.82 mmol) and Etl (128 mg, 0.82 mmol) in DMF (4 mL) was stirred at 35 °C overnight. H 2 O was added and the mixture was extracted with EtOAc (2 x 15 mL). The combined organic layers were washed with brine (15 mL), dried over anhydrous NaaSO*, and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give fert-butyl N-[(8S,14S)-22-ethyl-21-[4-(methoxymethyl)pyridin-3-yl]-18,1 8- 15 dimethyl-9,15-dioxo-4-[(triisopropylsilyl)oxy]-16-oxa-10,22, 28- triazapentacyclo[18.5.2.1 A [2,6].1 A [10,14].0 A [23,27]]nonacosa-1 (26),2,4,6(29),20,23(27),24-heptaen-8-yl] carbamate (350 mg, 97% yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for C50H71N5O7S1 881.5; found 882.6.

Step 4. A mixture of fert-butyl N-[(8S,14S)-22-ethyl-21-[4-(methoxymethyl)pyridin-3-yl]-18,1 8-

20 dimethyl-9,15-dioxo-4-[(triisopropylsilyl)oxy]-16-oxa-10,22, 28- triazapentacyclo[18.5.2.1 A [2,6].1 A [10,14].0 A [23,27]]nonacosa-1 (26),2,4,6(29),20,23(27),24-heptaen-8-yl] carbamate (350 mg, 0.4 mmol) and 1 M TBAF in THF (0.48 mL, 0.480 mmol) in THF (3 mL) at 0 °C under an atmosphere of Ar was stirred for 1 h. The mixture was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give fert-butyl N-[(8S,14S)-22-ethyl-4-

25 hydroxy-21 -[4- (methoxymethyl)pyridin-3-yl]-18,18-dimethyl-9,15-dioxo-16-ox a-10,22,28- triazapentacyclo[18.5.2.1 A [2,6].1 A [10,14].0 A [23,27]]nonacosa-1 (26),2,4,6(29),20,23(27),24-heptaen-8- yljcarbamate (230 mg, 80% yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for C41H51N5O7725.4; found 726.6.

Step 5. To a mixture of fert-butyl N-[(8S,14S)-22-ethyl-4-hydroxy-21 -[4- (methoxymethyl)pyridin-

30 3-yl]-18,18-dimethyl-9,15-dioxo-16-oxa-10,22,28- triazapentacyclo[18.5.2.1 A [2,6].1 A [10,14].0 A [23,27]]nonacosa-1 (26),2,4,6(29),20,23(27),24-heptaen-8- yljcarbamate (200 mg, 0.28 mmol) in 1 ,4-dioxane (2 mL) at 0 °C under an atmosphere of Ar was added 4M HCI in 1 ,4-dioxane (2 mL, 8 mmol). The mixture was allowed to warm to rt and was stirred overnight, then concentrated under reduced pressure to give (8S,14S)-8-amino-22-ethyl-4-hydroxy-21-[4-

35 (methoxymethyl)pyridin-3-yl]-18,18-dimethyl-16-oxa-10,22,28- triazapentacyclo[18.5.2.1 A [2,6].1 A [10,14].0 A [23,27]]nonacosa-1 (26),2,4,6(29),20,23(27),24-heptaene-

9,15-dione (200 mg). LCMS (ESI): m/z [M+H] calc’d for C36H43N5O5625.3; found 626.5.

Step 6. To a mixture of (2S)-3-methyl-2-[N-methyl-1 -[(3S)-1 -(prop-2-enoyl)pyrrolidin-3- yl]formamido]butanoic acid (108 mg, 0.38 mmol) and (8S,14S)-8-amino-22-ethyl-4-hydroxy-21-[4-

40 (methoxymethyl)pyridin-3-yl]-18,18-dimethyl-16-oxa-10,22,28- triazapentacyclo[18.5.2.1 A [2,6].1 A [10,14].0 A [23,27]]nonacosa-1 (26),2,4,6(29),20,23(27),24-heptaene-

9,15-dione (200 mg, 0.32 mmol) in DCM (3 mL) at 0 °C was added DIPEA (165 mg, 1.3 mmol) and

858

SUBSTITUTE SHEET (RULE 26) COMU (274 mg, 0.64 mmol) in portions. The mixture was stirred at 0 °C for 2 h, h½0 added and extracted with EtOAc (2 x 10 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography then prep-HPLC to give (2S)-/\A[(8S,14S)-22-ethyl-4-

5 hydroxy-21 -[4-(methoxymethyl)pyridin-3-yl]-18,18-dimethyl-9,15-dioxo-1 6-oxa-10,22,28- triazapentacyclo[18.5.2.1 Λ [2,6].1 A [10,14].0 A [23,27]]nonacosa-1 (26),2,4,6(29),20,23(27),24-heptaen-8-yl]- 3-methyl-2-[N-methyl-1-[(3S)-1-(prop-2-enoyl)pyrrolidin-3-yl ]formamido]butanamide (16 mg, 5.6% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for CsoHeaNyOe 889.5; found 890.6; 1 H NMR (400 MHz, DMSO- flfe) 69.33 (dd, J = 9.1 , 6.9 Hz, 1H), 8.79 - 8.46 (m, 2H), 7.93 (s, 1H), 7.68 - 7.58 (m, 2H), 7.53 (t, J= 8.5

10 Hz, 1 H), 7.26 - 6.98 (m, 2H), 6.71 - 6.47 (m, 2H), 6.24 - 6.07 (m, 1 H), 5.80 - 5.60 (m, 1 H), 5.49 - 5.18 (m,

1 H), 4.45 - 4.07 (m, 4H), 4.08 - 3.87 (m, 3H), 3.87 - 3.64 (m, 4H), 3.64 - 3.40 (m, 5H), 3.34 (s, 2H), 3.30 (s, 2H), 3.23 (d, J= 1.8 Hz, 1H), 2.94 - 2.74 (m, 6H), 2.16 - 2.01 (m, 3H), 1.82 - 1.47 (m, 3H), 1.08 (q, J = 8.9, 8.0 Hz, 1H), 1.00 - 0.88 (m, 6H), 0.82 (d, J = 10.8 Hz, 4H), 0.76 - 0.66 (m, 2H), 0.44 (d, J= 14.2 Hz,

3H).

15

859

SUBSTITUTE SHEET (RULE 26) Example A316. Synthesis of (2/7)-2-(((1-(4-(dimethylamino)-4-methylpent-2-ynoyl)azetidi n-3- yl)oxy)methyl)-N-((6 3 S,4S}-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)py rldln-3-yl)-1 O.10-dlmethyl-SJ-dloxo- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-lndola-6(1 ,3)-pyridazlna-2(1 ,3)- benzenacycloundecaphane-4-yl)-3-methylbutanamlde

Step 1. To a mixture of 2-(((1-((benzyloxy)carbonyl)azetidin-3-yl)oxy)methyl)-3-meth ylbutanoic acid (650 mg, 2 mmol) and di-tert-butyl dicarbonate (883 mg, 4 mmol) in BuOH (10 mL) was added 4- dimethylaminopyridine (124 mg, 1 mmol). The mixture was heated to 30 °C and stirred for 1 h, then diluted with H 2 O (50 mL) and extracted with EtOAc (50 mL x 3). The combined organic layers were

860

SUBSTITUTE SHEET (RULE 26) concentrated under reduced pressure and the residue was purified by silica gel column chromatography to afford benzyl 3-(2-(fert-butoxycarbonyl)-3-methylbutoxy)azetidine-1-carbox ylate (450 mg, 56% yield) as an oil. LCMS (ESI): m/z [M+Na] calc'd for CaiHsiNOsNa 400.2; found 400.2.

Step 2. A mixture of benzyl 3-(2-(fert-butoxycarbonyl)-3-methylbutoxy)azetidine-1 -carboxylate 5 (450 mg, 1.19 mmol) and Pd/C (50 mg) in THF (30 mL) was stirred for 2 h under an atmosphere of hfe (15 psi). The mixture was filtered and the filtrate was concentrated under reduced pressure to give fert-butyl 2-((azetidin-3-yloxy)methyl)-3-methylbutanoate (300 mg, 100% yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for CiaHasNOa 243.2; found 244.2; 1 H NMR (400 MHz, CDCIa) 64.35 - 4.25 (m, 1 H), 3.71 - 3.63 (m, 2H), 3.63 - 3.56 (m, 2H), 3.50 (t, J= 8.0 Hz, 1H), 3.43 (dd, J = 9.0, 4.0 Hz, 1H), 2.37 - 2.26 (m, 1H), 2.21 10 (br. s, 1 H), 1.92 - 1.81 (m, 1 H), 1.47 (s, 9H), 0.93 (d, J = 6.8 Hz, 6H).

Step 3. To a mixture of fert-butyl 2-((azetidin-3-yloxy)methyl)-3-methylbutanoate (270 mg, 1.11 mmol), 4-(dimethylamino)-4-methylpent-2-ynoic acid (860 mg, 5.55 mmol) and DIPEA (1.56 g, 11.1 mmol) in DMF (20 mL) at 0 °C was added TaP (2.12 g, 6.7 mmol). The mixture was stirred at 0 °C for 1 h, diluted with EtOAc (200 mL), then washed with H 2 O (30 mL x 5), brine (30 mL), dried over anhydrous NaaSO*

15 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give fert-butyl 2-(((1-(4-(dimethylamino)-4-methylpent-2-ynoyl)azetidin-3- yl)oxy)methyl)-3-methylbutanoate (200 mg, 47% yield) as an oil. LCMS (ESI): m/z [M+H] calc'd for CaiHseNaO* 380.3; found 381.3.

Step 4. To a mixture of fert-butyl 2-(((1 -(4-(dimethylamino)-4-methylpent-2-ynoyl)azetidin-3- 20 yl)oxy)methyl)-3-methylbutanoate (190 mg, 0.5 mmol) in DCM (4 mL) was added TFA (2 mL). The mixture was stirred for 1 h, then concentrated under reduced pressure to give 2-(((1 -(4-(dimethylamino)-4- methylpent-2-ynoyl)azetidin-3-yl)oxy)methyl)-3-methylbutanoi c add (162 mg, 100% yield) as an oil, which was used directly in the next step without further purification. LCMS (ESI): m/z [M+H] calc'd for C17H28N 2 O4324.2; found 325.3.

25 Step 5. To a solution of (2S)-3-(3-bromophenyl)-2-[(fert-butoxycarbonyl)amino]propano ic acid (100 g, 290 mmol) in DMF (1 L) at room temperature was added NaHCOs (48.8 g, 581.1 mmol) and Mel (61.9 g, 435.8 mmol). The reaction mixture was stirred for 16 h and was then quenched with H 2 O (1 L) and extracted with EtOAc (3 x 1 L). The combined organic layers were washed with brine (3 x 500 mL), dried over NaaS04, filtered, and concentrated under reduced pressure. The residue was purified by silica 30 gel chromatography (13% EtOAc/pet. ether) to give methyl (S)-3-(3-bromophenyl)-2-((fert- butoxycarbonyl)amino)propanoate (109 g, crude). LCMS (ESI): m/z [M+Na] calc'd for CisH2oBrN04 380.05; found 380.0.

Step 6. To a stirred solution of methyl (2S)-3-(3-bromophenyl)-2-[(fert- butoxycarbonyl)amino]propanoate (108 g, 301.5 mmol) and bis(pinacolato)diboron (99.53 g, 391.93 35 mmol) in 1 ,4-dioxane (3.2 L) was added KOAc (73.97 g, 753.70 mmol) and Pd(dppf)Cla (22.06 g, 30.15 mmol). The reaction mixture was heated to 90 °C for 3 h and was then cooled to room temperature and extracted with EtOAc (2 x 3 L). The combined organic layers were washed with brine (3 x 800 mL), dried over NaaS04, filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (5% EtOAc/pet. ether) to give methyl (S)-2-((fert-butoxycarbonyl)amino)-3-(3-(4,4,5,5- 40 tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)propanoate (96 g, 78.6% yield). LCMS (ESI): m/z [M+Na] calc’d for CaiHaaBNOe 428.22; found 428.1.

861

SUBSTITUTE SHEET (RULE 26) Step 7. To a mixture of methyl (2S)-2-[(terf-butoxycarbonyl)amino]-3-[3-(4,4,5,5-tetramethy l- 1 ,3,2-dioxaborolan-2-yl)phenyl]propanoate (94 g, 231.9 mmol) and 3-(5-bromo-1 H-indol-3-yl)-2,2- dimethylpropyl acetate (75.19 g, 231.93 mmol) in 1 ,4-dioxane (1.5 L) and H 2 O (300 mL) was added K2CO3 (64.11 g, 463.85 mmol) and Pd(DtBPF)Cl 2 (15.12 g, 23.19 mmol). The reaction mixture was 5 heated to 70 °C and stirred for 4 h. The reaction mixture was extracted with EtOAc (2 x 2 L) and the combined organic layers were washed with brine (3 x 600 mL), dried over NaaSO*, filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (20% EtOAc/pet. ether) to give methyl (S)-3-(3-(3-(3-acetoxy-2,2-dimethylpropyl)-1 H-indol-5-yl)phenyl)-2-((te/i- butoxycarbonyl)amino)propanoate (130 g, crude). LCMS (ESI): m/z [M+H] calc'd for CsoHaeNaOe 523.28;

10 found 523.1.

Step 8. To a solution of methyl (2S)-3-(3-[3-[3-(acetyloxy)-2,2-dimethylpropyl]-1 H-indol-5- yl]phenyl)-2-[(te/1-butoxycarbonyl)amino]propanoate (95.0 g, 181.8 mmol) and iodine (36.91 g, 145.41 mmol) in THF (1 L) at -10 °C was added AgOTf (70.0 g, 272.7 mmol) and NaHCOa (22.9 g, 272.65 mmol). The reaction mixture was stirred for 30 min and was then quenched by the addition of sat. NaaSaOa (100

15 mL) at 0 °C. The resulting mixture was extracted with EtOAc (3 x 1 L) and the combined organic layers were washed with brine (3 x 500 mL), dried over NaaSO*, filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (50% EtOAc/pet. ether) to give methyl (S)-3-(3-(3-(3-acetoxy-2,2-dimethylpropyl)-2-iodo-1H-indol-5 -yl)phenyl)-2-((tert- butoxycarbonyl)amino)propanoate (49.3 g, 41.8% yield). LCMS (ESI) m/z. [M + H] calcd for CsoHsyINaOe:

20 649.18; found 649.1.

Step 9. To a solution of methyl (2S)-3-(3-[3-[3-(acetyloxy)-2,2-dimethylpropyl]-2-iodo-1 H-indol-5- yl]phenyl)-2-[(te/t-butoxycarbonyl)amino]propanoate (60 g, 92.5 mmol) in THF (600 mL) was added a solution of LiOH»H 2 O (19.41 g, 462.5 mmol) in H 2 O (460 mL). The resulting solution was stirred overnight and then the pH was adjusted to 6 with HCI (1 M). The resulting solution was extracted with EtOAc (2 x

25 500 mL) and the combined organic layers was washed with sat. brine (2 x 500 mL), dried over NaaSO*. filtered, and concentrated under reduced pressure to give (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(3-(3- hydroxy-2,2-dimethylpropyl)-2-iodo-1H-indol-5-yl)phenyl)prop anoic acid (45 g, 82.1% yield). LCMS (ESI): m/z [M+Na] calc'd for CarHwINaOe 615.13; found 615.1.

Step 10. To a solution of (2S)-2-[(terf-butoxycarbonyl)amino]-3-[3-[3-(3-hydroxy-2,2-

30 dimethylpropyl)-2-iodo-1 H-indol-5-yl]phenyl]propanoic add (30 g, 50.6 mmol) and methyl (3S)-1 ,2- diazinane-3-carboxylate (10.9 g, 75.9 mmol) in DCM (400 mL) was added NMM (40.97 g, 405.08 mmol), HOST (2.05 g, 15.19 mmol), and EDCI (19.41 g, 101.27 mmol). The reaction mixture was stirred overnight and then the mixture was washed with sat. NH4CI (2 x 200 mL) and sat. brine (2 x 200 mL), and the mixture was dried over NaaSO*, filtered, and concentrated under reduced pressure to give methyl (S)-

35 1-((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(3-(3-hydroxy-2,2 -dimethylpropyl)-2-iodo-1H-indol-5- yl)phenyl)propanoyl)hexahydropyridazine-3-carboxylate (14 g, 38.5% yield). LCMS (ESI): m/z [M+H] calc’d for C33H43IN4O6718.23; found 719.4.

Step 11. To a solution of methyl (S)-1-((S)-2-((fe/t-butoxycarbonyl)amino)-3-(3-(3-(3-hydroxy - 2,2-dimethylpropyl)-2-iodo-1 H-indol-5-yl)phenyl)propanoyl)hexahydropyridazine-3-carboxyl ate (92 g,

40 128.0 mmol) in THF (920 mL) at 0 °C was added a solution of LiOH«H 2 O (26.86 g, 640.10 mmol) in H 2 O (640 mL). The reaction mixture was stirred for 2 h and was then concentrated under reduced pressure to give (S)-1-((S)-2-((fe/t-butoxycarbonyl)amino)-3-(3-(3-(3-hydroxy -2,2-dimethylpropyl)-2-iodo-1H-indol-5-

862

SUBSTITUTE SHEET (RULE 26) yl)phenyl)propanoyl)hexahydropyridazine-3-carboxylic acid (90 g, crude). LCMS (ESI): m/z [M+H] calc’d for 0 3 2Η4ΐΙΝ 4 0β 705.22; found 705.1.

Step 12. To a solution of of (3S)-1-[(2S)-2-[(fert-butoxycarbonyl)amino]-3-[3-[3-(3-hydro xy-2,2- dimethylpropyl)-2-iodo-1 H-indol-5-yl]phenyl]propanoyl]-1 ,2-diazinane-3-carboxylic acid (90 g, 127.73 5 mmol) in DCM (10 L) at 0 °C was added HOBt (34.52 g, 255.46 mmol), DIPEA (330.17 g, 2554.62 mmol) and EDCI (367.29 g, 1915.96 mmol). The reaction mixture was stirred for 16 h and was then concentrated under reduced pressure. The mixture was extracted with DCM (2 x 2 L) and the combined organic layers were washed with brine (3 x 1 L), dried over NaaSCX filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (50% EtOAc/pet. ether) to give 10 fert-butyl ((6 3 S,4S)-1 2 -iodo-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 ' H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (70 g, 79.8% yield). LCMS (ESI): m/z [M+H] calc’d for C32H39IN4O5687.21 ; found 687.1.

Step 13. A 1 L round-bottom flask was charged with fert-butyl ((6 3 S,4S)-1 2 -iodo-10,10-dimethyl- 5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 8 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- 15 benzenacycloundecaphane-4-yl)carbamate (22.0 g, 32.042 mmol), toluene (300.0 mL), Pdz(dba)3 (3.52 g, 3.845 mmol), S-Phos (3.95 g, 9.613 mmol), and KOAc (9.43 g, 96.127 mmol) at room temperature. To the mixture was added 4,4,5,5-tetramethyl-1 ,3,2-dioxaborolane (26.66 g, 208.275 mmol) dropwise with stirring at room temperature. The resulting solution was stirred for 3 h at 60 °C. The resulting mixture was filtered, and the filter cake was washed with EtOAc. The filtrate was concentrated under reduced pressure 20 and the remaining residue was purified by silica gel column chromatography to afford fert-butyl ((6 3 S,4S)- 10,10-dimethyl-5,7-dioxo-12-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro- 1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (22 g, 90 % yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for C38H51BN4O7687.3; found 687.4.

Step 14. A mixture of fert-butyl ((6 3 S,4S)-10,10-dimethyl-5,7-dioxo-1 2 -(4,4,5,5-tetramethyl-1 ,3,2- 25 dioxaborolan-2-yl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate (2.0 g, 2.8 mmol), 3-bromo-2-[(1 S)-1-methoxyethyl]pyridine (0.60 g, 2.8 mmol), Pd(dppf)Cl 2 (0.39 g, 0.5 mmol), and K3PO4 (1.2 g, 6.0 mmol) in 1 ,4-dioxane (50 mL) and H 2 O (10 mL) under an atmosphere of N 2 was heated to 70 °C and stirred for 2 h. The mixture was diluted with H 2 O (50 mL) and extracted with EtOAc (3 x 50 mL). The combined organic layers were 30 washed with brine (3 x 50 mL), dried over anhydrous Na 2 SO 4 , and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give fert- butyl ((6 3 S,4S)-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 ' H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (1.5 g, 74% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C40H49N5O6695.4; found 696.5.

35 Step 15. To a solution of fert-butyl ((6 3 S,4S)-1 2 -(2-((S)-1-methoxyethyl) pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 Ή-8-oxa-l (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl) carbamate (20 g, 28.7 mmol) and CS2CO3 (18.7 g, 57.5 mmol) in DMF (150 mL) at 0 °C was added a solution of ethyl iodide (13.45 g, 86.22 mmol) in DMF (50 mL). The resulting mixture was stirred overnight at 35 °C and was then diluted with H 2 O (500 mL). The mixture was 40 extracted with EtOAc (2 x 300 mL) and the combined organic layers were washed with brine (3 x 100 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography to give fert-butyl ((6 3 S,4S)-1 '-ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-

863

SUBSTITUTE SHEET (RULE 26) 10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate (4.23 g, 18.8% yield) and the atropisomer (5.78 g, 25.7% yield) as solids. LCMS (ESI): m/z [M+H] calc’d for C42H53N5O6724.4; found 724.6.

Step 16. A mixture of fert-butyl ((6 3 S,4S)-1 '-ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10- 5 dimethyl-5, 7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate (1.3 g, 1.7 mmol) in TFA (10 mL) and DCM (20 mL) was stirred at 0 °C for 2 h. The mixture was concentrated under reduced pressure to afford (6 3 S,4S)-4-amino- 1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa- 1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-5,7-dione (1.30 g, crude) as a solid.

10 LCMS (ESI): m/z [M+H] calc’d for C37H45N5O4623.3; found 624.4.

Step 17. To a mixture of (6 3 S,4S)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10- dimethyl-6',6 2 , 6 3 , 6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione (258 mg, 0.41 mmol) and 2-(((1-(4-(dimethylamino)-4-methylpent- 2-ynoyl)azetidin-3-yl)oxy)methyl)-3-methylbutanoic acid (162 mg, 0.5 mmol) in DMF (4 mL) at 0 °C was 15 added a mixture of HATU (188 mg, 0.5 mmol) and DIPEA (534 mg, 4.14 mmol) in DMF (2 mL). The mixture was stirred at 0 °C for 1 h, then diluted with HzO (30 mL) and extracted with EtOAc (30 mL x 3). The combined organic layers were concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 2-(((1-(4-(dimethylamino)-4-methylpent-2-ynoyl)azetidin-3- yl)oxy)methyl)-W-((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo- 20 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 W-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-

4-yl)-3-methylbutanamide (250 mg, 64% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C54H71N7O7 929.5; found 930.5; 1 H NMR (400 MHz, CD3OD) 68.90 - 8.79 (m, 1 H), 8.54 - 8.21 (m, 1 H), 8.15 - 7.91

(m, 2H), 7.88 - 7.67 (m, 2H), 7.65 - 7.52 (m, 2H), 7.47 - 7.15 (m, 2H), 5.80 - 5.52 (m, 1H), 4.53 - 4.23 (m,

5H), 4.23 - 3.93 (m, 3H), 3.90 - 3.76 (m, 2H), 3.75 - 3.58 (m, 3H), 3.57 - 3.44 (m, 1 H), 3.38 (s, 1 H), 3.29 - 25 3.26 (m, 2H), 3.21 - 2.85 (m, 8H), 2.82 - 2.65 (m, 3H), 2.51 - 2.30 (m, 1 H), 2.24 - 2.03 (m, 1 H), 1.99 - 1.87

(m, 1 H), 1 .86 - 1.69 (m, 6H), 1.67 - 1.57 (m, 2H), 1 .57 - 1.39 (m, 4H), 1.45 - 1.05 (m, 2H), 1.04 - 0.96 (m,

3H), 0.96 - 0.88 (m, 3H), 0.88 - 0.79 (m, 3H), 0.79 - 0.63 (m, 3H), 0.56 (s, 1H).

Step 18. 2-(((1 -(4-(dimethylamino)-4-methylpent-2-ynoyl)azetidin-3-yl)oxy)m ethyl)-A/-((6 3 S,4S)- 1 '-ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 H- 30 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)-3-methylbutanamide (180 mg, 0.194 mmol) was purified by prep-HPLC to afford (2fl)-2-(((1-(4-(dimethylamino)-4-methylpent-2- ynoyl)azetidin-3-yl)oxy)methyl)-N- ((6 3 S,4S)-1 '-ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-6',6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 'H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-3-methylbutanamide (41.8 mg, 23.2% yield) as a solid. LCMS (ESI): m/z 35 [M+H] calc'd for C54H71N7O7930.5; found 930.5; Ή NMR (400 MHz, MeOD) 68.74 (d, J = 4.0 Hz, 1 H),

8.53 - 8.30 (m, 1 H), 8.10 - 7.95 (m, 1 H), 7.94 - 7.80 (m, 2H), 7.68 (t, J = 8.0 Hz, 1 H), 7.65 - 7.58 (m,

1 H), 7.58 - 7.46 (m, 2H), 7.38 - 7.17 (m, 2H), 5.73 - 5.60 (m, 1 H), 4.52 - 4.40 (m, 1 H), 4.35 - 4.15 (m,

4H), 4.14 - 3.95 (m, 2H), 3.90 - 3.72 (m, 3H), 3.71 - 3.45 (m, 4H), 3.30 - 3.20 (m, 3H), 3.06 - 2.72 (m,

5H), 2.49 - 2.28 (m, 4H), 2.28 - 2.20 (m, 3H), 2.18 - 2.06 (m, 1 H), 2.00 - 1.90 (m, 1 H), 1.90 - 1.52 (m,

40 4H), 1.52 - 1.40 (m, 5H), 1.40 - 1.22 (m, 4H), 1.09 - 0.92 (m, 8H), 0.90 - 0.75 (m, 3H), 0.71 - 0.52 (m,

3H) and (2S)-2-(((1 -(4-(dimethylamino)-4-methylpent-2-ynoyl)azetidin-3-yl)oxy)m ethyl)-N-((6 3 S,4S)-1 '- ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10, 10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-

864

SUBSTITUTE SHEET (RULE 26) oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)-3-methylbutanamide (51.2 mg, 28.4% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for C54H71N7O7930.5; found 930.3; 1 H NMR (400 MHz, MeOD) 68.74 (d, J = 4.0 Hz, 1 H), 8.28 - 8.20 (m, 0.6H), 8.11 - 7.98 (m, 1 H), 7.97 - 7.80 (m, 2H), 7.73 - 7.48 (m, 4H), 7.46 - 7.36 (m, 0.4H), 7.33 - 7.26 (m, 1 H), 7.25 - 7.13 (m, 1 H), 5.79 - 5.66 (m,

5 1 H), 4.54 - 4.43 (m, 1 H), 4.42 - 4.01 (m, 7H), 3.90 - 3.75 (m, 2H), 3.73 - 3.48 (m, 4H), 3.27 - 3.12 (m, 3H), 3.08 - 2.99 (m, 1 H), 2.96 - 2.85 (m, 2H), 2.84 - 2.69 (m, 2H), 2.69 - 2.49 (m, 6H), 2.41 - 2.29 (m, 1H), 2.15 - 2.05 (m, 1H), 1.95 - 1.85 (m, 1H), 1.84 - 1.71 (m, 1H), 1.71 - 1.38 (m, 11H), 1.14 - 1.00 (m, 3H), 1.00 - 0.71 (m, 9H), 0.70 - 0.56 (m, 3H).

10 Example A427. Synthesis of 3-((1-(4-(dimethylamlno)-4-methylpent-2-ynoyl)azetidln-3- yl)oxy)-AA((6 3 S,4S,Z)-1 1 -ethyH 2 -(2-((S)-1 -methoxyethyl)pyrldln-3-yl)-10,10-dlmethyl-5,7-dloxo- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-thiazola-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-4-yl)propanamkJe

15 Step 1. To a mixture of tert-butyl prop-2-ynoate (5 g, 40 mmol) and [3-(3-hydroxyazetidin-1 - yl)phenyl]methyl formate (4.1 g, 20 mmol) in DCM (150 mL) was added DMAP (9.8 g, 80 mmol). The mixture was stirred for 2 h, then diluted with HzO and washed with HzO (60 mL x 3). The organic layer was dried over NazSO*, filtered, the filtrate was concentrated under reduced pressure and the residue purified by silica gel column chromatography to give benzyl (E)-3-((3-(fert-butoxy)-3-oxoprop-1 -en-1 -

20 yl)oxy)azetidine-1 -carboxylate (6.6 g, 90% yield) as an oil. LCMS (ESI): m/z [M+Na] calc’d for CieHzsNOsNa 356.2; found 356.2.

Step 2. A mixture of benzyl (E)-3-((3-(fert-butoxy)-3-oxoprop-1 -en-1 -yl)oxy)azetidine-1 - carboxylate (1.4 g, 4 mmol) and Pd/C (200 mg) in THF (10 mL) was stirred under an atmosphere of Hz (1 atmosphere) for 16 h. The mixture was filtered and the filtrate and was concentrated under reduced

25 pressure to give fert-butyl 3-(azetidin-3-yloxy)propanoate, which was used directly in the next step. LCMS (ESI): m/z [M+H] calc'd for C10H19NO3201.1 ; found 202.2.

Step 3. To a mixture of fert-butyl 3-(azetidin-3-yloxy)propanoate (300 mg, 1.5 mmol) and 4- (dimethylamino)-4-methylpent-2-ynoic acid (2.3 g, 15 mmol) in DMF (15 mL) at 5 °C was added DIPEA (1.9 g, 15 mmol) and T3P (4.77 g, 7.5 mmol) dropwise. The mixture was stirred at 5 °C for 2 h, then HzO

30 and EtOAc (80 mL) were added. The organic and aqueous layers were separated and the organic layer was washed with HzO (20 mL x 3), brine (30 mL), dried over anhydrous NBZSOA and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by preparative-HPLC to afford

865

SUBSTITUTE SHEET (RULE 26) tort-butyl 3-((1-(4-(dimethylamino)-4-methylpent-2-ynoyl)azetidin-3-yl) oxy)propanoate (60 mg, 12% yield) as an oil. LCMS (ESI): m/z [M+H] calc'd for CieHsoN^ 338.2; found 339.2.

Step 4. A mixture tort-butyl 3-((1-(4-(dimethylamino)-4-methylpent-2-ynoyl)azetidin-3- yl)oxy)propanoate (70 mg, 0.21 mmol) in TFA/ DCM (1 :3, 2 mL) was stirred at 0 - 5 °C for 1 h, then 5 concentrated under reduced pressure to give 3-({1 -[4-(dimethylamino)-4-methylpent-2-ynoyl]azetidin-3- yl}oxy)propanoic acid (56 mg, 95% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C14H22N 2 O4282.2; found 283.3.

Step 5. To a mixture of 3-((1-(4-(dimethylamino)-4-methylpent-2-ynoyl)azetidin-3- yl)oxy)propanoic acid (56 mg, 0.19 mmol), (6 3 S,4S,2)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-

10 3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 ' H-8-oxa-2(4,2)-thiazola-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-5,7-dione (90 mg, 0.14 mmol) and DIPEA (200 mg, 1.9 mmol) in DMF (1 mL) at 0 °C was added HATU (110 mg, 0.38 mmol) portion-wise. The mixture was stirred at 0 °C for 1 h, then H 2 O added and the mixture extracted with EtOAx (150 mL x 2). The combined organic layers were washed with H 2 O (150 mL) and brine (150 mL), then dried over anhydrous NazS04 and filtered. The

15 filtrate was concentrated under reduced pressure and the residue was purified by preparative-HPLC to give 3-((1 -(4-(dimethylamino)-4-methylpent-2-ynoyl)azetidin-3-yl)oxy)- N-((6 3 S,4S,2)-1 1 -ethyl-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-2(4,2)- thiazola-1(5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)propanamide (12.6 mg, 7.5 % yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for C48He2N e 07S 894.5; found 895.3; 1 H NMR (400 MHz, CDaOD) 6

20 8.73 (dd, J = 4.8, 1.6 Hz, 1 H), 8.57 (s, 1 H), 8.28 (s, 0.3H), 7.84 (m, 1 H), 7.71 (m, 1 H), 7.52 (m, 3H), 5.76 (dd, J= 30.2, 7.8 Hz, 1H), 4.40 (m, 4H), 4.32 - 4.12 (m, 4H), 4.06 (dd, J= 12.4, 6.0 Hz, 1H), 3.97 - 3.86 (m, 1 H), 3.79 - 3.66 (m, 4H), 3.46 (dd, J = 14.8, 4.8 Hz, 1 H), 3.41 - 3.33 (m, 3H), 3.29 - 3.19 (m, 1 H), 3.17 - 3.05 (m, 1H), 2.79 (m, 1H), 2.73 - 2.50 (m, 3H), 2.49 - 2.43 (m, 3H), 2.38 (s, 3H), 2.21 (dd, J= 12.6, 9.6 Hz, 1 H), 1.95 (d, J = 12.8 Hz, 1 H), 1.86 - 1.73 (m, 1 H), 1.61 (dd, J = 12.6, 3.6 Hz, 1 H), 1.51 (s, 2H), 1.46 -

25 1.43 (m, 4H), 1.38 - 1.27 (m, 3H), 1.01 - 0.86 (m, 6H), 0.44 (d, J= 11.6 Hz, 3H).

866

SUBSTITUTE SHEET (RULE 26) Example A716. Synthesis of (3S)-1 -acrytoyl-AA((2S)-1 -(((6 3 ¾4S)-1 '-ethyl-l ^-((SM - methoxyethyl)pyrldin-3-yl)-10,10-dimethyl-5,7-dioxo-2 1 ,2 2 ,2 3 ,2 ® ,6 1 ,6 2 ,6 3 ,6 4 ,6 6 ,6 6 -decahydro-1 1 H- 8- oxa-ltS^^indola-etl^pyridazina^tS.IH’yridinacycloundecapha ne-A-yOamino^S-methyl-l- oxobutan-2-yl)-N-methylpyrrolldlne-3-carboxamlde

5

Step 1. To a solution of methyl (tert-butoxycarbonyl)-L-serinate (10 g, 45 mmol) in anhydrous MeCN (150 mL), was added DIPEA (17 g, 137 mmol). The reaction mixture was stirred at 45 °C for 2 h to give methyl 2-((tert-butoxycarbonyl)amino)acrylate in solution. LCMS (ESI): m/z [M+Na] calc’d for C9H15NO4201.1 ; found 224.1.

10 Step 2. To a solution of methyl 2-((fert-butoxycarbonyl)amino)acrylate (12 g, 60 mmol) in anhydrous MeCN (150 mL) at 0 °C, was added 4-DMAP (13 g, 90 mmol) and (ΒοφΟ (26 g, 120 mmol). The reaction was stirred for 6 h, then quenched with HzO (100 mL) and extracted with DCM (200 mL x 3). The combined organic layers were washed with brine (150 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column

15 chromatography to give methyl 2-(bis(terf-butoxycarbonyl)amino)acrylate (12.5 g, 65% yield) as solid. LCMS (ESI): m/z [M+Na] calc’d for CuHaNOe 301.2; found 324.1.

Step 3. To a mixture of 5-bromo-1 ,2,3,6-tetrahydropyridine (8.0 g, 49 mmol) in MeOH (120 mL) under an atmosphere of Ar was added methyl 2-{b/s[(fert-butoxy)carbonyl]amino}prop-2-enoate (22 g, 74 mmol). The mixture was stirred for 16 h, then concentrated under reduced pressure and the residue was

20 purified by silica gel column chromatography to give methyl 2-(bis(tert-butoxycarbonyl)amino)-3-(5- bromo-3,6-dihydropyridin-1(2H)-yl)propanoate (12 g, 47% yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for CigHaiBrNaOe 462.1 ; found 463.1.

867

SUBSTITUTE SHEET (RULE 26) Step 4. To a mixture of methyl 2-(bis(fert-butoxycarbonyl)amino)-3-(5-bromo-3,6-dihydropyri din- 1 (2/7)-yl)propanoate (14 g, 30 mmol) in 1 ,4-dioxane (30 mL) and H 2 O (12 mL) was added LIOH (3.6 g,

151 mmol). The mixture was heated to 35 °C and stirred for 12 h, then 1 M HCI was added and the pH adjusted to -3-4. The mixture was extracted with DCM (300 mL x 2) and the combined organic layers 5 were dried over anhydrous NaaSO* and filtered. The filtrate was concentrated under reduced pressure to give 3-(5-bromo-3,6-dihydropyridin-1(2H)-yl)-2-((te/t-butoxycarbo nyl)amino)propanoic acid (10 g, 85% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for CiaHaiBrNaO* 348.1; found 349.0.

Step 5. To a mixture of 3-(5-bromo-3,6-dihydropyridin-1 (2/7)-yl)-2-((tert- butoxycarbonyl)amino)propanoic acid (10 g, 30 mmol), DIPEA (12 g, 93 mmol) and methyl (3S)-1 ,2- 10 diazinane-3-carboxylate (5.4 g, 37 mmol) in DMF (100 mL) at 0 °C under an atmosphere of Ar was added HATU (13 g, 34 mmol). The mixture was stirred at 0 °C for 2 h, then H 2 O added and the mixture extracted with EtOAc (300 mL x 2). The combined organic layers were dried over anhydrous Na 2 SO 4 , filtered, the filtrate was concentrated under reduced pressure and the residue was purified by preparative-HPLC to give methyl (3S)-1 -(3-(5-bromo-3,6-dihydropyridin-1 (2/7)-yl)-2-((tert- 15 butoxycarbonyl)amino)propanoyl)hexahydropyridazine-3-carboxy late (9.0 g, 55% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for CigHaiBrlsUOs 474.1 ; found 475.1.

Step 6. A mixture of methyl (3S)-1 -(3-(5-bromo-3,6-dihydropyridin-1 (2H)-y\)-2-((tert- butoxycarbonyl)amino)propanoyl)hexahydropyridazine-3-carboxy late (9.0 g, 18 mmol), K2CO3 (4.5 g, 32 mmol), Pd(dppf)Cl 2 .DCM (1.4 g, 2 mmol), 3-(1 -ethyl-2-{2-[(1 S)-1-methoxyethyl]pyridin-3-yl}-5-(4, 4,5,5- 20 tetramethyl-1 ,3,2-dioxaborolan-2-yl)indol-3-yl)-2,2-dimethylpropan-1 -ol (9.8 g, 20 mmol) in 1 ,4-dioxane

(90 mL) and H 2 O (10 mL) under an atmosphere of Ar was heated to 75 °C and stirred for 2 h. H 2 O was added and the mixture was extracted with EtOAc (200 mL x 3). The combined organic layers were dried over NaaSO*, filtered, the filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give methyl (3S)-1 -(2-((ferf-butoxycarbonyl)amino)-3-(5-(1 -ethyl- 25 3-(3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)py ridin-3-yl)-1H-indol-5-yl)-3,6-dihydropyridin- 1 (2H)-yl)propanoyl)hexahydropyridazine-3-carboxylate (4.0 g, 25% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for C42H60N6O7760.5; found 761.4.

Step 7. To a mixture of methyl (3S)-1-(2-((fert-butoxycarbonyl)amino)-3-(5-(1-ethyl-3-(3-hy droxy- 2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1 H-indol-5-yl)-3,6-dihydropyridin-1(2H)- 30 yl)propanoyl)hexahydropyridazine-3-carboxylate (4.1 g, 5.0 mmol) in THF (35 mL) at 0 °C was added LiOH (0.60 g, 27 mmol). The mixture was stirred at 0 °C for 1.5 h, then 1 M HCI added to adjust pH to ~6- 7 and the mixture extracted with EtOAc (200 mL x 3). The combined organic layers were dried over Na 2 SO 4 , filtered and the filtrate was concentrated under reduced pressure to give (3S)-1-(2-((tert- butoxycarbonyl)amino)-3-(5-(1-ethyl-3-(3-hydroxy-2,2-dimethy lpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3- 35 yl)-1 H- indol-5-yl)-3,6-dihydropyridin-1 (2/7)-yl)propanoyl)hexahydropyridazine-3-carboxylic acid (3.6 g,

80% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C^HseNeO? 746.4; found 747.4.

Step 8. To a mixture of (3S)-1 -(2-((fert-butoxycarbonyl)amino)-3-(5-(1 -ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1 -methoxyethyl)pyridin-3-yl)-1 H-indol-5-yl)-3,6-dihydropyridin-1 (2/7)- yl)propanoyl)hexahydropyridazine-3-carboxylic acid (3.6 g , 5.0 mmol) and DIPEA (24 g ,190 mmol) in 40 DCM (700 mL) under an atmosphere of Ar was added EDCI.HCI (28 g, 140 mmol) and HOBT (6.5 g, 50 mmol). The mixture was heated to 30 °C and stirred for 16 h at 30 °C, then concentrated under reduced pressure. The residue was diluted with EtOAc (200 mL) and washed with H 2 O (200 mL x 2), brine (200

868

SUBSTITUTE SHEET (RULE 26) mL), dried over NaaSO* and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give terf-butyl ((6 3 S)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -decahydro-1 1 H- 8-oxa-

1(5,3)-indola-6(1 ,3)-pyridazina-2(5,1)-pyridinacycloundecaphane-4-yl)carbamat e (1.45 g, 40% yield) as a 5 solid. LCMS (ESI): m/z [M+H] calc’d for C+iHseNeOe 728.4; found 729.4.

Step 9. To a mixture of fert-butyl ((6 3 S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-2 1 ,2 2 , 2 3 ,2 6 ,6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -decahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(5,1 )-pyridinacydoundecaphane-4-yl)carbamate (130 mg, 0.20 mmol) in DCM (1.0 mL) at 0 °C was added TFA ( 0.3 mL). The mixture was warmed to room temperature and stirred for 2 h, then 10 concentrated under reduced pressure to give (6 3 S)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3- yl)-10,10-dimethyl-2 1 ^ 2 ^, 2 ® ,6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -decahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(5,1 )-pyridinacycloundecaphane-5,7-dione, which was used directly in the next step directly without further purification. LCMS (ESI): m/z [M+H] calc’d for CaFfcaNeQ* 628.4; found 629.4.

Step 10. To a mixture of ((6 3 S)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10- 15 dimethyl-2 1 ,2 2 , 2 3 ,2 e ,6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -decahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(5,1 )- pyridinacycloundecaphane-5,7-dione (130 mg, 0.2 mmol), DIPEA (270 mg, 2.0 mmol) and (2S)-3-methyl- 2-{N-methyl-1-[(3S)-1-(prop-2-enoyl)pyrrolidin-3-yl]formamid o}butanoic acid (118 mg, 0.40 mmol) in DMF (3.0 mL) at 0 °C under an atmosphere of Ar was added HATU (87 mg, 0.30 mmol) in portions. The mixture was stirred at 0 °C for 1 h, then diluted with H 2 O extracted with EtOAc (30 mL x 2). The combined 20 organic layers were dried over anhydrous Na 2 SO 4 , filtered, the filtrate was concentrated under reduced pressure and the residue was purified by preparative-HPLC to give (3S)-1 -acryloyl-N-((2S)-1 -(((6 3 S,4S)- 1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 1 ,2 2 ,2 3 , 2« 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - decahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(5,1 )-pyridinacydoundecaphane-4-yl)amino)-3- methyl-1-oxobutan-2-yl)-N- methylpyrrolidine-3-carboxamide (17.2 mg, 10% yield ) as a solid. LCMS 25 (ESI): m/z [M+H] calc’d for CsoHeeNeO* 892.5; found 893.5; 1 H NMR (400 MHz, CD3OD) 68.74 (d, J = 4.4 Hz, 1H), 7.93 - 7.90 (m, 1H), 7.56 - 7.51 (m, 3H), 7.43 (d, J= 4.4 Hz, 1H), 6.63 - 6.53 (m, 2H), 6.33 - 6.23 (m, 2H),5.83 - 5.70 (m, 1 H), 4.73 - 4.70 (d, J = 11.0 Hz, 1 H), 4.48 - 4.45 (d, J = 13.0 Hz, 1 H), 4.12 - 4.10 (m, 3H), 3.86 - 3.81 (m, 4H), 3.79 - 3.75 (m, 1 H), 3.72 - 3.69 (m, 3H), 3.57 - 3.47 (m, 2H), 3.21 - 3.09 (m, 1H), 3.07 - 3.04 (q, 4H), 3.02 - 2.95 (m, 3H), 2.86 - 2.82(m, 3H), 2.66 - 2.48 (m, 2H), 2.29 - 2.17 (m, 4H), 30 2.11 - 1.98 (m, 2H), 1.95 - 1.91 (m,1 H), 1.45 (d, J = 6.2 Hz, 3H), 1.23 - 1.16 (m, 2H), 1.09 - 1 .04 (m, 1 H),

0.97 - 0.93 (m, 3H), 0.92 - 0.81 (m, 5H), 0.67 - 0.63 (m, 3H).

869

SUBSTITUTE SHEET (RULE 26) Example A663. The synthesis of (2fl)-2-(((1-(4-(dimethylamlno)-4-methylpent-2- ynoyl)870yrldine870-3-yl)oxy)methyl)-N-((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)870yridine-3- yl)-10,10-dimethyl-5,7-dioxo-2 1 ,6 2 > 6 3 f 6 4 ,6 5 ,6 6 -decahydro-1 1 /f8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(5,1)-pyridinacycloundecaphane-4-yl)-3-methylbut anamide

5

To a mixture of (6 3 S,4S)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)870yridine-3-yl)-10,10- dimethyl-2 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -decahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(5,1 )- pyridinacycloundecaphane-5,7-dione (100 mg, 0.16 mmol), ®-2-(((1-(4-(dimethylamino)-4-methylpent-2- ynoyl)870yridine870-3-yl)oxy)methyl)-3-methylbutanoic acid (80 mg, 0.24 mmol) and DIPEA (825 mg, 6.4

10 mmol) in DMF (2 mL) at 0 °C, was added HATU (95 mg, 0.24 mmol). The reaction mixture was stirred at 0 °C for 1 h, then poured into HzO (60 mL), extracted with EtOAc (80 mL x 2). The combined organic layers were washed with HzO (80 mL) and brine (80 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography to afford (2fl)-2-(((1-(4-(dimethylamino)-4-methylpent-2-ynoyl)870yrid ine870-3-yl)oxy)methyl)-N-((6 3 S,4S)-

15 1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)870yridine-3-yl)-10,10-dimethyl-5,7-dioxo-2 1 ,2 2 , 2 3 ,2 e ,6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - decahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(5,1 )-pyridinacycloundecaphane-4-yl)-3- methylbutanamide (55 mg, 36% yield) as solid. 1 H NMR (400 MHz, CDsOD) 68.76 - 8.70 (m, 1 H), 8.49 (dd, J = 4.3, 1.4 Hz, 0.1 H), 7.93 - 7.87 (m, 1 H), 7.58 - 7.50 (m, 3H), 7.41 (dd, J = 8.8, 3.2 Hz, 1 H), 6.26 (d, J = 16.8 Hz, 1 H), 5.96 (t, J = 9.6 Hz, 1 H), 4.47 (d, J = 12.8 Hz, 1 H), 4.39 - 4.28 (m, 2H), 4.21 - 3.97

20 (m, 5H), 3.96 - 3.70 (m, 5H), 3.68 - 3.54 (m, 3H), 3.51 - 3.35 (m, 1 H), 3.11 (d, J = 22.7 Hz, 3H), 3.00 - 2.67 (m, 5H), 2.46 - 2.30 (m, 7H), 2.24 (s, 3H), 2.11 (d, J = 12.4 Hz, 1H), 1.92 (d, J = 13.2 Hz, 1H), 1.85 - 1.60 (m, 3H), 1.45 (d, J = 7.8 Hz, 6H), 1.32 (d, J = 16.0 Hz, 3H), 1.12 (dt, J = 24.5, 6.8 Hz, 3H), 0.95 (m, 6H), 0.76 (m, 6H). LCMS (ESI): m/z [M+H] calc’d for CsaHy+NeO? 934.6; found 935.5.

25 Example A646. The synthesis of (2FQ-2-(((1-(4-(dimethylamino)-4-methylpent-2- ynoyl)azetidin-3-yl)oxy)methyl)-W-((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10- dlmethyl-S.Z-dloxo-e 1 ,6 2 ,6 3 ,6 4 ,6 e ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyrldazina-2(3,1 )- plperldlnacycloundecaphane-4-yl)-3-methylbutanamide

870

SUBSTITUTE SHEET (RULE 26) Step 1. A mixture of tert-butyl ((6 3 S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-2 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -decahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(5,1 )-pyridinacydoundecaphane-4-yl)carbamate (0.2 g, 0.28 mmol) and Pd/C (0.2 g, 2 mmol) in MeOH (10 mL) was stirred at 25 °C for 16 h under an Ha atmosphere. The reaction mixture was filtered through 5 Celite, concentrated under reduced pressure to afford tert-butyl ((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(3,1)-piperidinacycloundecaphane-4-yl)carbam ate as solid. LCMS (ESI): m/z [M+H] calc’d for C-nHseNeOe 730.4; found 731.4.

Step 2. To a solution of tert-butyl ((6 3 S,4S)-1 '-ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10- 10 dimethyl-5, 7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(3,1 )- piperidinacycloundecaphane-4-yl)carbamate (150 mg, 0.2 mmol) in DCM (1 .5 mL) at 0 °C was added TFA (0.5 mL). The reaction mixture was stirred at 20 °C for 1 h, then concentrated under reduced pressure to afford (6 3 S,4S)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(3,1 )-piperidinacycloundecaphane-5,7-dione as

15 solid. LCMS (ESI): m/z [M+H] calc’d for CseHsoNeCb 630.4; found 631.4.

Step 3. To a mixture of (6 3 S,4S)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10- dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(3,1 )- piperidinacycloundecaphane-5,7-dione (240 mg, 0.4 mmol), DIPEA (982 mg, 2 mmol) and (fl)-2-(((1 -(4- (dimethylamino)-4-methylpent-2-ynoyl)azetidin-3-yl)oxy)methy l)-3-methylbutanoic acid (148 mg, 0.45 20 mmol) in DMF (4 mL) at 0 °C under argon atmosphere, was added HATU (173 mg, 0.46 mmol) in portions. The reaction mixture was stirred at 0 °C under an argon atmosphere for 1 h, then quenched with H 2 O at 0 °C. The resulting mixture was extracted with EtOAc (30 mL x 2). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by reverse phase chromatography to afford (2fl)-2-(((1 -(4-(dimethylamino)-4-methylpent-2-ynoyl)azetidin- 25 3-yl)oxy)methyl)-N-((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-

6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(3,1 )- piperidinacycloundecaphane-4-yl)-3-methylbutanamide (150 mg, 38% yield) as solid. Ή NMR (400 MHz, CD30D) δ 8.72 (d, J = 4.8 Hz, 1H), 7.88 (d, J = 8.0 Hz, 1H), 7.53-7.49(m, 2H), 7.42 (d, J = 8.4 Hz, 1H), 7.18 (d, J = 8.4 Hz, 1H), 5.95-5.91 (m, 1H), 4.52-4.49 (m, 1H), 4.37-4.25 (m, 3H), 4.18-4.15 (m, 2H), 3.99- 30 3.98 (m, 2H), 3.90-3.86 (m, 1 H), 3.76-3.68 (m, 2H), 3.55-3.50(m, 2H), 3.39-3.36 (m, 2H), 3.20 (s, 3H), 3.02

(s, 3H), 2.89-2.79 (m, 3H), 2.62-2.50 (m, 2H), 2.36 (s, 3H), 2.35-2.30 (m, 1H), 2.26(s, 3H), 2.20-1.15 (m, 1 H), 1.97-1.93 (m, 3H), 1.81 -1.76 (m, 4H), 1.64-1.61 (m, 2H), 1.46-1.43 (m, 6H), 1.36 (d, J = 14.8 Hz, 3H), 1.02 (s, 3H), 0.94 (m, 6H), 0.81 (s, 3H), 0.65 (s, 3H). LCMS (ESI): m/z [M+H] calc’d for CsaHyeNeO? 936.6; found 937.5.

871

SUBSTITUTE SHEET (RULE 26) Example A740. Synthesis of (3S)-1 -aery loyl· W-((2S)-1 -(((2 3 S,6 3 S,4S)-1 1 -ethyH 2 -(2-((S)-1 - methoxyethyl)pyrldln-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)- indola-eil^J-pyrldazlna^^lJ-plperldlnacycloundecaphane-t-yOa minoJ-S-methyl-l-oxobutan-Z- yl)-N- methylpyrrolldlne-3-carboxamlde

5

To a mixture of (6 3 S,4S)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(3,1 )- piperidinacycloundecaphane-5,7-dione (140 mg, 0.20 mmol), DIPEA (570 mg, 4.4 mmol) and (2S)-3- methyl-2-{yV-methyl-1-[(3S)-1-(prop-2-enoyl)pyrrolidin-3-yl] formamido}butanoic acid (124 mg, 0.40 mmol)

10 in DMF (3.0 mL) at 0 °C under an atmosphere of Ar was added HATU (100 mg, 0.30 mmol) in portions. The mixture was stirred at 0 °C for 1 h, then H 2 O was added and the mixture extracted with EtOAc (2 x 30 mL). The combined organic layers were dried over anhydrous NaaSCX and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by preparative-HPLC to give (3S)-1 -acryloyl-N-((2S)- 1 -(((2 3 S,6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-

15 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(3,1 )- piperidinacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2 -yl)-N-methylpyrrolidine-3-carboxamide (41 mg, 20% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for CsoHvoNeO? 894.5; found 895.5; Ή NMR (400 MHz, CD3OD) 68.72 (d, J = 4.8 Hz, 1H), 7.87 (d, J= 7.6 Hz, 1H), 7.51 - 7.49 (m, 2H), 7.42 - 7.37 (m, 1 H), 7.18 - 7.14 (m, 1 H), 6.64 - 6.54 (m, 1 H), 6.30 - 6.23 (m, 1 H), 5.77 - 5.70 (m, 2H), 4.65 - 4.60 (m, 1 H),

20 4.50 - 4.40 (m, 1 H), 4.27 - 4.16 (m, 2H), 4.00 - 3.95 (m, 2H), 3.83 - 3.78(m, 2H), 3.73 - 3.60 (m, 4H), 3.51 - 3.36 (m, 3H), 3.22 - 3.19 (m, 4H), 3.07 (d, J= 6.8 Hz, 2H), 2.99 (d, J= 12.0 Hz, 3H), 2.90 - 2.78 (m, 2H), 2.75 - 2.64 (m, 3H), 2.20 - 2.10 (m, 4H), 2.02 - 1 .93 (m, 3H), 1.87 - 1.64 (m, 4H), 1.45 (d, J = 4.8 Hz, 3H), 1.06 - 1.00 (m, 4H), 0.97 - 0.89 (m, 3H), 0.83 - 0.79 (m, 3H), 0.66 (s, 3H).

872

SUBSTITUTE SHEET (RULE 26) Example A534. (2S)-2-((S)-7-(4-(dlmethylamino)-4-methylpent-2-ynoyl)-1-oxo -2,7- diazaspiro[4.4]nonan-2-yl)-A#-((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyrldin-3-yl)-10,10- dimethyl-5,7-dloxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-lndola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-3-methylbutanamlde o

-C

~ H L N

Λ. CJ

HASTU. DIPEAr H

DMF h-

5 k

Step 1. To a mixture of 1 -fert-butyl 3-methyl pyrrolidine-1 ,3-dicarboxylate (20.0 g, 87.2 mmol) in THF (150 mL) at -78 °C under an atmosphere of nitrogen was added 1M LiHMDS in THF (113.4 mL, 113.4 mmol). After stirring at -78 °C for 40 min, allyl bromide (13.72 g, 113.4 mmol) was added and the mixture was allowed to warm to room temperature and stirred for 4 h. The mixture was cooled to 0 °C,

10 saturated NaCI (30 mL) was added and the mixture extracted with EtOAc. The combined organic layers were dried over anhydrous NaaSO* and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 1 -(fert-butyl) 3-methyl 3- allylpyrrolidine-1 ,3-dicarboxylate (17 g, 72% yield) as an oil. 1 H NMR (300 MHz, CDCIs) 65.80 - 5.60 (m, 1 H), 5.16 - 5.02 (m, 2H), 3.71 (s, 4H), 3.42 (d, J = 9.3 Hz, 2H), 3.27 (t, J= 11.2 Hz, 1H), 2.42 (d, J = 7.6

15 Hz, 2H), 2.38 - 2.24 (m, 1H), 2.05 (s, 1H), 1.85 (dt, J= 14.3, 7.5 Hz, 1H), 1.46 (s, 10H), 1.27 (t, J= 7.1 Hz, 1H).

Step 2. To a mixture of 1 -(fert-butyl) 3-methyl 3-allylpyrrolidine-1 ,3-dicarboxylate (4.0 g, 14.9 mmol) and 2,6-dimethylpyridine (3.18 g, 29.7 mmol) in 1 ,4-dioxane (200 mL) and H 2 O (100 mL) at 0 °C was added Ka0s042Ha0 (0.11 g, 0.3 mmol) in portions. The mixture was stirred for 15 min at 0 °C, then

20 NalC>4 (6.35 g, 29.7 mmol) was added in portions. The mixture was stirred at room temperature for 3 h at room temperature, then cooled to 0 °C and saturated aqueous NaaSaOa (50 mL) added. The mixture was extracted with EtOAc (3 x 100 mL) and the combined organic layers were washed with 2 M HCI, then dried over anhydrous NaaS04 and filtered. The filtrate was concentrated under reduced pressure to give 1 -(fert-butyl) 3-methyl 3-(2-oxoethyl)pyrrolidine-1 ,3-dicarboxylate (4 g, 52% yield) as an oil. 1 H NMR (300

873

SUBSTITUTE SHEET (RULE 26) MHz, CDCI3) δ 5.80 - 5.60 (m, 1H), 5.16 - 5.04 (m, 2H), 3.72 (s, 3H), 3.41 (s, 3H), 3.28 (d, J= 11.0 Hz,

1 H), 2.44 (s, 2H), 2.31 (d, J = 9.1 Hz, 1 H), 1.85 (dt, J = 12.7, 7.5 Hz, 1 H), 1 .69 (s, 1 H), 1.47 (s, 10H).

Step 3. To a mixture of 1 -(iert-butyl) 3-methyl 3-(2-oxoethyl)pyrrolidine-1 ,3-dicarboxylate (6.30 g, 23.2 mmol), in MeOH (70 mL) at 0 °C was added benzyl (2S)-2-amino-3-methylbutanoate (7.22 g, 34.8 5 mmol) and ZnCfe (4.75 g, 34.8 mmol). The mixture was warmed to room temperature and stirred for 30 min, then cooled to 0 °C and NaCNBHa (2.92 g, 46.4 mmol) was added in portions. The mixture was warmed to room temperature and stirred for 2 h, then cooled to 0 °C and saturated aqueous NH4CI added. The mixture was extracted with EtOAc (3 x 200 mL) and the combined organic layers were washed with brine (150 mL), dried over anhydrous Na 2 SO 4 and filtered. The filtrate was concentrated 10 under reduced pressure and the residue was purified by silica gel column chromatography to give 1 -{tert- butyl) 3-methyl 3-(2-(((S)-1-(benzyloxy)-3-methyl-1-oxobutan-2-yl)amino)ethy l)pyrrolidine-1 ,3- dicarboxylate (6.4 g, 54% yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for C25H38N 2 O6462.3; found 463.4.

Step 4. To a mixture of l-(terf-butyl) 3-methyl 3-(2-(((S)-1 -(benzyloxy)-3-methyl-1 -oxobutan-2- 15 yl)amino)ethyl)pyrrolidine-1 ,3-dicarboxylate (4.50 g, 9.7 mmol) in toluene (50 mL) was added DIPEA (12.57 g, 97.3 mmol) and DMAP (1 .19 g, 9.7 mmol). The resulting mixture was heated to 80 °C and stirred for 24 h, then concentrated under reduced pressure and the residue was purified by preparative- HPLC, then by chiral-HPLC to give tert-butyl (fl)-7-((S)-1 -(benzyloxy)-3-methyl-1 -oxobutan-2-yl)-6-oxo- 2,7-diazaspiro[4.4]nonane-2-carboxylate (1.0 g, 32% yield) and fert-butyl (S)-7-((S)-1 -(benzyloxy)-S- 20 methyl-1 -oxobutan-2-yl)-6-oxo-2,7-diazaspiro[4.4]nonane-2-carboxylat e (1.0 g, 32% yield) and as a an oil. LCMS (ESI): m/z [M+H] calc’d for C24H34N 2 O5430.5; found 431.2 and LCMS (ESI): m/z [M+H] calc’d for C24H34N 2 O5430.3; found 431.2.

Step 5. A mixture of fert-butyl (fl)-7-((S)-1 -(benzyloxy)-3-methyl-1 -oxobutan-2-yl)-6-oxo-2,7- diazaspiro[4.4]nonane-2-carboxylate (4.0 g) and 10% Pd/C (1 g) in MeOH (40 mL) was stirred at room 25 temperature under an atmosphere of H2. The mixture was filtered through a pad of Celite pad and the filtrae was concentrated under reduced pressure to give (S)-2-((fl)-7-(fert-butoxycarbonyl)-1-oxo-2,7- diazaspiro[4.4]nonan-2-yl)-3-methylbutanoic acid (4.9 g) as a solid. LCMS (ESI): m/z [M-H] calc’d for C17H28N 2 O5340.2; found 339.3.

Step 6. To a mixture of (6 3 S,4S)-4-amino-1 1 -ethyl- 1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10- 30 dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 8 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione (500 mg, 0.8 mmol) in DCM at 0 °C were added DIPEA (829 mg, 6.4 mmol), ((S)-2-((fl)-7-(tert-butoxycarbonyl)-1 -oxo-2,7-diazaspiro[4.4]nonan-2-yl)-3-methylbutanoic acid (273 mg, 0.8 mmol) and HATU (396 mg, 1.0 mmol) in portions over 1 min. The mixture was allowed to warm to room temperature and stirred 2 h, then concentrated under reduced pressure and the residue 35 was purified by preparative-TLC to give iert-butyl (5fl)-7-((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacydoundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)-6-oxo-2,7- diazaspiro[4.4]nonane-2-carboxylate (500 mg, 64% yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for C54H71N7O8945.5; found 946.5.

40 Step 7. To a mixture of fert-butyl (5fl)-7-((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan- 2-yl)-6-oxo-2,7-

874

SUBSTITUTE SHEET (RULE 26) diazaspiro[4.4]nonane-2-carboxylate (1.0 g, 1.06 mmol) in DCM (10 mL) at 0 °C was added TFA (3 mL) dropwise. The mixture was warmed to room temperature and stirred for 1 h, then concentrated under reduced pressure to give (2S)-N-((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl- 5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- 5 benzenacycloundecaphane-4-yl)-3-methyl-2-((S)-1 -oxo-2,7-diazaspiro[4.4]nonan-2-yl)butanamide (1 .3 g). LCMS (ESI): m/z [M-H] calc'd for C49H63N 7 06846.1 ; found 845.5.

Step 8. To a mixture of (2S)-N-((6 3 S,4S)-1 ’-ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-3-methyl-2-((S)-1 -oxo-2, 7-diazaspiro[4.4]nonan-2-yl)butanamide (500 10 mg, 0.59 mmol) and DIPEA (764 mg, 5.9 mmol) in DMF (5 mL) at 0 °C were added 4-(dimethylamino)-4- methylpent-2-ynoic acid (110 mg, 0.71 mmol) and HATU (292 mg, 0.77 mmol) in portions. The mixture was warmed to room temperature and stirred for 1 h, then H 2 O (10 mL) was added and the mixture extracted with EtOAc (10 mLx 3). The combined organic layers were washed with brine (10 mL), dried over anhydrous NaaSO* and filtered. The filtrate was concentrated under reduced pressure and the 15 residue was purified by preparative-HPLC to give (2S)-2-((S)-7-(4-(dimethylamino)-4-methylpent-2-ynoyl)- 1 -oxo-2, 7-diazaspiro[4.4]nonan-2-yl)-N-((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-3-methylbutanamide (177 mg, 28.94% yield) as a white solid. LCMS (ESI): m/z [M+H] calc’d for CsyFUNeO 982.6; found 983.8; Ή NMR (400 MHz, DMSO-Ob) 68.76 (dd, J = 20 4.7, 1.7 Hz, 1 H), 8.52 (d, J = 7.9 Hz, 1 H), 7.99 (d, J = 1.7 Hz, 1 H), 7.83 (d, J = 10.2 Hz, 2H), 7.74 - 7.58

(m, 3H), 7.53 (dd, J = 7.7, 4.8 Hz, 1H), 7.24 (t, J= 7.6 Hz, 1H), 7.12 (d, J= 7.6 Hz, 1H), 5.32 (d, J= 9.7 Hz, 2H), 4.33 - 4.20 (m, 4H), 4.03 (dd, J= 15.0, 8.6 Hz, 2H), 3.88 - 3.82 (m, 1 H), 3.63 (dq, J = 20.5, 10.3 Hz, 4H), 3.42 - 3.34 (m, 2H), 3.21 (s, 1H), 3.13 (d, J= 2.8 Hz, 3H), 2.87 (s, 2H), 2.83 - 2.72 (m, 2H), 2.69 - 2.62 (m, 1H), 2.21 (d, J= 22.6 Hz, 6H), 2.12 - 1.76 (m, 7H), 1.75 - 1.47 (m, 2H), 1.46 - 1.28 (m, 9H),

25 0.99 - 0.89 (m, 6H), 0.79 - 0.71 (m, 6H), 0.52 (s, 3H).

875

SUBSTITUTE SHEET (RULE 26) Example A341. Synthesis of (3S)-1 -acryloyl-N- ((2S)-1 -(((6 3 ¾4S)-2 5 -(dlf luoromethyl)-! '-ethyl- 1 z -(2-((S)-1 -methoxyethyl)pyrldln-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 f 6 4 ,6 5 ,6 6 -hexahydro-1 1 H· 8- oxa-1 (5,3)-indola-6(1 ,3)-pyrklazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amir»o)-3-methyl-1 - oxobutan-2-yl)-N-methylpyrrolldlne-3-carboxamlde

Step 1. To a mixture of 3-bromo-5-iodobenzaldehyde (4.34 g, 14.0 mmol) in DCM at 0 °C under an atmosphere of Nz was added BAST (6.8 g, 30.7 mmol) and EtOH (129 mg, 2.8 mmol) dropwise. The mixture was heated with microwave heating at 27 °C for 14 h. HzO (500 mL) was added and the mixture was extracted with DCM (200 mL x 3), the combined organic layers were concentrated under reduced

10 pressure and the residue was purified by silica gel column chromatography to give 1 -bromo-3- (difluoromethyl)-5-iodobenzene (3.2 g, 65% yield) as a solid. 1 H NMR (300 MHz, DMSO-cfe) δ 8.16 (p, J = 1.2 Hz, 1 H), 7.94 (p, J = 1.3 Hz, 1 H), 7.81 (p, J = 1.3 Hz, 1 H), 7.00 (t, J = 55.3 Hz, 1 H).

Step 2. A mixture of Zn (2.28 g, 34.8 mmol) and I2 (442 mg, 1.74 mmol) in DMF (20 mL) under an atmosphere of Ar was stirred at 50 °C for 0.5 h. To this mixture was added a solution of methyl (methyl

15 (fl)-2-((tert-butoxycarbonyl)amino)-3-iodopropanoate (2.39 g, 7.25 mmol) in DMF (20 mL) and the mixture was stirred at 50 °C for 2 h. After cooling, the mixture was added to 1 -bromo-3-(difluoromethyl)-5- iodobenzene (2.90 g, 8.7 mmol), Pdz(dba)3 (239 mg, 0.26 mmol) and tri-2-furylphosphine (162 mg, 0.7 mmol) in DMF (20 mL). The mixture was heated to 70 °C and stirred for 2 h, then H 2 O (200mL) was added and the mixture extracted with EtOAc (200 mL x 3). The combined organic layers were

20 concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give methyl (S)-3-(3-bromo-5-(difluoromethyl)phenyl)-2-((fert-butoxycarb onyl)amino)propanoate (560 mg, 19% yield) as a solid. 1 H NMR (300 MHz, DMSO-db) δ 7.65 (d, J= 10.0 Hz, 2H), 7.47 (s, 1H), 7.36 (d, J= 8.4 Hz, 1H), 7.00 (t, J = 55.6 Hz, 1H), 4.25 (td, J= 9.6, 4.7 Hz, 1H), 3.64 (s, 3H), 3.11 (dd, J =

13.6, 4.9 Hz, 1H), 3.00 - 2.80 (m, 1H), 1.32 (s, 9H).

876

SUBSTITUTE SHEET (RULE 26) Step 3. To a mixture of methyl (S)-3-(3-bromo-5-(difluoromethyl)phenyl)-2-((tert- butoxycarbonyl)amino)propanoate (650 mg, 1 .6 mmol) in THF (1.5 mL) at 0 °C under an atmosphere of N 2 was added LIOH (114 mg, 4.8 mmol) in H 2 O (1.50 mL). The mixture was stirred at 0 °C for 1 h, then acidified to pH 5 with 1 M HCI. The mixture was extracted with DCM / MeOH (10/1 ) (100 mL x 3) and the 5 combined organic layers were dried over anhydrous NapSO* and filtered. The filtrate was concentrated under reduced pressure to give (S)-3-(3-bromo-5-(difluoromethyl)phenyl)-2-((tert- butoxycarbonyl)amino)propanoic acid (500 mg), which was used directly in the next step without further purification. LCMS (ESI): m/z [M+H] calc’d for CisHieBrFzNO* 393.0; found 392.1.

Step 4. To a mixture of methyl (3S)-1 ,2-diazinane-3-carboxylate (475 mg, 3.3 mmol) in DCM (10 10 mL) at 0 °C under an atmosphere of Na were added N- methylmorpholine (3.34 g, 33.0 mmol) and (S)-3- (3-bromo-5-(difiuoromethyl)phenyl)-2-((fe/t-butoxycarbonyl)a mino)propanoic acid (650 mg, 1.7 mmol) and HOBt (45 mg, 0.33 mmol) and EDCI (632 mg, 3.3 mmol). The mixture was warmed to room temperature and stirred for 16 h, then diluted with DCM (100 mL) and HzO. The organic and aqueous layer was separated and the aqueous layer was extracted with DCM (100 mL x 3). The combined organic layers 15 were dried over anhydrous NaaSO* and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give methyl (S)-1-((S)-3-(3-bromo-5- (difluoromethyl)phenyl)-2-((te/t-butoxycarbonyl)amino)propan oyl)hexahydropyridazine-3-carboxylate (510 mg, 56% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for CaiHaeBrFaNaOs 519.1 ; found 520.3.

Step 5. To a mixture of 4,4,5,5-tetramethyl-2-(tetramethyl-1 ,3,2-dioxaborolan-2-yl)-1 ,3,2- 20 dioxaborolane (488 mg, 1.92 mmol) and methyl (S)-1-((S)-3-(3-bromo-5-(difluoromethyl)phenyl)-2-((fert- butoxycarbonyl)amino)propanoyl)hexahydropyridazine-3-carboxy late (500 mg, 0.96 mmol) in 1 ,4-dioxane (5 mL) was added Pd(dppf)Cl 2 (70 mg, 0.07 mmol) and KOAc (236 mg, 2.4 mmol) in portions. The mixture was heated to 90 °C and stirred for 4 h then diluted with H 2 O (100 mL). The mixture was extracted with DCM (100 mL x 3) and the combined organic layers were dried over anhydrous Na 2 SO 4 25 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give methyl (S)-1-((S)-2-((fert-butoxycarbonyl)amino)-3-(3-(difluorometh yl)- 5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)propanoyl)hexahydropyridazine- 3-carboxylate (423 mg, 73% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for C27H40BF2N3O7567.3; found 568.2.

Step 6. To a mixture of methyl (S)-1 -((S)-2-((te/t-butoxycarbonyl)amino)-3-(3-(difluoromethyl)-5 - 30 (4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)propanoyl)hexahydropyridazine- 3-carboxylate (260 mg, 0.47 mmol), (S)-3-(5-bromo-1 -ethyl-2-(2-(1 -methoxyethyl)pyridin-3-yl)-1 H-indol-3-yl)-2,2- dimethylpropan-1 -ol and Pd(dppf)Cl 2 (34 mg, 0.05 mmol) in 1 ,4-dioxane (3 mL) and H 2 O (0.6 mL) was added K2CO3 (163 mg, 1.12 mmol). The mixture was heated to 60 °C and stirred for 16 h, then diluted with H 2 O (100 mL) and extracted with DCM (100 mL x 3). The combined organic layers were dried over 35 anhydrous Na2SO< and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give methyl (S)-1 -((S)-2-((tert- butoxycarbonyl)amino)-3-(3-(difluoromethyl)-5-(1-ethyl-3-(3- hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1- methoxyethyl)pyridin-3-yl)-1 H-indol-5-yl)phenyl)propanoyl)hexahydropyridazine-3-carboxyl ate (350 mg, 78% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C44H57F2N5O7805.4; found 806.6.

40 Step 7. To a mixture of methyl (S)-1 -((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(difluoromethyl)-5 - (1 -ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1 -methoxyethyl)pyridin-3-yl)-1 H-indol-5- yl)phenyl)propanoyl)hexahydropyridazine-3-carboxylate (350 mg, 0.43 mmol) in THF (2.8 mL) at 0 °C

877

SUBSTITUTE SHEET (RULE 26) was added LiOH H 2 O (54 mg, 1.3 mmol) in H 2 O (0.7 mL). The mixture was warmed to room temperature and stirred for 2 h, then acidified to pH 5 with 1 M HCI and extracted with EtOAc (50 mL x 3). The combined organic layers were dried over anhydrous NaaSO*, filtered and the filtrate was concentrated under reduced pressure to give (S)-1 -((S)-2-((fert-butoxycarbonyl)amino)-3-(3-(difluoromethyl)-5 -(1 -ethyl- 5 3-(3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1 -methoxyethyl)pyridin-3-yl)-1 H- indol-5- yl)phenyl)propanoyl)hexahydropyridazine-3-carboxylic acid (356 mg) was used directly in the next step without further purification. LCMS (ESI): m/z [M+H] calc’d for C43H55F2N5O7791.4; found 792.6.

Step 8. To a mixture of S)-1-((S)-2-((fert-butoxycarbonyl)amino)-3-(3-(difluoromethy l)-5-(1-ethyl- 3-(3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)py ridin-3-yl)-1 H- indol-5- 10 yl)phenyl)propanoyl)hexahydropyridazine-3-carboxylic acid (356 mg, 0.45 mmol) and DIPEA (1.74 g, 13.5 mmol) in DCM were added EDCI (2.41 g, 12.6 mmol) and HOBt (304 mg, 2.3 mmol). The mixture was stirred for 16 h then H 2 O was added and the mixture extracted with EtOAc (200 mL x 3). The combined organic layers were washed with brine (50 mL x 4), dried over anhydrous NaaSO* and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column 15 chromatography to give fert-butyl ((6 3 S,4S)-2 5 -(difluoromethyl)-1 1 -ethyl- 1 2 -(2-((S)-1 -methoxyethyl)pyridin- 3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (202 mg, 51% yield). LCMS (ESI): m/z [M+H] calc’d for C43H53F2N5O6773.4; found 774.6.

Step 9. To a mixture of fert-butyl ((6 3 S,4S)-2 5 -(difluoromethyl)-1 1 -ethyl-1 2 -(2-((S)-1 - 20 methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-

6(1 ,3)-pyridazina-2(1 ,3)-benzenacyctoundecaphane-4-yl)carbamate (202 mg, 0.26 mmol) in DCM (2 mL) at 0 °C was added TFA (1.0 mL) dropwise. The mixture was stirred at 0 °C for 1.5 h, then concentrated under reduced pressure and dried azeotropically with toluene (3 mL x 3) to give (6 3 S,4S)-4-amino-2 5 - (difluoromethyl)-l 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - 25 hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-5,7-dione, which was used directly in the next without further purification. LCMS (ESI): m/z [M+H] calc'd for C38H45F2NSO4 673.3; found 674.5.

Step 10. To a mixture of (6 3 S,4S)-4-amino-2 5 -(difluoromethyl)-1 '-ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)- 30 pyridazina-2(1 ,3)-benzenacycloundecaphane-5,7-dione (202 mg, 0.3 mmol) and (2S)-2-[(fert- butoxycarbonyl)(methyl)amino]-3-methylbutanoic acid (139 mg, 0.6 mmol) in THF under an atmosphere of Ar were added DIPEA (581 mg, 4.5 mmol), EDCI (86 mg, 0.45 mmol) and HOBt (61 mg, 0.45 mmol). The mixture was stirred for 16 h, then H 2 O (100 mL) added and the mixture extracted with EtOAc (200 mL x 3). The combined organic layers were washed with brine (50 mL x 3), dried over anhydrous NaaS04 and 35 filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give fert-butyl ((2S)-1 -(((6 3 S,4S)-2 s -(difluoromethyl)-1 1 -ethyl-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 ' H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamate (135 mg, 46% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C49H64F2N6O7 40 886.5; found 887.6.

Step 11. To a mixture of fert-butyl ((2S)-1 -(((6 3 S,4S)-2 s -(difluoromethyl)-1 1 -ethyl-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 ' H- 8-oxa-1 (5,3)-indola-

878

SUBSTITUTE SHEET (RULE 26) 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamate (130 mg, 0.15 mmol) in DCM at 0 °C under an atmosphere of Na was added TFA (1.0 mL) dropwise. The mixture was stirred at 0 °C for 1.5 h, then concentrated under reduced pressure and dried azeotropically with toluene (3 mLx 3) to give (2S)-N-((6 3 S,4S)-2 5 -(difluoromethyl)-1 1 -ethyl-1 2 -(2- 5 ((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)bu tanamide (130 mg), which was used directly in the next step without further purification. LCMS (ESI): m/z [M+H] calc’d for C44H56F2N6O5786.4; found 787.6.

Step 12. To a mixture of (2S)-N-((6 3 S,4S)-2 5 -(difluoromethyl)-1 '-ethyl-1 2 -(2-((S)-1- 10 methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-

6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)bu tanamide (130 mg, 0.17 mmol) and (3S)-1-(prop-2-enoyl)pyrrolidine-3-carboxylic acid (56 mg, 0.33 mmol) in MeCN (1.5 mL) at 0 °C under an atmosphere of Na were added DIPEA (427 mg, 3.3 mmol) and GIF (69 mg, 0.25 mmol). The mixture was stirred at 0 °C for 1 h, then H 2 O (100 mL) was added and the mixture extracted with 15 EtOAc (200 mL x 3). The combined organic layers were washed with brine (50 mL x 3), dried over anhydrous NaaSO* and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by preparative-HPLC to give (3S)-1-acryloyl-N-((2S)-1-(((6 3 S,4S)-2 5 -(difluoromethyl)-1 1 -ethyl- 1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa- 1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)- 20 N-methylpyrrolidine-3-carboxamide (58 mg, 36% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for

CsaHesFaNyOy 937.4; found 938.1 ; 1 H NMR (400 MHz, DMSO-db) 68.78 (dd, J = 4.8, 1.7 Hz, 1 H), 8.43 - 8.21 (m, 1H), 8.02 (s, 2H), 7.93 - 7.81 (m, 2H), 7.76 (dd, J= 9.3, 3.9 Hz, 1H), 7.66 (d, J= 8.7 Hz, 1H),

7.56 (dd, J = 7.7, 4.8 Hz, 1 H), 7.34 (d, J = 5.4 Hz, 1 H), 7.20 - 6.86 (m, 1 H), 6.80 - 6.40 (m, 1 H), 6.15 (ddt, J= 16.8, 4.9, 2.4 Hz, 1H), 5.90 - 5.60 (m, 1H), 5.59 - 5.19 (m, 2H), 4.71 (dd, J= 10.7, 3.1 Hz, 1H), 4.40 - 25 4.17 (m, 3H), 4.12 - 3.90 (m, 3H), 3.85 - 3.71 (m, 1H), 3.61 (tdd, J = 23.4, 9.9, 4.3 Hz, 6H), 3.40 - 3.30 (m,

2H), 3.11 (d, J= 6.8 Hz, 3H), 3.08 - 2.90 (m, 2H), 2.87 (s, 2H), 2.84 (s, 3H), 2.69 - 2.30 (d, J= 16.5 Hz, 1H), 2.30 - 1.79 (m, 5H), 1.75 - 1.45 (m, 2H), 1.40 (d, J= 6.1 Hz, 3H), 1.05 - 0.85 (m, 6H), 0.85 - 0.66 (m, 6H), 0.57 (d, J= 11.8 Hz, 3H).

879

SUBSTITUTE SHEET (RULE 26) Example A741. Synthesis of (3S)-1-acryloyl-AF((2S)-1-(((2 3 S;6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)pyrldin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 Wt-oxa-1 (5,3 y indola-6(1 ,3)-pyridazina-2(1 ,3)-plperldlnacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yO-N-methylpyrrolidine-3-carboxamide

Step 1. A solution of tert-butyl (3fl)-3-(hydroxymethyl) piperidine-1 -carboxylate (10 g, 46.45 mmol) in DCM (200 mL) at 0 °C, was added PPha (15.8 g, 60.4 mmol), Imidazole (4.7 g, 69.7 mmol) and I2 (14.1 g, 55.74 mmol). The reaction suspension was stirred at 20 °C for 17 h, then concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford tert-butyl (3 R)- 10 3-(iodomethyl) piperidine-1 -carboxylate (10 g, 66% yield) as oil. LCMS (ESI): m/z [M+H] calc'd for C11H20INO2325.1 ; found no mass.

Step 2. To a mixture of 3-isopropyl-2,5-dimethoxy-3,6-dihydropyrazine (10.8 g, 58.9 mmol) in THF (150 mL) at -60 "C under an atmosphere of N 2 was added n-BuLi (47 mL, 2.5 M in hexane, 117.7 mmol) dropwise. The mixture was warmed to 0 °C and was stirred for 2 h, then re-cooled to -60 °C, and a solution 15 of tert-butyl (3fl)-3-(iodomethyl)piperidin-1 -yl formate (9.60 g, 29.4 mmol) in THF (50 mL) was slowly added dropwise. The mixture was stirred at -60 °C for 2 h then warmed to room temperature and stirred for 2 h. Saturated NHACI (150 mL) was slowly added and the mixture extracted with EtOAc (150 mL x 2). The combined organic layers were washed with brine (200 mL), dried over anhydrous Na2SO* and filtered. The filtrate was reduced under reduced pressure and the residue was purified by silica gel column 20 chromatography to give tert-butyl (3S)-3-{[(2S)-5-isopropyl-3,6-dimethoxy-2,5-dihydropyrazin-2 - yl]methyl}piperidin-1 -yl formate (5.3 g, 46% yield) as a gum. LCMS (ESI): m/z [M+H] calc’d for C20H35N3O4 381.5; found 382.3.

Step 3. A mixture of tert-butyl (3S)-3-{[(2S)-5-isopropyl-3,6-dimethoxy-2,5-dihydropyrazin-2 - yl]methyl}piperidin-1 -yl formate (5.30 g, 13.9 mol) in MeCN (4 mL) was added 1M HCI (27.7 mL, 27.7 25 mmol) dropwise. The mixture was stirred for 2 h, then saturated NaHCOa until ~pH 7-8, then extracted with DCM (30 mL x 2). The combined organic layers was dried over anhydrous Na 2 SO 4 , filtered and

880

SUBSTITUTE SHEET (RULE 26) concentrated under reduced pressure to give methyl (Sj-tert- butyl 3-((S)-2-amino-3-methoxy-3- oxopropyl)piperidine-1-carboxylate (4.3 g, 95% yield) as an oil, which was used in next step without further purification. LCMS (ESI): m/z [M+H] calc’d for CuHasNaCU 286.2; found 287.3.

Step 4. To a mixture of methyl (S)-fert-butyl 3-((S)-2-amino-3-methoxy-3-oxopropyl)piperidine-1- 5 carboxylate (4.30 g, 15.0 mmol) in EtOAc (30 mL) and H 2 O (20 mL) at -10 °C was added NaHCOa (3.77 g, 44.88 mmol). The mixture was stirred at -10 °C for 10 min, then a solution of benzyl chloroformate (3.83 g, 22.44 mmol) was added dropwise. The mixture was warmed to 0 °C and stirred for 1 h, then H 2 O (50 mL) was added and the mixture extracted with EtOAc (50 mL x 2). The combined organic layers were dried over anhydrous NaaSO*, filtered, the filtrate was concentrated under reduced pressure to give tert- 10 butyl (3S)-3-[(2S)-2-{[(benzyloxy)carbonyl] amino}-3-methoxy-3-oxopropyl]piperidine-1 -carboxylate (4.0 g, 60% yield) as a gum. LCMS (ESI): m/z [M-Boc+H] calc'd for C17H24N 2 O4320.2; found 321.3.

Step 5. To a mixture of fert-butyl (3S)-3-[(2S)-2-{[(benzyloxy)carbonyl] amino}-3-methoxy-3- oxopropyl]piperidine-1 -carboxylate (1.0 g, 2.38 mmol) in EtOAc (8 mL) was added 2M HCI in EtOAc (11.9 mL, 23.8 mmol). The mixture was stirred for 2 h, then saturated NaHCOa added until ~pH 8-9, and the 15 mixture extracted with DCM (30 mL x 3). The combined organic layer was dried over anhydrous NaaS04, filtered and concentrated under reduced pressure to give (2S)-2-{[(benzyloxy)carbonyl]amino)-3-[(3S)- piperidin-3-yl]propanoate (740 mg, 91% yield) as a gum. LCMS (ESI): m/z [M+H] calc’d for C17H24N 2 O4 320.2; found 321.2.

Step 6. To a mixture of (3-{3-[(fert-butyldiphenylsilyl)oxy]-2,2-dimethylpropyl}-1-e thyl-2-{2-[(1 S)- 20 1-methoxyethyl]pyridin-3-yl}indol-5-yl)boranediol (5.47 g, 8.43 mmol) and methyl (2S)-2-

{[(benzyloxy)carbonyl]amino}-3-[(3S)-piperidin-3-yl]propa noate (2.70 g, 8.43 mmol) in DCM (70mL) was added Cu(OAc)a (6.06 g, 16.86 mmol) and pyridine (2.0 g , 25.3 mmol). The mixture was stirred under an atmosphere of O2 for 48 h, then diluted with DCM (200 mL) and washed with H 2 O (150 mL x 2). The organic layer was dried over anhydrous Na 2 SO 4 , filtered, concentrated under reduced pressure and the 25 residue was purified by silica gel column chromatography to give methyl 2-{[(benzyloxy)carbonyl]amino}- 3-[(3S)-1 -(3-{3-[(iert-butyldiphenylsilyl)oxy]-2,2-dimethylpropyl}-1 -ethyl-2-{2-[(1 S)-1 -methoxyethyljpyridin- 3-yl}indol-5-yl)piperidin-3-yl]propanoate (3.6 g, 42% yield) as a solid. LCMS (ESI): m/z [M/2+H] calc’d for C 5 eH7oN40eSi 462.3; found 462.3.

Step 7. To a mixture of methyl 2-{[(benzyloxy)carbonyl]amino}-3-[(3S)-1-(3-{3-[(terf- 30 butyldiphenylsilyl)oxy]-2,2-dimethylpropyl}-1 -ethyl-2-{2-[(1 S)-1 -methoxyethyl]pyridin-3-yl}indol-5- yl)piperidin-3-yl]propanoate (3.60 g, 3.57 mmol) in THE (60 mL) and H 2 O (30 mL) was added LiOH (342 mg, 14.28 mmol). The mixture was stirred for 2 h, then diluted with H 2 O (150 mL), then 1 M HCI was added slowly until ~pH 3-4 and the mixture extracted with EtOAc (200 mL x 3). The combined organic layers were dried over anhydrous Na 2 SO 4 , filtered and the filtrate concentrated under reduced pressure to 35 give 2-{[(benzyloxy)carbonyl]amino}-3-[(3S)-1 -(3-{3-[(fert-butyldiphenylsilyl)oxy]-2,2-dimethylpropyl}-1 - ethyl-2-{2-[(1S)-1-methoxyethyl]pyridin-3-yl}indol-5-yl)pipe ridin-3-yl]propanoic acid (3.3 g, 85% yield) as a solid, which was used directly in the next step without further purification. LCMS (ESI): m/z [M/2+H] calc'd for C 55 HeeN40eSi 455.3; found 455.3.

Step 8. To a mixture of methyl 2-{[(benzyloxy)carbonyl]amino}-3-[(3S)-1-(3-{3-[(tert- 40 butyldiphenylsilyl)oxy]-2,2-dimethylpropyl}-1 -ethyl-2-{2-[(1 S)-1 -methoxyethyl]pyridin-3-yl}indol-5- yl)piperidin-3-yl]propanoate (3.30 g, 2.91 mmol) in DMF (40 mL) was added methyl (3S)-1 ,2-diazinane-3- carboxylate (0.42 g, 2.91 mmol), HATU (2.21 g, 5.82 mmol) and DIPEA (2.26 g, 17.46 mmol). The mixture

881

SUBSTITUTE SHEET (RULE 26) was stirred for 3 h, then poured into ice-H 2 O and extracted with EtOAc (120 mL x 2). The combined organic layers were washed with saturated NaHCOs (150 mL), brine (150 mL), dried over anhydrous NaaSO* and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give methyl (3S)-1-[(2S)-2-{[(benzyloxy)carbonyl]amino}-3-[(3S)-

5 1 -(3-{3-[(fert-butyldiphenylsilyl)oxy]-2,2-dimethylpropyl}-1 -ethyl-2-{2-[(1 S)-1 -methoxyethyl]pyridin-3- yl}indol-5-yl)piperidin-3-yl]propanoyl]-1 ,2-diazinane-3-carboxylate (2.9 g, 95% yield) as a gum. LCMS (ESI): m/z [M/2+H] calc’d for ΟβιΗτβΝβΟβΐ 518.3; found 518.3.

Step 9. To a mixture of methyl (3S)-1-[(2S)-2-{[(benzyloxy)carbonyl]amino}-3-[(3S)-1-(3-{3- [(fert- butyldiphenylsilyl)oxy]-2,2-dimethylpropyl}-1-ethyl-2-{2-[(1 S)-1-methoxyethyl]pyridin-3-yl)indol-5-

10 yl)piperidin-3-yl]propanoyl]-1 ,2-diazinane-3-carboxylate (1.70 g, 1.64 mmol) was added a mixture of 1 M TBAF in THE (19.68 mL, 19.68 mmol) and AcOH (1.18 g, 19.68 mmol). The reaction was heated to 60 °C and stirred for 22 h, then diluted with EtOAc (80 mL) and washed with saturated NaHCOs (80 mL), H 2 O (60 mL x 2) and brine (60 mL). The organic layer was dried over anhydrous NaaSO*, filtered, the filtrate was concentrated under reduced pressure and the residue was purified by preparative-HPLC to give

15 methyl (3S)-1 -[(2S)-2-{[(benzyloxy)carbonyl]amino}-3-[(3S)-1 -[1 -ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2- {2-[(1 S)-1-methoxyethyl]pyridin-3-yl}indol-5-yl]piperidin-3-yl]pro panoyl]-1 ,2-diazinane-3-carboxylate (1.0 g, 73% yield) as a solid. LCMS (ESI): m/z [M/2+H] calc'd for CAsHeoNeOy 399.2; found 399.4.

Step 10. To a mixture m methyl (3S)-1 -[(2S)-2-{[(benzyloxy)carbonyl]amino}-3-[(3S)-1 -[1 -ethyl-3- (3-hydroxy-2,2-dimethylpropyl)-2-{2-[(1 S)-1-methoxyethyl]pyridin-3-yl}indol-5-yl]piperidin-3-yl]pro panoyl]-

20 1 ,2-diazinane-3-carboxylate (1.0 g, 1.1 mmol) in 1 ,2-dichloroethane (10 mL) was added MesSnOH (1.42 g 7.84 mmol). The mixture was heated to 65 °C and stirred for 10 h, then filtered and the filtrate was concentrated under reduced pressure to give (3S)-1-[(2S)-2-{[(benzyloxy)carbonyl]amino}-3-[(3S)-1-[1- ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-{2-[(1 S)-1-methoxyethyl]pyridin-3-yl}indol-5-yl]piperidin-3- yl]propanoyl]-1 ,2-diazinane-3-carboxylic acid (1.0 g, 99% yield) as a gum. The product was used in the

25 next step without further purification. LCMS (ESI): m/z [M/2+H] calc'd for CuHseNeO? 392.2; found 392.3.

Step 11. To a mixture of (3S)-1-[(2S)-2-{[(benzyloxy)carbonyl]amino}-3-[(3S)-1-[1-eth yl-3-(3- hydroxy-2,2-dimethylpropyl)-2-{2-[(1 S)-1-methoxyethyl]pyridin-3-yl}indol-5-yl]piperidin-3-yl]pro panoyl]-1 ,2- diazinane-3-carboxylic acid (1.0 g, 1.1 mmol) in DCM (30 mL) at 0 °C was added HOST (1.51 g, 11.2 mmol), N-(3-dimethylaminopropyl)-/V-ethylcarbodiimide HCI (6.44 g, 33.6 mmol) and DIPEA (5.79 g, 44.8

30 mmol). The mixture was warmed to room temperature and stirred for 6 h, then diluted with H 2 O and extracted with EtOAc (100 mL x 3). The combined organic layers were dried over anhydrous Na 2 SO 4 , filtered, the filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give benzyl N-[(6S,8S,14S)-22-ethyl-21-{2-[(1 S)-1-methoxyethyl]pyridin-3-yl}- 18,18-dimethyl-9,15-dioxo-16-oxa-2,10,22,28-tetraazapentacyc lo [18.5.2.1 A {2,6}.1 A {10,

35 14}.0 A {23,27}]nonacosa-1(26),20,23(27),24-tetraen-8-yl]carba mate (340 mg, 36% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C^HseNeOe 383.2; found 383.3.

Step 12. A mixture of benzyl N-[(6S,8S,14S)-22-ethyl-21 -{2-[(1 S)-1 -methoxyethyl]pyridin-3-yl}- 18,18-dimethyl-9,15-dioxo-16-oxa-2,10,22,28-tetraazapentacyc lo [18.5.2.1 A {2,6}.1 A {10, 14}.0 A {23,27}]nonacosa-1(26),20,23(27),24-tetraen-8-yl]carba mate (250 mg, 0.33 mmol), Pd/C (100 mg)

40 and NhUCI (353 mg, 6.6 mmol) in MeOH (5 mL) was stirred under an atmosphere of H2 for 4 h. The mixture was filtered through Celite and the filtrate was concentrated under reduced pressure. The residue was dissolved in DCM (30 mL) and washed with saturated NaHCOs (20 mL), H 2 O (20 mL) and brine (20

882

SUBSTITUTE SHEET (RULE 26) mL). The organic layer was dried over Na2SC>4, filtered, and the filtrate concentrated under reduced pressure to give (6S,8S,14S)-8-amino-22-ethyl-21 -{2-[(1 S)-1-methoxyethyl]pyridin-3-yl}-18,18-dimethyl·

1 e-oxa-2,10,22,28-tetraazapentacyclo[18.5.2.1 A {2,6}.1 A {10,14}.0 A {23,27}]nonacosa-1 (2β),20,23(27),24- tetraene-9,15-dione, which was used in next step without further purification. LCMS (ESI): m/z [M+H]

5 calc'd for CaeHsoNeO* 631.4; found 631.4.

Step 13. To a mixture of (6S,8S,14S)-8-amino-22-ethyl-21-{2-[(1 S)-1-methoxyethyl]pyridin-3-yl>- 18,18-dimethyH6-oxa-2,10,22,28-tetraazapentacyclo[18.5.2.1 A {2,6}.1 A {10,14}.0 A {23,27}]nonacosa- 1(26), 20, 23(27), 24-tetraene-9,15-dione (300 mg, 0.48 mmol), (2S)-3-methyl-2-{N- methyl-1-[(3S)-1-(prop- 2-enoyl)pyrrolidin-3-yl]formamido}butanoic acid (136 mg, 0.48 mmol) and DIPEA (620 mg, 4.8 mmol) in

10 DMF (5 mL) at 0 °C was added HATU (183 mg, 0.48 mmol). The mixture was stirred at 0-5 °C for 1 h, then diluted with EtOAc (50 mL), washed with HzO (50 mL x 2), brine (50 mL), dried over N82S04 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give (2S)-N- [(6S,8S,14S)-22-ethyl-21-{2-[(1 S)-1-methoxyethyl]pyridin-3-yl}-18,18- dimethyl-9,15-dioxo-16-oxa-2,10,22,28-tetraazapentacyclo[18. 5.2.1 A {2,6}.1 A {10,14}.0 A {23,27}]nonacosa-

15 1 (26) ,20 ,23(27) ,24-tetraen-8-yl]-3-methyl-2-{N- methyl-1 -[(3S)-1 -(prop-2-enoyl)pyrrolidin-3- yl]fomnamido}butanamide (90 mg, 20% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for CsoHyoNeO? 895.5; found 895.4; Ή NMR (400 MHz, CDsOD) 68.71 (dd, J = 4.8, 1.5 Hz, 1 H), 7.86 (dd, J = 7.7, 1.5 Hz, 1H), 7.51 (dd, J = 7.7, 4.8 Hz, 1H), 7.36 (dd, J = 8.9, 1.9 Hz, 1H), 7.22 (d, J = 12.1 Hz, 1H), 7.09 (dd, J = 8.9, 1.9 Hz, 1H), 6.59 (dt, J = 16.9, 9.9 Hz, 1H), 6.26 (ddd, J = 16.8, 5.0, 1.9 Hz, 1H), 5.80 - 5.67 (m, 1H),

20 5.59 - 5.46 (m, 1H), 4.93 (d, J = 12.4 Hz, 1H), 4.66 (dd, J = 11.1, 6.4 Hz, 1H), 4.45 (d, J = 12.6 Hz, 1H), 4.28 - 4.19 (m, 1H), 4.13 (dd, J= 14.5, 7.2 Hz, 1H), 4.02 - 3.87 (m, 1H), 3.87 - 3.36 (m, 11H), 3.16 (s, 2H), 3.10 (d, J = 3.4 Hz, 2H), 2.76 (dd, J = 26.9, 13.5 Hz, 3H), 2.61 (s, 1H), 2.35 - 1.97 (m, 5H), 1.78 (dd, J = 25.4, 22.1 Hz, 10H), 1.45 (d, J = 6.2 Hz, 3H), 1.04 (d, J = 6.2 Hz, 3H), 0.95 (dd, J = 6.5, 1.8 Hz, 3H), 0.83 (d, J = 6.6 Hz, 3H), 0.72 (d, J = 31.8 Hz, 6H).

25 Example A715. Synthesis of benzyl ((2 3 S,6 3 S,4S)-1 1 -ethyH 2 -(2-((S)-1-methoxyethyl)pyrldin- 3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 M-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(1,3)-piperldinacycloundecaphane-4-yl)carbamate

To a solution of ((2 3 S,6 3 S,4S)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-

30 dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- piperidinacycloundecaphane-5,7-dione (50 mg, 0.08 mmol), (fl)-2-(((1-(4-(dimethylamino)-4-methylpent-2- ynoyl)azetidin-3-yl)oxy)methyl)-3-methylbutanoic acid (26 mg, 0.08 mmol) and DIPEA (31 mg, 0.24mmol) in DMF (1 mL) at 0 °C, was added HATU (30 mg, 0.08 mmol). The reaction mixture was stirred at 0-5 °C for 1 h, then diluted with EtOAc (20 mL), washed with H 2 O (20 mL x 2) and brine (20 mL). The organic

35 phase was separated and dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give (2/?)-2-(((1 -(4-

883

SUBSTITUTE SHEET (RULE 26) (dimethylamino)-4-methylpent-2-ynoyl)azetidin-3-yl)oxy)methy l)-N-((2 3 S,6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-piperidinacycloundecaphane-4-yl)-3-methylbutanamide as solid. 1 H NMR (400 MHz, CDaOD) 68.71 (d, J = 4.7 Hz, 1 H), 8.24 (s, 1 H), 8.10 (dd, J = 26.7, 7.8 Hz, 1 H), 7.86 (d, J = 7.7 Hz,

5 1 H), 7.51 (dd, J = 7.7, 4.8 Hz, 1 H), 7.39 (dd, J = 8.9, 3.1 Hz, 1 H), 7.26 (d, J = 16.6 Hz, 1 H), 7.11 (d, J = 8.8 Hz, 1 H), 5.61 (s, 1 H), 4.50 - 4.28 (m, 3H), 4.27 - 4.07 (m, 3H), 3.98 (ddd, J = 25.6, 13.4, 5.1 Hz, 2H), 3.84 - 3.72 (m, 2H), 3.62 (dd, J = 10.7, 4.8 Hz, 2H), 3.55 (d, J = 7.1 Hz, 2H), 3.47 (d, J = 6.5 Hz, 2H), 3.16 (s, 3H), 3.03 - 2.91 (m, 1 H), 2.76 (dd, J = 28.7, 15.2 Hz, 3H), 2.62 (s, 1 H), 2.40 (t, J = 7.0 Hz, 3H), 2.33 (dd, J = 14.3, 5.0 Hz, 4H), 2.05 (d, J = 11.6 Hz, 1 H), 1.99 - 1.64 (m, 10H), 1.64 - 1.55 (m, 1 H), 1.51 - 1.42 (m,

10 6H), 1.37 (d, J = 12.3 Hz, 3H), 1.05 (s, 3H), 0.94 (ddd, J = 9.3, 6.7, 2.0 Hz, 6H), 0.76 (d, J = 3.8 Hz, 3H), 0.69 (s, 3H). LCMS (ESI): m/z [M+H] calc’d for CsaHyeNeO? 936.6; found 937.4.

Example A347. Synthesis of (2S)-A#-[(7¾13S)-21-ethyl-20-{2-[(1S)-1-methoxyethyllpyridi n-3- ylJ-17,17-dimethyl-8,14-dioxo-15-oxa-4-th la-9,21 ,25,27,28- 15 pentaazapentacyclo[17.5.2.1 A {2,5}.1 A {9,13}.0 A {22,26}]octacosa-1 (25),2,5(28),19,22(26),23-hexaen-

7-yl]-3-methyl-2-{N-methyl-1-[(3S)-1-(prop-2-enoyl)pyrrol idin-3-yl]formaml<io}butanamide

Step 1. To a mixture of (5-chloro-1 H- pyrrolo[3,2-b]pyridin-3-yl)methanol (3.5 g, 19 mmol), and ((1 -methoxy-2-methylprop-1 -en-1 -yl)oxy)trimethylsilane (6.7 g, 38 mmol) in THF (50 ml) at 0 °C was

20 added TMSOTf (3.8 g, 17 mmol) dropwise. The mixture was stirred at 0-5 °C for 2 h, then diluted with EtOAc (100 mL) and washed with saturated NaHCOs (50 mL) and brine (50 nriLx 2). The organic layer

884

SUBSTITUTE SHEET (RULE 26) was dried over Na 2 SO 4 , filtered and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give methyl 3-(5-chloro-1 H- pyrrolo[3,2-b]pyridin-3-yl)- 2,2-dimethylpropanoate (3.0 g, 59% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C13H15CIN 2 O2 266.1; found 267.1.

5 Step 2. To a mixture of methyl 3-(5-chloro-1 H- pyrrolo[3,2-b]pyridin-3-yl)-2,2-dimethylpropanoate (3.0 g, 11 mmol) in anhydrous THF (50 mL) at 0 °C was added AgOTf (4.3g, 17 mmol) and I2 (2.9 g, 11 mmol). The mixture was stirred at 0 °C for 2 h, then saturated NaaSOa (20 mL) and EtOAc (50 mL) added. The mixture was filtered and the filtrate was washed with brine (50 mL), dried over NazSO* and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column

10 chromatography to give methyl 3-(5-chloro-2-iodo-1 H- pyrrolo[3,2-b]pyridin-3-yl)-2,2-dimethylpropanoate (2.3 g, 52% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for C13H15CIIN 2 O2392.0; found 393.0.

Step 3. To a mixture of methyl 3-(5-chloro-2-iodo-1 H-pyrrolo[3,2-b]pyridin-3-yl)-2,2- dimethylpropanoate (2.3 g, 5.9 mmol) and 2-(2-(2-methoxyethyl)phenyl)-4,4,5,5-tetramethyl-1 ,3,2- dioxaborolane (1.6 g, 7.1 mmol) and K2CO3 (2.4 g, 18 mol) in 1 ,4-dioxane (25 mL) and H 2 O (5 mL) under

15 an atmosphere of N 2 was added Pd(dppf)Cl 2 .DCM (480 mg, 0.59 mmol). The mixture was heated to 70 °C and for 4 h, then diluted with EtOAc (200 mL) and washed with brine (25 mL), dried over NazSO* and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give methyl (S)-3-(5-chloro-2-(2-(1 -methoxyethyl)pyridin-3-yl)-1 H- pyrrolo[3,2-b]pyridin-3-yl)-2,2-dimethylpropanoate (2.0 g, 84% yield) as a solid. LCMS (ESI): m/z [M+H]

20 calc’d for C21H24CIN3O3401.2; found 402.2.

Step 4. A mixture of ethyl 3-(5-chloro-2-(2-((S)-1 -methoxyethyl)pyridin-3-yl)-1 H- pyrrolo[3,2- b]pyridin-3-yl)-2-methylpropanoate (2.0 g, 5.0 mmol), CS2CO3 (3.3 g, 10 mmol) and Etl (1.6 g, 10 mmol) in DMF (30 mL) was stirred for 10 h. The mixture was diluted with EtOAc (100 mL) and washed with brine (20 mL x 4), dried over NazS04 and filtered. The filtrate was concentrated under reduced pressure and

25 the residue was purified by silica gel column chromatography to give two diastereomers of methyl (S)-3- (5-chloro-1-ethyl-2-(2-(1-methoxyethyl)pyridin-3-yl)-1 H-pyrrolo[3,2-b]pyridin-3-yl)-2,2-dimethylpropanoate (0.7 g, 32% yield; 0.6 g, 28% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for C23H28CIN3O3429.2; found 430.2.

Step 5. To a mixture of methyl (S)-3-(5-chloro-1-ethyl-2-(2-(1-methoxyethyl)pyridin-3-yl)-1 H-

30 pyrrolo[3,2-ti]pyridin-3-yl)-2,2-dimethylpropanoate (1.9 g, 4.4 mmol) in anhydrous THF (20 mL) at 0 °C was added LiBhU (200 mg, 8.8 mmol). The mixture was heated to 60 °C and stirred for 4 h, then saturated NhUCI (20 mL) and EtOAc (50 mL) added. The aqueous and organic layers were separated and the organic layer was washed with brine (30 mL), dried over NapSO* and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography

35 to give (S)-3-(5-chloro-1 -ethyl-2-(2-(1 -methoxyethyl)pyridin-3-yl)-1 H- pyrrolo[3,2-b]pyridin-3-yl)-2,2- dimethylpropan-1 -ol (1.5 g, 85% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for C22H28CIN3O2401.2; found 402.2.

Step 6. To a mixture of (S)-3-(5-chloro-1-ethyl-2-(2-(1-methoxyethyl)pyridin-3-yl)-1 H- pyrrolo[3,2- b]pyridin-3-yl)-2,2-dimethylpropan-1-ol (550 mg, 1.37 mmol), (S)-(2-(2-((tert-butoxycarbonyl)amino)-3-

40 methoxy-3-oxopropyl)thiazol-4-yl)boronic acid (907.4 mg, 2.74 mmol, 2 eq) and K2CO3 (568 mg, 4.11 mmol) in 1 ,4-dioxane (25 mL) and H 2 O (5 mL) under an atmosphere of Nz was added Pd(dppf)Cl 2 .DCM (89 mg, 0.14 mmol). The mixture was heated to 70 °C and stirred for 4 h, then H 2 O (50 mL) added and

885

SUBSTITUTE SHEET (RULE 26) the mixture extracted with EtOAc (100 mL x 3). The combined organic layers were washed with brine (50 mL), dried over NaaSO* and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give methyl (S)-2-((tert- butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3-hydroxy-2,2-dimethy lpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3- 5 yl)-1 H-pyrrolo[3,2-b]pyridin-5-yl)thiazol-2-yl)propanoate (440 mg, 22% yield) as a solid, which was used directly in the next step. LCMS (ESI): m/z [M+H] calc’d for C 3 4H4 5 N 5 O 6 S 651.3; found 652.3.

Step 7. To a mixture of (2S)-methyl 2-((tert-butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3-hydroxy- 2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1 H-pyrrolo[3,2-b]pyridin-5-yl)thiazol-2- yl)propanoate (280 mg, 0.43 mmol) in MeOH (4 mL) was added a solution of LiOH (51 mg, 2.2 mmol) in 10 H 2 O (2 mL). The mixture was stirred for 5 h, then pH adjusted to ~3-4 by addition of 1 M HCI. The mixture was diluted with H 2 O (30 mL) and extracted with EtOAc (15 mL χ 3). The combined organic layers were washed with brine (10 mL), dried over anhydrous NaaS04, filtered and the filtrate was concentrated under reduced pressure to give (2S)-2-((te/i-butoxycarbonyl)amino)-3-(4-(1 -ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1 /fpyrrolo[3,2-b]pyridin-5-yl)thiazol-2-yl)propanoic 15 acid (280 mg) as solid, which was used directly in the next step without further purification. LCMS (ESI): m/z [M+H] calc’d for CaaH^NsOeS 637.3; found 638.3.

Step 8. To a mixture of (2S)-2-((fert-butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3-hydro xy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1 H-pyrrolo[3,2-b]pyridin-5-yl)thiazol-2-yl)propanoic acid (274 mg, 0.43 mmol) and methyl (3S)-1 ,2-diazinane-3-carboxylate (280 mg, 0.64 mmol) in DMF (3 20 mL) at 0-5 °C was added a solution of HATU (245 mg, 0.64 mmol) and DIPEA (555 mg, 4.3 mmol) in DMF (2 mL). The mixture was stirred for 1 h, then diluted with EtOAc (20 mL) and H 2 O (20 mL). The aqueous and organic layers were partitioned and the organic layer was washed with H 2 O (20 mL x 3), brine (20 mL), dried over anhydrous NaaSO* and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give methyl (3S)-1-[(2S)-2- 25 {[(te/†-butoxy)carbonyl]amino}-3-{4-[1 -ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-{2-[(1 S)-1 - methoxyethyl]pyridin-3-yl}pyrrolo[3,2-b]pyridin-5-yl]-1,3-th iazol-2-yl}propanoyl]-1,2-diazinane-3- carboxylate (230 mg, 70% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for C39H53N7O7S 763.4; found 764.3.

Step 9. To a mixture of methyl (3S)-1-[(2S)-2-{[(tert-butoxy)carbonyl]amino}-3-{4-[1-ethyl- 3-(3-

30 hydroxy-2,2-dimethylpropyl)-2-{2-[(1S)-1-methoxyethyl]pyridi n-3-yl}pyrrolo[3,2-b]pyridin-5-yl]-1,3-thiazol- 2-yl}propanoyl]-1 ,2-diazinane-3-carboxylate (230 mg, 0.3 mmol) in DCE (3 mL) under an atmosphere of Na was added MeaSnOH (300 mg). The mixture was heated to 65 °C and stirred for 16 h, then concentrated under reduced pressure. The residue was diluted with EtOAc (20 mL), washed with H 2 O (20 mL) and brine (10 mL), dried over NaaSO* and filtered. The filtrate was concentrated under reduced

35 pressure to give (3S)-1-[(2S)-2-{[(tert-butoxy)carbonyl]amino}-3-[4-[1-ethyl- 3-(3-hydroxy-2,2- dimethylpropyl)-2-{2-[(1 S)-1 -methoxyethyl]pyridin-3-yl}pyrrolo[3,2-b]pyridin-5-yl]-1 ,3-thiazol-2- yl}propanoyl]-1 ,2-diazinane-3-carboxylic acid (200 mg) as a foam, which was used directly in the next step without further purification. LCMS (ESI): m/z [M+H] calc’d for CaeHsiNyOyS 749.4; found 750.3.

Step 10. To a mixture of (3S)-1-[(2S)-2-[[(te/i-butoxy)carbonyl]amino}-3-{4-[1-ethyl- 3-(3-hydroxy-

40 2,2-dimethylpropyl)-2-{2-[(1 S)-1-methoxyethyl]pyridin-3-yl}pyrrolo[3,2-ti]pyridin-5-yl]- 1,3-thiazol-2- yl}propanoyl]-1 ,2-diazinane-3-carboxylic acid (245 mg, 0.32 mmol) in DCM (50 mL) at 0-5 °C were added HOST (432 mg, 3.2 mmol), EDCI HCI (1.8 g, 9.6 mmol) and DIPEA (1.65 g, 12.8 mmol). The mixture was

886

SUBSTITUTE SHEET (RULE 26) warmed to room temperature and stirred for 16 h, then concentrated under reduced pressure. The residue was diluted with EtOAc (20 mL) and H 2 O (20 mL) and the aqueous and organic layers were partitioned. The organic layer was washed with H 2 O (30 mL x 3), brine (30 mL), dried over anhydrous NaaSO* and filtered. The filtrate was concentrated under reduced pressure and the residue was purified 5 by preparative-TLC to give tert-butyl ((6 3 S,4S,2)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-thiazola-1 (5,3)-pyrrolo[3,2-b]pyridina- 6(1 ,3)-pyridazinacycloundecaphane-4-yl)carbamate (100 mg, 43% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for Cs^NTOeS 731.4; found 732.3.

Step 11. A mixture of fert-butyl ((6 3 S,4S,Z)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10- 10 dimethyl-5, 7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-thiazola-1 (5,3)-pyrrolo[3,2-b]pyridina-

6(1 ,3)-pyridazinacycloundecaphane-4-yl)carbamate(80 mg, 0.11 mmol) in DCM (0.6 mL) and TEA (0.2 mL) was stirred for 1 h. The mixture was concentrated under reduced pressure to give (6 3 S,4S,2)-4- amino-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8- oxa-2(4,2)-thiazola-1 (5,3)-pyrrolo[3,2-b]pyridina-6(1 ,3)-pyridazinacycloundecaphane-5,7-dione (72 mg,

15 95% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C33H41N7O4S 631.3; found 632.3.

Step 12. To a mixture of (6 3 S,4S,2)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)- 10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 , 6 5 ,6 ® -hexahydro-1 1 H- 8-oxa-2(4,2)-thiazola-1 (5,3)-pyrrolo[3,2-b]pyridina-6(1 ,3)- pyridazinacycloundecaphane-5,7-dione (120 mg, 0.39 mmol) and DIPEA (335 mg, 2.6 mmol) in DMF (1 mL) at 0 °C was added HATU (60 mg, 0.16 mmol). The mixture was stirred at 0 °C for 1 h, then diluted 20 with H 2 O (110 mL) and extracted with EtOAc (80 mL x 2). The combined organic layers were washed with H 2 O (100 mL), brine (100 mL), dried over anhydrous Na 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by preparative-TLC to give (3S)-1 -acryloyl-N-((2S)- 1 -(((6 3 S,4S,Z)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 1 H- 8-oxa-2(4,2)-thiazola-1 (5,3)-pyrrolo[3,2-b]pyridina-6(1 ,3)-pyridazinacycloundecaphane-4- 25 yl)amino)-3-methyl-1 -oxobutan-2-yl)-N-methylpyrrolidine-3-carboxamide (1.8 mg, 2% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C47H61N9O7S 895.4; found 896.3; 1 H NMR (400 MHz, CDsOD) 68.72 (d, J= 4.5 Hz, 1H), 7.98 - 7.77 (m, 3H), 7.72 (dd, J= 12.0, 8.6 Hz, 1H), 7.54 (dd, J= 7.6, 4.8 Hz, 1 H),

6.67 - 6.54 (m, 1 H), 6.26 (m, 1 H), 5.79 - 5.58 (m, 2H), 4.83 - 4.75 (m, 1 H), 4.39 - 4.16 (m, 4H), 4.02 (dd, J = 28.0, 10.6 Hz, 2H), 3.89 - 3.65 (m, 6H), 3.50 (m, 4H), 3.34 (d, J= 6.2 Hz, 3H), 3.12 (d, J= 4.0 Hz, 2H), 30 3.00 (s, 1 H), 2.73 (m, 1 H), 2.48 - 2.37 (m, 1 H), 2.31 - 2.07 (m, 4H), 1.88 (d, J = 11.2 Hz, 1 H), 1.71 (d, J =

12.8 Hz, 1H), 1 .44 (m, 7H), 0.97 (dd, J= 6.2, 4.4 Hz, 3H), 0.92 - 0.84 (m, 8H), 0.41 (d, J= 6.2 Hz, 3H).

887

SUBSTITUTE SHEET (RULE 26) Example 647. Synthesis of 1 -(4-(dimethylamlno)-4-methylpent-2-ynoyl)-N- ((2S)-1 - (((2 Z S,6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyrkfln-3-yl)-10,10-dlmethyl-5 f 7-dloxo- 6 1 ,6 2 ,6 3 ,6 4 f 6 6 ,6 e -hexahydro-1 1 H-8-oxa-2(4,2)-morpholina-1 (5,3)-indola-6(1 ,3)- pyridazlnacycloundecaphane-4-yl)amlno)-3-methyH-oxobutan-2-y l)-4-fluoro-N-methylplperldine- 5 4-carboxamlde

UsJ*

(COCfc. El j N “ O

Boc ^

Et j N. T3P O

N N I s

DMF DMF

HO

TFA 'Ν' I

N,

DCM

CbzHN O^O r°H X z- MeO^D

FXOKOCHala OyO EMPO.TCCA ^^N S.S-Et-Duphoe-Rh

T IMG HCbz H? -Ai HQ

N aHCQs.EtOAc MeCN M EtOAc i ^NHCbz

N cT B O JO oOH

Bo oer BocN

HO

X ! NHCbz

,OH >T8S HOyO

HN A^o

MeQ RuPhoa, Po(dppOC: Cs£¾R uPhotP^ 2 >TBS ^NHCbz

T8SCI , LOH Mi I

.Br .B Me0

N· f

DCM dioxane THF/H5O O

I Ό

<<

IntermedM» 1 k k

_ΥϊΥ 0 H

IBWJ. Z^NHCbz

HATU. DiPEA 'NHCbz

TBAF/AcOH LOH x» x

MeQ j—- i. Mi ? MeQ

DMF THF THF/H j O

N. N· O N· O k k k

‘N 4 I

Vr H H I r ^NHCte > NH 2

Mi !

HOBt, EDCI, DIPEA M< Pd/C. NH4OAC HATU, DIPEA

DCM O MeOH o DMF k k i N' i

H N N «x

? H a O hr k

Step 1. A mixture of 1-[(fert-butoxy)carbonyl]-4-fluoropiperidine-4-carboxylic acid (2.0 g, 8.1 mmol) in DCM (20 mL) was added oxalic dichloride (1.34 g, 10.5 mmol) and DMF (30 mg, 0.4 mmol). The resulting solution was stirred at room temperature for 1 h. EtsN (3.2 g, 3.2 mmol) and (2S)-3-methyl-2-

10 (methylamino)butanoic acid (1.25 g, 9.5 mmol) were added and the mixture was stirred at room temperature for 1 h. H 2 O (100 mL) was added and the mixture was extracted with EtOAc (50 mL x 3). The combined organic layers were concentrated under reduced pressure and the residue was purified by

888

SUBSTITUTE SHEET (RULE 26) silica gel column chromatography to give tert- butyl (S)-4-((1 -(fert-butoxy)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamoyl)-4-fluoropiperidine-1-carboxylate (1.34 g, 45% yield) as a solid. LCMS (ESI): m/z [M+Na] calc'd for C2iH 3 7FN 2 0 5 Na 439.3; found 439.3.

Step 2. A mixture of fert-butyl (S)-4-((1-(fert-butoxy)-3-methyl-1-oxobutan-2- 5 yl)(methyl)carbamoyl)-4-fluoropiperidine-1-carboxylate (290 mg, 0.70 mmol) in DCM (4 mL) and TFA (2 mL) was stirred at room temperature for 2 h, then concentrated under reduced pressure to give N-{ 4- fluoropiperidine-4-carbonyl)-N-methyl-L-valine, which was used directly in the next step without further purification. LCMS (ESI): m/z [M+H] calc’d for C12H21FN 2 O3260.2; found 261.2.

Step 3. To a solution of the fert-butyl N-(4-fluoropiperidine-4-carbonyl)-N-methyl-L-valinate (1.7 10 g, 5.3 mmol), sodium 4-(dimethylamino)-4-methylpent-2-ynoate (1.67 g, 9.4 mmol) and EfaN (2.73 g, 36.9 mmol) in DMF (20 mL) stirred at 5 °C was added T3P (4.11 g, 10.7 mmol, 50wt% in EtOAc). The reaction mixture was stirred at 5 °C for 1 h. The resulting mixture was quenched with H 2 O (100 mL) and extracted with EtOAc (50 mL x 3). The combined organic layers were concentrated and purified by silica gel column chromatography to give fert-butyl N-(1-(4-(dimethylamino)-4-methylpent-2-ynoyl)-4-fluoropiperi dine-4- 15 carbonyl)-N-methyl-L-valinate (1.6 g, 74.0% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for C24H40FN3O4453.3; found 454.2.

Step 4. To a solution of fert-butyl N-(1-(4-(dimethylamino)-4-methylpent-2-ynoyl)-4- fluoropiperidine-4-carbonyl)-N-methyl-L-valinate (50 mg, 0.11 mmol) in DCM (2 mL) was added TFA (1 mL). The reaction mixture was stirred at 20 °C for 2 h, then concentrated under reduced pressure to 20 afford crude ΛΑ(1 -(4-(dimethylamino)-4-methylpent-2-ynoyl)-4-fluoropiperidine -4-carbonyl)-N-methyl-L- valine. It was used for the next step directly without further purification. LCMS (ESI): m/z [M+H] calc'd for C20H32FN3O4397.2; found 398.3.

Step 5. To a solution of fert-butyl (2fl)-2-(hydroxymethyl)morpholin-4-yl formate (50 g, 230 mmol) in EtOAc (1 L) was added TEMPO (715 mg, 4.6 mmol) and NaHCOs (58 g, 690 mmol) at 20 °C. The 25 mixture was cooled to -50 °C, then TCCA (56 g, 241 mmol) in EtOAc (100 mL) was added dropwise over 30 min. The reaction mixture was warmed to 5 °C for 2 h, then quenched with 10% NaaSaOs (200 mL) and stirred for 20 min. The resulting mixture was filtered and the organic phase was separated from filtrate. The aqueous phase was extracted with EtOAc (100 mL x 2). The combined organic layers were washed with H 2 O (100 mL) and brine (100 mL), and dried over anhydrous Na 2 SO 4 . The organic layer was 30 concentrated under reduced pressure to afford fert-butyl (2fl)-2-formylmorpholin-4-yl formate (50 g, crude) as an oil.

Step 6. To a solution of fert-butyl (2fl)-2-formylmorpholin-4-yl formate (49 g, 153 mmol) and methyl 2-{[(benzyloxy)carbonyl]amino)-2-(dimethoxyphosphoryl)acetat e (60 g, 183 mmol) in CAN (300 mL) was added tetramethylguanidine (35 g, 306 mmol) at 0~10 °C. The reaction mixture was stirred at 10

35 °C for 30 min then warmed to 20 °C for 2 h. The reaction mixture was diluted with DCM (200 mL) and washed with Citric acid (10%, 200 mL) and 10% NaHCOs aqueous solution (200 mL). The organic phase was concentrated under reduced pressure, and purified by silica gel column chromatography to afford fert-butyl (S,Z)-2-(2-(((benzyloxy)carbonyl)amino)-3-methoxy-3-oxoprop- 1 -en-1 -yl)morpholine-4- carboxylate (36 g, 90% yield) as solid. LCMS (ESI): m/z [M+Na] calc’d for C21H28N 2 O4420.2; found: 443.1

40 Step 7. To a solution of fert-butyl (S,Z)-2-(2-(((benzyloxy)carbonyl)amino)-3-methoxy-3-oxoprop- 1-en-1-yl)morpholine-4-carboxylate (49 g, 0.12 mol) in MeOH (500 mL) was added (S,S)-Et-DUPHOS-Rh (500 mg, 0.7 mmol). The mixture was stirred at 25 °C under an Ha (60 psi) atmosphere for 48 h. The

889

SUBSTITUTE SHEET (RULE 26) reaction was concentrated and purified by chromatography to give tert-butyl (S)-2-((S)-2- (((benzyloxy)carbonyl)amino)-3-methoxy-3-oxopropyl)morpholin e-4-carboxylate (44 g, 89.8% yield) as solid. LCMS (ESI): m/z [M+Na] calc’d for C21H30N 2 O7422.2; found: 445.2.

Step 8. To a stirred solution of fert-butyl (S)-2-((S)-2-(((benzyloxy)carbonyl)amino)-3-methoxy-3- 5 oxopropyl)morpholine-4-carboxylate (2.2 g, 5.2 mmol) in EtOAc (2 mL) was added HCI/EtOAc (25 mL) at 15 °C. The reaction was stirred at 15 °C for 2 h, then concentrated under reduced pressure to afford methyl (S)-2-(((benzyloxy)carbonyl)amino)-3-((S)-morpholin-2-yl)pro panoate (1.51 g, 90.4% yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for C16H22N 2 O5322.1 ; found 323.2.

Step 9. To a solution of 3-(5-bromo-1-ethyl-2-{2-[(1 S)-1-methoxyethyl]pyridin-3-yl}indol-3-yl)-2,2- 10 dimethylpropan-1 -ol( 100 g, 0.22 mol) and 1 H-imidazole(30.6 g, 0.45 mol) in DCM (800 mL) was added TBSCI (50.7 g, 0.34 mol) in DCM (200 mL) at 0 °C. The reaction was stirred at 25 °C for 2 h. The resulting solution was washed with H 2 O (300 mL x 3) and brine (200 mL x 2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified with silica gel column chromatography to give (S)-5-bromo-3-(3-((tert-butyldimethylsilyl)oxy)-2,2-dimethyl propyl)-1 -ethyl- 15 2-(2-(1 -methoxyethyl)pyridin-3-yl)-1 //-indole (138 g, 90% yield) as an solid. LCMS (ESI): m/z [M+H] calc'd for C29H43BrN 2 02Si 558.2; found 559.2.

Step 10. To a stirred solution of Intermediate 1 (50 g, 89.3 mmol) in dioxane (500 mL) was added methyl (2S)-2-[[(benzyloxy)carbonyl]amino}-3-[(2S)-morpholin-2-yl]p ropanoate from step 1 (31.7 g, 98.2 mmol), RuPhos (16.7 g, 35.7 mmol), Di-mu-chlorobis(2-amino-1 ,1-biphenyl-2-yl-C,N)dipalladium(ll) 20 (2.8 g, 4.4 mmol) and cesium carbonate (96 g, 295 mmol) followed by RuPhos-Pd-G2 (3.5 g, 4.4 mmol) at 105 °C under an N 2 atmosphere. The reaction mixture was stirred for 6 h at 105 °C under an N 2 atmosphere. The resulting mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by prep-TLC chromatography to afford methyl (2S)-2-

{[(benzyloxy)carbonyl]amino}-3-[(2S)-4-(3-{3-[(te/i-butyl dimethylsilyl)oxy]-2,2-dimethylpropyl}-1-ethyl-2-{2- 25 [(1 S)-1 -methoxyethyl]pyridin-3-yl}indol-5-yl)morpholin-2-yl]propano ate (55 g, 73% yield) as a solid. LCMS

(ESI): m/z [M+H] calc’d for C45H64N4O7S1 800.5; found 801.5.

Step 11. To a solution of methyl (2S)-2-{[(benzyloxy)carbonyl]amino)-3-[(2S)-4-(3-[3-[(fert- butyldimethylsilyl)oxy]-2,2-dimethylpropyl}-1-ethyl-2-{2-[(l S)-1-methoxyethyl]pyridin-3-yl}indol-5- yl)morpholin-2-yl]propanoate (10 g, 12 mmol) in THF (270 mL) was added LiOH (1.3 g, 31 mmol) in H 2 O

30 (45 mL) at 20 °C. The reaction was stirred at 20 °C for 2 h, then treated with 1 N HCI to adjust pH to 4~5 at 0~5 °C. The resulting mixture was extracted with EtOAc (50 mL x 2). The combined organic layers were washed with brine and dried over anhydrous NaaSO*. The organic phase was then concentrated under reduced pressure to afford (2S)-2-{[(benzyloxy)carbonyl]amino}-3-[(2S)-4-(3-[3-[(tert- butyldimethylsilyl)oxy]-2,2-dimethylpropyl)-1-ethyl-2-{2-[(1 S)-1-methoxyethyl]pyridin-3-yl}indol-5-

35 yl)morpholin-2-yl]propanoic acid (9.5 g, 97% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C44He2N407Si 786.4; found 787.4.

Step 12. To a stirred solution of (2S)-2-{[(benzyloxy)carbonyl]amino}-3-[(2S)-4-(3-[3-[(tert- butyldimethylsilyl)oxy]-2,2-dimethylpropyl)-1-ethyl-2-{2-[(1 S)-1-methoxyethyl]pyridin-3-yl}indol-5- yl)morpholin-2-yl]propanoic acid (10 g, 12.7 mmol) in DMF (150 mL), was added methyl (S)-

40 hexahydropyridazine-3-carboxylate (2 g, 14 mmol), then cooled to 0 °C, DIPEA (32.8 g, 254 mmol) was added followed by HATU (9.7 g, 25.4 mmol) at 0~5 °C. The reaction mixture was stirred at 0~5 °C for 1 h. The resulting mixture was diluted with EtOAc (500 mL) and H 2 O (200 mL). The organic layer was separated

890

SUBSTITUTE SHEET (RULE 26) and washed with H 2 O (100 mL x 2) and brine (100 mL), dried over anhydrous sodium sulfate. The solution was filtered and concentrated under reduced pressure, and the residue was purified by silica gel column chromatography to afford methyl (S)-1-((S)-2-(((benzyloxy)carbonyl)amino)-3-((S)-4-(3-(3-((t ert- butyldimethylsilyl)oxy)-2,2-dimethylpropyl)-1-ethyl-2-(2-((S )-1-methoxyethyl)pyridin-3-yl)-1H-indol-5- 5 yl)morpholin-2-yl)propanoyl)hexahydropyridazine-3-carboxylat e (8 g, 70% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for CsoHreNeOeSi 912.5; found 913.4.

Step 13. A solution of methyl (S)-1-((S)-2-(((benzyloxy)carbonyl)amino)-3-((S)-4-(3-(3-((f e/t- butyldimethylsilyl)oxy)-2,2-dimethylpropyl)-1-ethyl-2-(2-((S )-1-methoxyethyl)pyridin-3-yl)-1 /-Aindol-5- yl)morpholin-2-yl)propanoyl)hexahydropyridazine-3-carboxylat e (8.5 g, 9 mmol) in THF (8 mL) was added 10 a mixture of tetrabutylammonium fluoride (1 M in THF, 180 mL, 180 mmol) and AcOH (11 g, 200 mmol) at 20 °C. The reaction mixture was stirred at 75 °C for 3 h. The resulting mixture was diluted with EtOAc (150 mL) and washed with H 2 O (20 mL x 6). The organic phase was concentrated under reduced pressure to give methyl (S)-1 -((S)-2-(((benzyloxy)carbonyl)amino)-3-((S)-4-(1 -ethyl-3-(3-hydroxy-2,2-dimethylpropyl)- 2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1 /findol-5-yl)morpholin-2-yl)propanoyl)hexahydropyridazine-3- 15 carboxylate (7.4 g, 100% yield) as solid. LCMS (ESI): m/z [M+H] calc'd for C^HseNeOe 799.4; found 798.4.

Step 14. To a solution of methyl (S)-1-((S)-2-(((benzyloxy)carbonyl)amino)-3-((S)-4-(1-ethyl- 3-(3- hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin -3-yl)-1 H- indol-5-yl)morpholin-2- yl)propanoyl)hexahydropyridazine-3-carboxylate (8 g, 10 mmol) in THF (200 mL) was added lithium hydroxide (600 mg, 25 mmol) in H 2 O (30 mL). The reaction mixture was stirred at 20 °C for 1 h, then treated 20 with 1 N HCI to adjust pH to 4~5 at 0~5 °C, and extracted with EtOAc (500 mL x 2). The organic phase was washed with brine, and concentrated under reduced pressure to afford (S)-1 -((S)-2- (((benzyloxy)carbonyl)amino)-3-((S)-4-(1-ethyl-3-(3-hydroxy- 2,2-dimethylpropyl)-2-(2-((S)-1- methoxyethyl)pyridin-3-yl)-1 H- indol-5-yl)morpholin-2-yl)propanoyl)hexahydropyridazine-3-ca rboxylic acid (8 g, crude) as a solid. LCMS (ESI): m/z [M+H] calc'd for C^HseNeOe 784.4; found 785.4.

25 Step 15. To a stirred solution of (S)-1-((S)-2-(((benzyloxy)carbonyl)amino)-3-((S)-4-(1-ethyl- 3-(3- hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin -3-yl)-1 H- indol-5-yl)morpholin-2- yl)propanoyl)hexahydropyridazine-3-carboxylic acid (8 g, 10.2 mmol) and DIPEA (59 g, 459 mmol) in DCM (800 mL) was added EDCI (88 g, 458 mmol) and HOST (27.6 g, 204 mmol) at 25 °C under an argon atmosphere. The reaction mixture was stirred at 25 °C for 16 h. The resulting mixture was concentrated 30 under reduced pressure, and the residue was purified by silica gel column chromatography to afford benzyl ((2 2 S,6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 ' H-8-oxa-2(4,2)-morpholina-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4- yl)carbamate (5 g, 66% yield) as a solid; LCMS (ESI): m/z [M+H] calc’d for C43H54N6O7766.4; found 767.4.

Step 16. To a solution of benzyl ((2 2 S,6 3 S,4S)-1 '-ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)- 35 10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-morpholina-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-4-yl)carbamate (400 mg, 0.5 mmol) in MeOH (20 mL) was added Pd/C (200 mg) and ammonium acetate (834 mg, 16 mmol) at 20 °C under an Hz atmosphere and the mixture was stirred for 2 h. Then resulting mixture was filtered and concentrated under reduced pressure. The residue was redissolved in DCM (20 mL) and washed with HzO (5 mL x 2), then concentrated under reduced 40 pressure to afford (2 2 S,6 3 S,4S)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl- 6 , ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-morpholina-1 (5,3)-indola-6(1 ,3)-

891

SUBSTITUTE SHEET (RULE 26) pyridazinacycloundecaphane-5,7-dione (320 mg, 97% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for CssH^eNeOs 632.4; found 633.3.

Step 17. To a solution of the (2 2 S,6 3 S,4S)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3- yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-2(4,2)-morpholina-1 (5,3)-indola-6(1 ,3)-

5 pyridazinacycloundecaphane-5,7-dione (50 mg, 0.079 mmol), N-(1-(4-(dimethylamino)-4-methylpent-2- ynoyl)-4-fluoropiperidine-4-carbonyl)-N-methyl-L-valine (47 mg, 0.12 mmol) in DMF (2 mL) stirred at 0 °C was added HATU (36 mg, 0.09 mmol) and DIPEA (153 mg, 1.2 mmol) dropwise. The reaction was stirred at 0 °C for 1 h. The resulting mixture was purified by reverse phase to afford 1 -(4-(dimethylamino)-4- methylpent-2-ynoyl)-N-((2S)-1 -(((2 2 S,6 3 S,4S)-1 '-ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-

10 dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-morpholina-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2 -yl)-4-fluoro-N-methylpiperidine-4- carboxamide (11.9 mg, 13.9% yield) as a solid. Ή NMR (400 MHz, CDaOD) 68.71 (dd, J= 4.8, 1.7 Hz,

1 H), 7.86 (d, J = 7.8 Hz, 1 H), 7.51 (dd, J = 7.8, 4.8 Hz, 1 H), 7.39 (d, J = 8.9 Hz, 1 H), 7.15 - 7.04 (m, 2H), 5.67 (d, J = 8.8 Hz, 1 H), 4.62 (d, J = 11.2 Hz, 1 H), 4.46 (d, J = 12.4 Hz, 1 H), 4.39 - 4.27 (m, 2H), 4.23 (d,

15 J= 6.1 Hz, 1H), 4.17- 4.08 (m, 1H), 3.93 (s, 2H), 3.86 (s, 1H), 3.84 - 3.76 (m, 2H), 3.74- 3.65 (m, 2H), 3.63 - 3.51 (m, 2H), 3.27 - 3.23 (m, 1 H), 3.22 - 3.11 (m, 6H), 3.0 - 2.89 (m, 2H), 2.85 - 2.75 (m, 2H), 2.74 - 2.55 (m, 2H), 2.36 (d, J = 8.2 Hz, 6H), 2.32 - 2.21 (m, 2H), 2.20 - 2.02 (m, 5H), 1.92 (d, J = 12.5 Hz, 2H), 1.69 (dd, J= 43.8, 12.6 Hz, 2H), 1.46 (dt, J= 8.0, 4.9 Hz, 9H), 1.03 (d, J= 3.5 Hz, 3H), 0.90 (dd, J= 48.3, 6.5 Hz, 6H), 0.77 (d, J= 3.0 Hz, 3H), 0.69 (s, 3H). LCMS (ESI): m/z [M+H] calc'd for

20 CssHyeFNgOe 1011.6; found 1012.5.

892

SUBSTITUTE SHEET (RULE 26) Example A375. Synthesis of (2fl)-2-(((1-(4-(dlmethylamlno)-4-methylpent-2-ynoyl)azetldl n-3- yl)oxy)methyl)-W-((2 2 S,6 3 S,4S)-1 2 -(2-((S)-1 -methoxyethyl)pyrldln-3-yl)-1 O.10-dlmethyl-S.T-dloxo-l 1 - (2,2,2-trif luoroethyl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 //-8-oxa-2(4,2)-morpholina-1 (5,3)-lndola-6(1 ,3)- pyridazlnacycloundecaphane-4-yl)-3-methylbutanamlde

.OTBDPS ,οτΒθΡε

>TBDPS

TfO^CF *

NaH .Br .Br

Br

DMF

^OTBDPS JOTBOPS ^OTBOPS ,

.«Cttt

MbB(OH)2, NSOH

MeOHAlgOm# n n o

YVY°

UOH Cbz TBAF/AOOH r nr

THF*½0 THF

CF,

,N

N Q

H

EOC:, HOflt DtPEA ! Cbz Ύ f ‘N'

NH r Λ Ν

RVC. NH^a HATU, DIPEA

DCM MeOH O OMF 0"

5 S*

Step 1. To a mixture of 5-bromo-3-{3-[(fert-butyldiphenylsilyl)oxy]-2,2-dimethylprop yl}-2-{2-[(1 S)- 1 -methoxyethyl]pyridin-3-yl}-1 //-indole (10.0 g, 15.2 mmol) in anhydrous DMF (120 mL) at 0 °C under an atmosphere of Nz was added NaH, 60% dispersion in oil (1.2 g, 30.4 mmol) and 2,2,2-trifluoroethyl trifluoromethanesulfonate (35.4 g, 152 mmol). The mixture was stirred at 0 °C for 1 h, then saturated

10 NHACI (30 mL) added and the mixture extracted with EtOAc (100 mLx 3). The combined organic layers were washed with brine, dried over anhydrous NazSO* and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give (S)-5-bromo- 3-(3-((tert-butyldiphenylsilyl)oxy)-2,2-dimethylpropyl)-2-(2 -(1-methoxyethyl)pyridin-3-yl)-1 -(2,2,2- trifluoroethyl)-1 //-indole (8 g) as an oil and the other atropisomer (6 g, 48% yield) as an oil. LCMS (ESI):

15 m/z [M+H] calc'd for CssH^BrFsNzOzSi 736.2; found 737.1.

Step 2. To a mixture of (S)-5-bromo-3-(3-((fert-butyldiphenylsilyl)oxy)-2,2-dimethyl propyl)-2-(2- (1-methoxyethyl)pyridin-3-yl)-1-(2,2,2-trifluoroethyl)-1 //-indole (7.2 g, 9.7 mmol) in toulene (80 mL) under an atmosphere of Nz was added 4,4,4' ,4' ,5,5,5' ,5' -octamethyl-2,2' -bi(1 ,3,2-dioxaborolane) (2.7 g, 10.6 mmol), KOAc (1.9 g, 19.4 mmol) and Pd(dppf)Clz DCM (0.8 g, 0.1 mmol). The mixture was heated to

20 90 °C and stirred for 8 h, then saturated NH4CI (30 mL) added and the mixture extracted with EtOAc (40 mL x 3). The combined organic layers were dried over anhydrous NazS04 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give (S)-3-(3-((fert-butyldiphenylsilyl)oxy)-2,2-dimethylpropyl)- 2-(2-(1-methoxyethyl)pyridin-3-yl)-5- (4,4,5,5-tetramethyM ,3,2-dioxaborolan-2-yl)-1 -(2,2,2-trifluoroethyl)-1 //-indole (6.1 g, 64% yield) as an oil.

25 LCMS (ESI): m/z [M+H] calc’d for C4sHseBF3Nz04Si 784.4; found 785.3.

893

SUBSTITUTE SHEET (RULE 26) Step 3. To a mixture of (S)-3-(3-((te/t-butyldiphenylsilyl)oxy)-2,2-dimethylpropyl)- 2-(2-(1- methoxyethyl)pyridin-3-yl)-5-(4,4,5,5-tetramethyl-1,3,2-diox aborolan-2-yl)-1-(2,2,2-trifluoroethyl)-1H-indole (33 g, 42 mmol) in THF (120 mL) and MeOH (330 mL) at 0 °C under an atmosphere of Nfe was added MeB(OH)2 (50.4 g, 841 mmol), then a mixture of NaOH (33.6 g, 841 mmol) in H 2 O (120 mL). The mixture 5 was warmed to room temperature and stirred for 16 h, then concentrated under reduced pressure. HzO (500 mL) was added to the residue and the mixture extracted with EtOAc (300 mL x 3). The combined organic layers were washed with brine (300 mL), HzO (300 mL), then concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give (S)-(3-(3-((fert- butyldiphenylsilyl)oxy)-2,2-dimethylpropyl)-2-(2-(1-methoxye thyl)pyridin-3-yl)-1-(2,2,2-trifluoroethyl)-1/+

10 indol-5-yl)boronic acid (20 g, 68% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for C39H46BF3N 2 0(Si 702.3; found 703.3.

Step 4. Note: Three reactions were run in parallel - the yield reflects the sum of the products.

A mixture of (S)-(3-(3-((te/i-butyldiphenylsilyl)oxy)-2,2-dimethylpropyl) -2-(2-(1- methoxyethyl)pyridin-3-yl)-1-(2,2,2-trifluoroethyl)-1 H-indol-5-yl)boronic acid (1.85 g, 5.6 mmol) and methyl 15 (S)-2-(((benzyloxy)carbonyl)amino)-3-((S)-morpholin-2-yl)pro panoate in DCM (150 mL) under air was added pyridine (1.35 g, 16.9 mmol) and Cu(OAc)2 (2.0 g, 11.3 mmol). The mixture was stirred for 48 h, then concentrated under reduced pressure. H 2 O (300 mL) was added to the residue and the mixture was extracted with EtOAc (300 mL x 2). The combined organic layers were washed with brine (100 mL), dried over anhydrous Na 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue 20 was silica gel column chromatography to give methyl (S)-2-(((benzyloxy)carbonyl)amino)-3-((S)-4-(3-(3- ((tert-butyldiphenylsilyl)oxy)-2,2-dimethylpropyl)-2-(2-((S) -1-methoxyethyl)pyridin-3-yl)-1 -(2,2,2- trifluoroethyl)-1 H- indol-5-yl)morpholin-2-yl)propanoate (9.2 g, 55% yield) as a solid. LCMS (ESI): m/z [M/2+H] calc'd for CssHesFaN^Si 490.2; found 490.3.

Step 5. To a mixture of methyl (S)-2-(((benzyloxy)carbonyl)amino)-3-((S)-4-(3-(3-((tert- 25 butyldiphenylsilyl)oxy)-2,2-dimethylpropyl)-2-(2-((S)-1-meth oxyethyl)pyridin-3-yl)-1-(2,2,2-trifiuoroethyl)- 1 H- indol-5-yl)morpholin-2-yl)propanoate (10.8 g, 11.0 mmol) in THF (50 mL) was added LiOH (528 mg,

22 mmol) in H 2 O (10 mL). The mixture was stirred for 1 h, then cooled to 0-5 °C and acidified to pH~7 using 2N HCI (10 mL). The mixture was extracted with DCM (100 mL x 2) and the combined organic layers were dried over Na 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure to give 30 (S)-2-(((benzyloxy)carbonyl)amino)-3-((S)-4-(3-(3-((tert-but yldiphenylsilyl)oxy)-2,2-dimethylpropyl)-2-(2- ((S)-1 -methoxyethyl)pyridin-3-yl)-1 - (2 ,2 ,2-tr if I uoro ethyl) - 1 H- indol-5-yl)morpholin-2-yl)propanoic acid (10.6 g, 100% yield) as a solid. LCMS (ESI): m/z [M/2+H] calc’d for C 5 4H 63 F 3 N407SI 483.2; found 483.3.

Step 6. To a mixture of (S)-2-(((benzyloxy)carbonyl)amino)-3-((S)-4-(3-(3-((tert- butyldiphenylsilyl)oxy)-2,2-dimethylpropyl)-2-(2-((S)-1-meth oxyethyl)pyridin-3-yl)-1-(2,2,2-trifiuoroethyl)- 35 1 H- indol-5-yl)morpholin-2-yl)propanoic acid (10.6 g, 11.0 mmol) and methyl (3S)-1 ,2-diazinane-3- carboxylate (15.8 g, 22.0 mmol) in DMF (150 mL) at 0 °C was added DIPEA (28.4 g, 220 mmol) and HATU (8.4 g, 22.0 mmol). The mixture was stirred at 0~5 °C for 1 h, then EtOAc (500 mL) was added and the mixture was washed with H 2 O (200 mL x 2), brine (100 mL), dried over anhydrous Na 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel 40 column chromatography to give methyl (S)-1 -((S)-2-(((benzyloxy)carbonyl)amino)-3-((S)-4-(3-(3-((te/f- butyldiphenylsilyl)oxy)-2,2-dimethylpropyl)-2-(2-((S)-1-meth oxyethyl)pyridin-3-yl)-1-(2,2,2-trifluoroethyl)-

894

SUBSTITUTE SHEET (RULE 26) 1 H- indol-5-yl)morpholin-2-yl)propanoyl)hexahydropyridazine-3-ca rboxylate (11 g, 90% yield) as a solid. LCMS (ESI): m/z [M/2+H] calc'd for CeoHTsFsNeOeSi 546.3; found 546.3.

Step 7. To a mixture of methyl (S)-1-((S)-2-(((benzyloxy)carbonyl)amino)-3-((S)-4-(3-(3-((t e/i- butyldiphenylsilyl)oxy)-2,2-dimethylpropyl)-2-(2-((S)-1-meth oxyethyl)pyridin-3-yl)-1-(2,2,2-trifluoroethyl)- 5 1 H- indol-5-yl)morpholin-2-yl)propanoyl)hexahydropyridazine-3-ca rboxylate (11.0 g, 10.1 mmol) in THF

(10 mL) was added a mixture of AcOH (21.2 g, 353 mmol) and 1 M TBAF in THF (300 mL, 300 mmol).

The mixture was heated to 80 °C and stirred for 16 h, then concentrated under reduced pressure. EtOAc (800 mL) was added to the residue and the mixture was washed with H 2 O (80 mL x 6), concentrated under reduced pressure and the residue was purified by preparative-HPLC to give methyl (S)-1 -((S)-2- 10 (((benzyloxy)carbonyl)amino)-3-((S)-4-(3-(3-hydroxy-2,2-dime thylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin- 3-yl)-1-(2,2,2-trifluoroethyl)-1 H- indol-5-yl)morpholin-2-yl)propanoyl)hexahydropyridazine-3-ca rboxylate (7.9 g, 91% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for CuHssFsNeOe 852.4; found 853.3.

Step 8. To a mixture of methyl (S)-1-((S)-2-(((benzyloxy)carbonyl)amino)-3-((S)-4-(3-(3-hyd roxy- 2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1 -(2,2,2-trifluoroethyl)-1H-indol-5-yl)morpholin- 15 2-yl)propanoyl)hexahydropyridazine-3-carboxylate (7.9 g, 9.3 mmol) in THF (50 mL) was added LiOH (443 mg, 18.5 mmol) in H 2 O (10 mL). The mixture was stirred for 1 h, then cooled to 0-5 °C and acidified to ~pH 7 with 2N HCI (9 mL). The mixture was extracted with DCM (100 mL x 2) and the combined organic layers were dried over NaaSO* and filtered. The filtrate was concentrated under reduced pressure to give (S)-1 -((S)-2-(((benzyloxy)carbonyl)amino)-3-((S)-4-(3-(3-hydroxy- 2,2-dimethylpropyl)-2-(2-((S)-1 - 20 methoxyethyl)pyridin-3-yl)-1 -(2,2,2-trifluoroethyl)-1 H-indol-5-yl)morpholin-2- yl)propanoyl)hexahydropyridazine-3-carboxylic acid (7.6 g, 98% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for CwHssFaNeOe 838.4; found 839.3.

Step 9. To a mixture of (S)-1 -((S)-2-(((benzyloxy)carbonyl)amino)-3-((S)-4-(3-(3-hydroxy- 2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1-(2, 2,2-trifluoroethyl)-1 H- indol-5-yl)morpholin-2- 25 yl)propanoyl)hexahydropyridazine-3-carboxylic acid (7.6 g, 9.0 mmol) and DIPEA (52.3 g, 405 mmol) in DCM (800 mL) under an atmosphere of Ar was added EDCI (77.6 g, 405 mmol) and HOST (12 g, 90 mmol). The mixture was stirred for 16 h, then concentrated under reduced pressure. The residue was diluted with EtOAc (500 mL), washed with H 2 O (100 mL x 2) and filtered. The organic layer was concentrated under reduced pressure and the residue was purified by silica gel column chromatography 30 to give benzyl ((2 2 S,6 3 S,4S)-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-1 1 -(2,2,2- trifluoroethyl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-morpholina-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-4-yl)carbamate (6.1 g, 74% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C43H51F3N6O7820.4; found 821.3.

Step 10. To a mixture of benzyl ((2 2 S,6 3 S,4S)-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10- 35 dimethyl-5, 7-dioxo-1 1 -(2, 2, 2-trifluoroethyl)-6 1 ,6 2 ,6 3 ,6 4 , 6 s , 6 6 -hexahydro-1 1 H- 8-oxa-2(4,2)-morpholina-

1 (5,3)-indola-6(1 ,3)-pyridazinacydoundecaphane-4-yl)carbamate (700 mg, 0.85 mmol) in MeOH (30 mL) was added 10% Pd on C (317 mg) and NHXI (909 mg). The mixture was stirred under an atmosphere of Ha (1 atm) for 16 h, then filtered through Celite and the filter cake was washed with MeOH (150 mL). The filtrate was concentrated under reduced pressure, DCM (20 mL) was added to the residue and the 40 mixture was washed with saturated NaHCOa (20 mL x 3). The organic layer was concentrated under reduced pressure to give (2 2 S,6 3 S,4S)-4-amino-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-1 1 - ^^-trifluoroethylj-e 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-morpholina-1 (5,3)-indola-6(1 ,3)-

895

SUBSTITUTE SHEET (RULE 26) pyridazinacycloundecaphane-5,7-dione (660 mg, 95% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for C35H45F3N6O5686.3; found 687.3; Ή NMR (400 MHz, CDCIs) 68.80 (dd, J= 4.7, 1.7 Hz, 1H), 7.66 (d, J = 7.4 Hz, 1H), 7.43 - 7.30 (m, 2H), 7.12 - 7.01 (m, 2H), 4.90 - 4.83 (m, 1H), 4.68 (d, J= 12.5 Hz, 1H), 4.57 (dd, J= 16.2, 8.1 Hz, 1H), 4.24 (q, J= 6.1 Hz, 1H), 4.08 (d, J= 10.6 Hz, 1H), 3.97 - 3.82 (m, 4H), 3.80 -

5 3.68 (m, 2H), 3.55 (d, J = 11.6 Hz, 1 H), 3.21 (d, J = 9.4 Hz, 1 H), 2.93 (dd, J = 19.9, 9.3 Hz, 3H), 2.66 (t, J = 11.6 Hz, 1 H), 2.47 (d, J = 14.5 Hz, 1 H), 2.19 - 2.04 (m, 4H), 1.96 (d, J = 13.6 Hz, 2H), 1.80 - 1.71 (m, 2H), 1.66 - 1.59 (m, 1 H), 1.47 (d, J = 6.1 Hz, 3H), 0.88 (s, 3H), 0.42 (s, 3H).

Step 11. To a mixture of (2 2 S,6 3 S,4S)-4-amino-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10- dimethyl-1 '-^^-trifluoroethyO-e 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-morpholina-1 (5,3)-indola-

10 6(1 ,3)-pyridazinacycloundecaphane-5,7-dione (300 mg, 0.4 mmol), (2fl)-2-[({1-[4-(dimethylamino)-4- methylpent-2-ynoyl]azetidin-3-yl}oxy)methyl]-3-methylbutanoi c acid (157 mg, 0.48 mmol) and DIPEA (569.0 mg, 0.4 mmol) in DMF (5 mL) at 0 °C was added HATU (217 mg, 0.57 mmol). The mixture was stirred at 0 °C for 0.5 h, then diluted with HzO and extracted with EtOAc (20 mL x 3). The combined organic layers were washed with brine (20 mL x 3), dried over NaaSO* and filtered. The filtrate was

15 concentrated under reduced pressure and the residue was purified by preparative-TLC to give (2fl)-2- (((1-(4-(dimethylamino)-4-methylpent-2-ynoyl)azetidin-3-yl)o xy)methyl)-N-((2 2 S,6 3 S,4S)-12-(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-1 '-(2,2,2-trifluoroethyl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro- 1 1 H- 8-oxa-2(4,2)-morpholina-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)-3-methylbutanamide (200 mg, 46% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for CaHyiFaNeOe 992.5; found 993.4; 1 H

20 NMR (400 MHz, CD3OD) 68.74 (m, 1H), 8.00 (m, 1H), 7.85 (d, J= 7.7 Hz, 1H), 7.60 - 7.43 (m, 2H), 7.14 (t, J = 9.5 Hz, 2H), 5.63 (s, 1 H), 5.06 (m, 1 H), 4.64 (s, 1 H), 4.52 - 4.31 (m, 3H), 4.27 - 4.05 (m, 3H), 3.97 (m, 1H), 3.92 - 3.66 (m, 6H), 3.59 (m, 2H), 3.46 (m, 2H), 3.25 (d, J= 5.3 Hz, 3H), 3.07 - 2.89 (m, 2H),

2.86 - 2.59 (m, 3H), 2.38 - 2.32 (m, 3H), 2.28 (s, 3H), 2.13 (m, 2H), 2.03 - 1.51 (m, 6H), 1.50 - 1.41 (m, 6H), 1.38 (d, J= 7.5 Hz, 3H), 0.98 (t, J= 8.7 Hz, 6H), 0.89 (t, J= 6.4 Hz, 3H), 0.54 (d, J= 8.4 Hz, 3H).

25

896

SUBSTITUTE SHEET (RULE 26) Example A722. Synthesis of 1-acryk>yl-AH(2S)-1-(((6 3 ^4S)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)pyrldin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)- morphollna-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)-4-fluoro-jV-methylplperldlne-4-carboxamlde

5

Step 1. To a mixture of N-(4-fluoropiperidine-4-carbonyl)-N-methyl-L-valine (190 mg, 0.73 mmol) and NaHCOa (306 mg, 3.6 mmol) in DCM (2 mL) and H 2 O (1 mL) at -10 °C was added prop-2-enoyl chloride (132 mg, 1.45 mmol). The mixture was stirred at 0-5 °C for 1 h, then diluted with DCM (20 mL) and washed with H 2 O (20 mL x 2), brine (20 mL) and the organic layer dried over NazSOt and filtered.

10 The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give N-(1-acryloyl-4-fluoropiperidine-4-carbonyl)-N-methyl-L-vali ne (120 mg, 52% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C15H23FN 2 O4314.2; found 315.2.

Step 2. To a mixture of (6 3 S,4S)-4-amino-1 1 -ethyl- 1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10- dimethyl-6 1 ,6 2 ,6 3 , 6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-morpholina-1 (5,3)-indola-6(1 ,3)-

15 pyridazinacycloundecaphane-5,7-dione (153 mg, 0.24 mmol), W-(1-acryloyl-4-fluoropiperidine-4- carbonyl)-N-methyl-L-valine (106 mg, 0.34 mmol) in DMF(2 mL) at 5 °C was added HATU (110 mg, 0.29 mmol) and DIPEA (468 mg, 3.6 mmol) dropwise. The mixture was stirred at 5 °C for 1 h, then purified by preparative-HPLC to give 1 -acryloyl-N-((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-2(4,2)-morpholina-1 (5,3)-indola-6(1 ,3)-

20 pyridazinacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2 -yl)-4-fluoro-W-methylpiperidine-4- carboxamide (69.5 mg, 29% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for CsoHesFNsOs 928.5; found 929.4; 1 H NMR (400 MHz, CD3OD) 68.71 (d, J= 3.2 Hz, 1 H), 7.86 (d, J= 7.7 Hz, 1H), 7.51 (dd, J = 7.7, 4.8 Hz, 1H), 7.39 (d, J= 8.9 Hz, 1H), 7.18 - 7.02 (m, 2H), 6.80 (dd, J= 16.8, 10.7 Hz, 1H), 6.23 (d, J = 16.8 Hz, 1 H), 5.77 (d, J = 10.6 Hz, 1 H), 5.67 (d, J = 6.5 Hz, 1 H), 4.61 (d, J = 11 .1 Hz, 1 H), 4.44 (t, J =

25 15.1 Hz, 2H), 4.23 (q, J = 6.1 Hz, 1 H), 4.18 - 4.10 (m, 1 H), 4.09 - 4.01 (m, 1 H), 3.99 - 3.83 (m, 3H), 3.83 - 3.65 (m, 4H), 3.58 - 3.46 (m, 2H), 3.27 (s, 1H), 3.21 - 3.11 (m, 6H), 3.00 - 2.91 (m, 2H), 2.85 - 2.75 (m, 2H), 2.73 - 2.64 (m, 1H), 2.62 - 2.54 (m, 1H), 2.36 - 2.21 (m, 2H), 2.19 - 2.01 (m, 5H), 1.92 (d, J= 12.7 Hz, 2H), 1.79 - 1.57 (m, 2H), 1.44 (d, J = 6.2 Hz, 3H), 1.04 (t, J = 6.3 Hz, 3H), 0.98 - 0.81 (m, 6H), 0.76 (s, 3H), 0.68 (s, 3H).

30

897

SUBSTITUTE SHEET (RULE 26) Example Α377. Synthesis of (2fl)-2-(((1-(4-(dlmethylamlno)-4-methylpent-2-ynoyl)azetldl n-3- yl)oxy)methyl)-W-((2 2 S,6 3 S,4S)-1 1 -ethyH 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7- dloxo-6 1 ,6 2 ,6 3 ,6 4 ,6 6 ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-morpholina-1 (5,3)-lndola-6(1 ,3)- pyridazlnacycloundecaphane-4-yl)-3-methylbutanamlde

5

Step 1. (2fl)-2-(((1 -(4-(dimethylamino)-4-methylpent-2-ynoyl)azetidin-3-yl)oxy)m ethyl)-N- ((2 2 S,6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 , 6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 1 H-8-oxa-2(4,2)-morpholina-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)-3- methylbutanamide was synthesized in a manner similar to (2fl)-2-(((1-(4-(dimethylamino)-4-methylpent-2-

10 ynoyl)azetidin-3-yl)oxy)methyl)-N-((2 2 S,6 3 S,4S)-12-(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl- 5,7-dioxo-1 1 -(2,2,2-trifluoroethyl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-morpholina-1 (5,3)-indola- 6(1 ,3)-pyridazinacycloundecaphane-4-yl)-3-methylbutanamide except (2 2 S,6 3 S,4S)-4-amino-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-1 , -(2,2,2-trifluoroethyl)-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa- 2(4,2)-morpholina-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-5,7-dione was substituted with

15 (2 2 S,6 3 S,4S)-4-amino-1 1 -ethyl- 1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 1 H-8-oxa-2(4,2)-morpholina-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-5,7-dione and 3-({1-[4-(dimethylamino)-4-methylpent-2-ynoyl]azetidin-3-yl} oxy)propanoic acid was substituted with (2fl)- 2-[({1-[4-(dimethylamino)-4-methylpent-2-ynoyl]azetidin-3-yl }oxy)methyl]-3-methylbutanoic acid to give the desired product (25.6 mg, 26% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C52H74NBOB 938.6;

20 found 939.5; 1 H NMR (400 MHz, CD3OD) 68.72 (dd, J= 4.8, 1.5 Hz, 1H), 7.97 (dd, J= 20.0, 6.8 Hz, 1 H), 7.87 (dd, J = 5.8, 2.6 Hz, 1 H), 7.54 - 7.51 (m, 1H), 7.41 (d, J= 8.9 Hz, 1H), 7.14 (dd, J= 36.0, 10.4 Hz, 2H), 5.65 (s, 1 H), 4.49 - 4.33 (m, 3H), 4.27 - 4.08 (m, 4H), 3.96 (d, J = 8.6 Hz, 2H), 3.87 (dd, J = 10.8, 3.6 Hz, 2H), 3.79 (dd, J= 10.8, 7.7 Hz, 3H), 3.69 - 3.58 (m, 3H), 3.43 (dd, J= 23.9, 11.7 Hz, 2H), 3.17 (d, J = 21.9 Hz, 3H), 3.00 - 2.95 (m, 1H), 2.70 (t, J= 14.0 Hz, 8H), 2.34 - 2.24 (m, 1H), 2.05 (d, J= 34.4 Hz, 3H),

25 1.92 - 1.82 (m, 2H), 1.69 - 1.62 (m, 5H), 1.57 (d, J = 11.5 Hz, 3H), 1.45 (d, J = 6.2 Hz, 3H), 1.33 (d, J = 12.6 Hz, 1H), 1.05 - 0.93 (m, 10H), 0.80 (d, J= 9.8 Hz, 3H), 0.64 (d, J= 12.2 Hz, 2H).

898

SUBSTITUTE SHEET (RULE 26) Example A643. Synthesis of (2/7)-2-(((1-(4-(dimethylamino)-4-methylpent-2-ynoyl)piperki in- 4-yl)oxy)methyl)-N-((2 2 S,6 3 S,4S)-1 1 -ethyl-1 z -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dlmethyl-5,7- dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 ® ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-morpholina-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-4-yl)-3-methylbutanamide

5

Step 1. A mixture of 1-(1-methylphenyl)piperidin-4-yl methanesulfonate (2 g, 7.4 mmol) and tert- butyl (2fl)-2-(hydroxymethyl)-3-methylbutanoate (1.39 g, 7.4 mmol) was stirred at 120 °C for 1 h, then purified by silica gel column chromatography to give tert-butyl (fl)-2-(((1-benzylpiperidin-4-yl)oxy)methyl)- 3-methylbutanoate (800 mg, 28% yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for C22H35NO3361.3;

10 found 362.3.

Step 2. A mixture of tert-butyl (R)- 2-(((1 -benzylpiperidin-4-yl)oxy)methyl)-3-methylbutanoate (700 mg, 1.9 mmol) , 10% wet Pd/C (411 mg, 3.9 mmol) and 20% wet Pd(OH)2/C (542mg, 3.9 mmol) in THF (30 mL) was stirred under an atmosphere of Ha (15 psi) for 16 h. The mixture was filtered and the filtrate was concentrated under reduced pressure to give tert-butyl (fl)-3-methyl-2-((piperidin-4-

15 yloxy)methyl)butanoate (440 mg, 80% yield) as a an oil. LCMS (ESI): m/z [M+H] calc’d for C15H29NO3 271.2; found 272.2.

Step 3. To a mixture of tert-butyl (fl)-3-methyl-2-((piperidin-4-yloxy)methyl)butanoate (440 mg,

1.6 mmol) 4-(dimethylamino)-4-methylpent-2-ynoic acid (3.77 g, 24.3 mmol) and DIPEA (2.09 g, 16.2 mmol) in DMF (50 mL) at 0 °C was added T3P (2.57 g, 8.1 mmol). The mixture was stirred at 0 °C for 1 h,

20 then poured into H 2 O (50 mL) and extracted with EtOAc (50mL x 3). The combined organic layers were washed with brine, concentrated under reduced pressure and the residue purified by silica gel column chromatography to give tert-butyl (fl)-2-(((1-(4-(dimethylamino)-4-methylpent-2-ynoyl)piperidi n-4- yl)oxy)methyl)-3-methylbutanoate (190 mg, 27% yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for C23H40N 2 O4408.3; found 409.4.

25 Step 4. To a mixture of tert-butyl (fl)-2-(((1-(4-(dimethylamino)-4-methylpent-2-ynoyl)piperidi n-4- yl)oxy)methyl)-3-methylbutanoate (180 mg, 0.47 mmol) in DCM (2 mL) was added TFA (1 mL). The mixture was stirred at room temperature for 1 h, then concentrated under reduced pressure to give ( R)-2 -

899

SUBSTITUTE SHEET (RULE 26) (((1 -(4-(dimethylamino)-4-methylpent-2-ynoyl)piperidin-4-yl)oxy) methyl)-3-methylbutanoic acid (170 mg, 98% yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for CigHaaNaCfc 352.2; found 353.2.

Step 5. (2fl)-2-(((1 -(4-(dimethylamino)-4-methylpent-2-ynoyl)piperidin-4-yl)oxy) methyl)-N- ((2 2 S,6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - 5 hexahydro-1 1 H-8-oxa-2(4,2)-morpholina-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)-3- methylbutanamide was synthesized in a manner similar to (2fl)-2-(((1-(4-(dimethylamino)-4-methylpent-2- ynoyl)azetidin-3-yl)oxy)methyl)-N-((2 2 S,6 3 S,4S)-12-(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl- 5,7-dioxo-1 1 -(2,2,2-trifluoroethyl)-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 /+8-oxa-2(4,2)-morpholina-1 (5,3)-indola- 6(1 ,3)-pyridazinacycloundecaphane-4-yl)-3-methylbutanamide except (2 2 S,6 3 S,4S)-4-amino-1 2 -(2-((S)-1- 10 methoxyethyl)pyridin-3-yl)-10,10-dimethyl-1 l -(2,2,2-trifluoroethyl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 'H-Q-oxa- 2(4,2)-morpholina-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-5,7-dione was substituted with (2 2 S,6 3 S,4S)-4-amino-1 '-ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 1 H-8-oxa-2(4,2)-morpholina-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-5,7-dione and 3-({1-[4-(dimethylamino)-4-methylpent-2-ynoyl]azetidin-3-yl} oxy)propanoic acid was substituted with (2R)- 15 2-[({1-[4-(dimethylamino)-4-methylpent-2-ynoyl]piperidin-4-y l}oxy)methyl]-3-methylbutanoic acid. (101 mg, 42% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for Cs+HyeNeOa 966.6; found 969.5; Ή NMR (400 MHz, CDaOD) 68.70 (d, J = 4.8 Hz, 1 H), 7.81 - 7.76 (m, 1 H), 7.56 - 7.46 (m, 1 H), 7.43 - 7.34 (m, 1 H), 7.24 - 7.01 (m, 2H), 5.66 - 5.54 (m, 1 H), 4.50 - 4.40 (m, 1 H), 4.31 - 4.22 (m, 1 H), 4.19 - 4.08 (m, 1 H), 4.02 - 3.82(m, 4H), 3.80 - 3.53 (m, 10H), 3.47 - 3.34 (m, 2H), 3.26 - 3.15 (m, 3H), 2.98 - 2.57 (m, 5H), 2.37 - 20 2.30 (m, 3H), 2.27 - 2.18 (m, 4H), 2.15 - 2.02 (m, 2H), 2.00 - 1.80 (m, 4H), 1.78 - 1.71 (m, 2H), 1.68 - 1 .55

(m, 3H), 1 .49 - 1.37 (m, 6H), 1.35 - 1.28 (m, 3H), 1 .05 - 0.92 (m, 9H), 0.85 - 0.72 (m, 3H), 0.68 - 0.51 (m,

3H).

900

SUBSTITUTE SHEET (RULE 26) Example A328. Synthesis of two atroplsomers of (3S)-1 -aery loy I- W-((2S)-1 -(((6 3 S,4S)-1 1 - ethyl-1 2 -(2-((S)-1 -methoxyethyl)-5-(4-methylplperazln-1 -yl)pyridin-3-yl)-10,10-dlmethyl-5,7-dloxo- 6 1 ,6 2 ,6 3 ,6 4 f 6 6 ,6 e -hexahydro-1 1 H- 8-oxa-1 (5,3)-lndola-6(1 ,3)-pyridazlna-2(1 ,3>- benzenacycloundecaphane-4-yl)amlno)-3-methyH-oxobutan-2-yl)- Af-methylpyrrolldlne-3- 5 carboxamide

Step 1. To a mixture of 3-bromo-5-iodo-2-[(1 S)-1 -methoxyethyljpyridine (2.20 g, 6.4 mmol) and fert-butyl piperazine-1 -carboxylate (1.20 g, 6.4 mmol) in toluene (50 mL) under an atmosphere of Ar were added ffiuONa (0.74 g, 7.7 mmol) and portion-wise addition of Pd2(dba)s (0.59 g, 0.64 mmol), followed by

10 portion-wise addition of Xanlphos (0.74 g, 1.3 mmol). The mixture was heated to 100 °C and stirred for 16 h then H 2 O added and the mixture extracted with EtOAc (400 mL x 3). The combined organic layers were washed with brine (150 mL x 3), dried over anhydrous NaaSO* and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by preparative-HPLC to give tort-butyl 4-[5-bromo- 6-[(1 S)-1 -methoxyethyl]pyridin-3-yl]piperazine-1 -carboxylate (1.7g, 61% yield) as a solid. LCMS (ESI):

15 m/z [M+H] calc’d for CizHaeBrNaOa 399.1 ; found 400.1.

Step 2. A mixture of tort-butyl 4-[5-bromo-6-[(1 S)-1 -methoxyethyl]pyridin-3-yl]piperazine-1 - carboxylate (1.76 g, 4.4 mmol) and 4,4,4',4 , ,5,5,5 , ,5'-octamethyl-2,2 , -bi(1 ,3,2-dioxaborolane) (1.67 g, 6.6 mmol) in toluene (18 mL) under an atmosphere of Ar were added KOAc (0.95 g, 9.7 mmol) and Pd(PPhs)2Cl 2 (0.31 g, 0.44 mmol) in portions. The mixture was heated to 80 °C and stirred for 16 h, then

20 diluted with H 2 O and the mixture extracted with EtOAc (500 mL x 3). The combined organic layers were washed with brine (100 mL x 3), dried over anhydrous Na 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give tort- butyl 4-[6-[(1 S)-1 -methoxyethyl]-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-3-yl]piperazine-1 -

901

SUBSTITUTE SHEET (RULE 26) carboxylate (1.4 g, 68% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for CzsHseBNsOs 447.4; found 448.2.

Step 3. To a mixture of fert-butyl ((6 3 S,4S)-1 2 -iodo-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate 5 (1.0 g, 1.5 mmol) in DCM (10 mL) at 0 ’C under an atmosphere of Nz was added TFA (5.0 mL, 67.3 mmol) in portions. The mixture was stirred at 0 °C for 1 h then concentrated under reduced pressure and dried azeotropically with toluene (3 mLx 3) to give (6 3 S,4S)-4-amino-1 2 -iodo-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-5,7-dione (1.0 g), which was used directly in the next step without further purification. LCMS (ESI): m/z [M+H] calc’d for 10 C27H31IN4O3586.1; found 587.3.

Step 4. To a mixture of (6 3 S,4S)-4-amino-1 2 -iodo-10,10-dimethyl-6 1 ,6 2 ,6 3 , 6 4 ,6 s ,6 6 -hexahydro- 1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-5,7-dione (1.0 g, 1 .7 mmol) in DMF (15 mL) at 0 °C under an atmosphere of Nz were added DIPEA (2.20 g, 17.0 mmol) and (2S)-2- [[(benzyloxy)carbonyl](methyl)amino]-3-methylbutanoic acid (0.90 g, 3.4 mmol) in portions, followed by 15 COMU (1.10 g, 2.6 mmol) in portions over 10 min. The mixture was stirred at 0 °C for 1.5 h, then diluted with HzO and the mixture was extracted with EtOAc (300 mL x 3). The combined organic layers were washed with brine (100 mL x 3), dried over anhydrous NazSO* and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by preparative-HPLC to give benzyl ((2S)-1 - (((6 3 S,4S)-1 2 -iodo-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- 20 pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)(methyl)carbamate (790 mg, 53% yield) as a solid.

Step 5. To a mixture of benzyl ((2S)-1 -(((6 3 S,4S)-1 2 -iodo-10,10-dimethyl-5,7-dioxo- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 e -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane- 4-yl)amino)-3-methyl-1-oxobutan-2-yl)(methyl)carbamate (480 mg, 0.58 mmol) and fert-butyl 4-[6-[(1 S)-1- 25 methoxyethyl]-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-3-yl]piperazine-1 -carboxylate (309 mg, 0.69 mmol) in 1 ,4-dioxane (8.0 mL) and HzO (1.6 mL) under an atmosphere of Ar were added K2CO3 (199 mg, 1 .4 mmol) and Pd(dppf)Clz (42 mg, 0.06 mmol) in portions. The mixture was heated to 70 °C and stirred for 16 h, then diluted with HzO and extracted with EtOAc (200 mL x 3). The combined organic layers were washed with brine (150 mL x 3), dried over anhydrous NazSO* and filtered. The filtrate was 30 concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give fert-butyl 4-(5-((6 3 S,4S)-4-((S)-2-(((benzyloxy)carbonyl)(methyl)amino)-3- methylbutanamido)- 10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-1 2 -yl)-6-((S)-1 -methoxyethyl)pyridin-3-yl)piperazine-1 -carboxylate (335 mg, 51% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C58H74N8O9 1026.6; found 1027.4.

35 Step 6. To a mixture of fert-butyl 4-(5-((6 3 S,4S)-4-((S)-2-(((benzyloxy)carbonyl)(methyl)amino)-3- methylbutanamido)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-1 2 -yl)-6-((S)-1 -methoxyethyl)pyridin-3-yl)piperazine-1 - carboxylate (335 mg, 0.33 mmol) in DMF (5 mL) at 0 °C under an atmosphere of Nz were added CszCOs (234 mg, 0.72 mmol) and iodoethane (102 mg, 0.65 mmol) in portions. The mixture was warmed to room 40 temperature and stirred for 16 h, then diluted with HzO and the mixture extracted with EtOAc (100 mL x 3). The combined organic layers were washed with brine (50 mL x 3), dried over anhydrous NazS04 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by

902

SUBSTITUTE SHEET (RULE 26) preparative-TLC to give fert-butyl 4-(5-((6 3 S,4S)-4-((S)-2-(((benzyloxy)carbonyl)(methyl)amino)-3- methylbutanamido)-1 1 -ethyl-10,10-dimethyl-5,7-d ioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-1 2 -yl)-6-((S)-1 -methoxyethyl)pyridin-3- yl)piperazine-1 -carboxylate (320 mg, 84% yield) as a light yellow solid. LCMS (ESI): m/z [M+H] calc’d for

5 CeoHyeNsOg 1054.6; found 1055.8.

Step 7. A mixture of fert-butyl 4-(5-((6 3 S,4S)-4-((S)-2-(((benzyloxy)carbonyl)(methyl)amino)-3- methylbutanamido)-1 1 -ethyl-10,10-dimethyl-5,7-d ioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-1 2 -yl)-6-((S)-1 -methoxyethyl)pyridin-3- yl)piperazine-1 -carboxylate (320 mg) in xx M HCI in 1 ,4-dioxane (3.0 mL) at 0 °C under an atmosphere of

10 Nz was stirred at room temperature for 2 h, then concentrated under reduced pressure to give benzyl ((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)-5-(piperazin-1 -yl)pyridin-3-yl)-10,10-dimethyl-5,7- dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)(methyl)carbamate, which was used directly in the next step further purification. LCMS (ESI): m/z [M+H] calc’d for CssHyoNeO? 954.5; found

15 955.3.

Step 8. To a mixture of benzyl ((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)-5- (piperazin-1 -yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamate (320 mg, 0.34 mmol) and HCHO (60 mg, 2.0 mmol) in MeOH (3.0 mL) at 0 °C

20 under an atmosphere of Nz were added NaCNBHs (42 mg, 0.67 mmol) and AcOH (60 mg, 1.0 mmol) in portions. The mixture was warmed to room temperature and stirred for 2 h, then diluted with H 2 O and the mixture extracted with DCM / MeOH (5:1) (200 mL x 3). The combined organic layers were washed with brine (100 mL x 3), dried over anhydrous NazSO* and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by preparative-HPLC to give benzyl ((2S)-1-(((6 3 S,4S)-1 1 -

25 ethyl-1 2 -(2-((S)-1 -methoxyethyl)-5-(4-methylpiperazin-1 -yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 e -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane- 4-yl)amino)-3-methyl-1-oxobutan-2-yl)(methyl)carbamate (160 mg, 59% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for CseHyzNeOy 968.6; found 969.6.

Step 9. To a mixture of benzyl ((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)-5-(4-

30 methylpiperazin-1 -yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 ' H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamate (160 mg, 0.17 mmol) in toluene (10 mL) and MeOH (1.0 mL) was added Pd/C (130 mg, 1 .2 mmol) in portions. The mixture was evacuated and re-filled with Ha (x 3), then stirred under an atmosphere of Hz for 16 h. The mixture was filtered and the filtrate was concentrated under reduced

35 pressure to give (2S)-N-((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)-5-(4-methylpiperazin-1 -yl)pyridin-3- yl)-10,10-d imethyl-5 ,7-d ioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)butana mide (140 mg), which was used directly in the next step without further purification. LCMS (ESI): m/z [M+H] calc'd for C^HeeNeOs 834.5; found 835.5.

40 Step 10. To a mixture of (2S)-N-((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)-5-(4- methylpiperazin-1 -yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)bu tanamide

903

SUBSTITUTE SHEET (RULE 26) (140 mg, 0.17 mmol) in ACN (2.0 mL) at 0 °C under an atmosphere of Na were added DIPEA (433 mg, 3.35 mmol), (3S)-1-(prop-2-enoyl)pyrrolidine-3-carboxylic acid (57 mg, 0.34 mmol) in portions and GIF (70 mg, 0.25 mmol) in portions over 10 min. The mixture was stirred at 0 °C for 1.5 h, then H 2 O added and the mixture extracted with EtOAc (150 mL x 3). The combined organic layers were washed with brine 5 (100 mL x 3), dried over anhydrous NaaSO* and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by preparative-HPLC to give two atropisomers of (3S)-1 -acryloyl-N- ((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)-5-(4-methylpiperazin-1 -yl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)-N-methylpyrrolidine-3-carboxamide 10 (40 mg, 24% yield) as a solid and (20 mg, 12% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for

C56H75N9O7985.6; found 986.7; Ή NMR (400 MHz, DMSO-db) 8.47 (t, J= 2.1 Hz, 1H), 8.00 (d, J= 4.7 Hz, 1H), 7.78 - 7.59 (m, 3H), 7.58 - 7.48 (m, 1H), 7.42 - 7.30 (m, 1H), 7.23 (dq, J= 8.0, 4.0, 3.5 Hz, 1H), 7.15 - 7.03 (m, 1 H), 6.75 - 6.50 (m, 1 H), 6.18 (dt, J = 16.8, 2.7 Hz, 1 H), 5.70 (tt, J = 9.3, 2.7 Hz, 1 H), 5.48

- 5.23 (m, 1H), 5.06 (dd, J= 31.1 , 12.3 Hz, 1H), 4.74 (dd, J= 11.0, 4.3 Hz, 1H), 4.33 - 4.15 (m, 2H), 4.01

15 (ddd, J = 36.1 , 12.6, 7.6 Hz, 2H), 3.91 - 3.56 (m, 6H), 3.52 - 3.39 (m, 2H), 3.31 - 3.28 (m, 2H), 3.24 (d, J =

5.7 Hz, 4H), 3.06 (s, 4H), 2.93 (d, J= 9.8 Hz, 2H), 2.81 (d, J= 5.4 Hz, 3H), 2.47 - 2.43 (m, 4H), 2.22 (s, 4H), 2.09 (tq, J = 12.0, 7.4, 6.6 Hz, 3H), 1.81 (s, 1 H), 1.74 (d, J = 11.7 Hz, 1 H), 1.56 (d, J = 11.7 Hz, 1 H),

1.20 (dd, J = 6.3, 1.5 Hz, 3H), 1.10 (td, J = 7.2, 2.4 Hz, 3H), 1.00 - 0.86 (m, 6H), 0.86 - 0.72 (m, 3H), 0.54 (d, J= 3.5 Hz, 3H) and LCMS (ESI): m/z [M-H] calc'd for C56H75N9O7985.6; found 984.4; Ή NMR (400 20 MHz, DMSO-Ob) 68.46 (d, J = 3.0 Hz, 2H), 7.98 (s, 1 H), 7.89 - 7.83 (m, 1 H), 7.76 - 7.57 (m, 3H), 7.24 (s, 2H), 7.07 (s, 1H), 6.70 - 6.58 (m, 1H), 6.17 (d, J= 16.5 Hz, 1H), 5.73 - 5.67(m, 1H), 5.36 - 5.30 (m, 1H), 4.31 - 3.97 (m, 6H), 3.83 - 3.77 (m, 2H), 3.74 - 3.49 (m, 6H), 3.48 - 3.41 (m, 1 H), 3.40 - 3.37 (m, 2H) 3.28

- 3.24 (m, 4H), 3.07 (s, 3H), 2.88 - 2.82 (m, 1 H), 2.80 - 2.64 (m, 7H), 2.49 - 2.44 (m, 4H), 2.22 (s, 3H), 2.04 (d, J= 26.1 Hz, 3H), 1.85 - 1.79 (m, 1H), 1.67 - 1.55 (m, 2H), 1.35 (d, J= 6.1 Hz, 3H), 1.27 - 1.22

25 (m, 1 H), 1 .05 - 0.93 (m, 4H), 0.89 (d, J = 6.9 Hz, 2H), 0.79 (d, J = 12.4 Hz, 5H), 0.73 (d, J = 6.5 Hz, 1 H),

0.56 (s, 3H).

904

SUBSTITUTE SHEET (RULE 26) Example A542. The synthesis of 1-(4-(dlmethylamino)-4-methylpent-2-ynoyl)-4-fluoro-/V· ((2SM -(((6 3 S,4S)-1 2 -(2-((S)-1 -methoxyethyl)-5-(4-methylplperazln-1 -yl)pyrldln-3-yl)-10,10-dimethyl- 5,7-dioxo-1 1 -(2,2,2-trlfluoroethyl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 I 6 e -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1,3)-benzsnacycloundecaphans-4-yl)amino)-3-msth yl-1-oxobutan-2-yl)-N- 5 methylpiperldlne-4-carboxamlde

Step 1. To a solution of (S)-3-bromo-5-iodo-2-(1 -methoxyethyl)pyridine (15 g, 43.86 mmol), and benzyl piperazine-1 -carboxylate (8.7 g, 39.48 mmol) in toluene (150 mL) at 0 °C, were added cesium carbonate (71.46 g, 219.32 mmol), BINAP (0.55 g, 0.88 mmol) and palladium acetate (0.49 g, 2.19 mmol)

10 in portions. The reaction mixture was stirred at 90 °C for 12 h under an argon atmosphere. The resulting mixture was cooled down to room temperature, filtered and the filter cake was washed with EtOAc (150 mL x 3). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford benzyl (S)-4-(5-bromo-6-(1 -methoxyethyl)pyridin-3-yl)piperazine-1 - carboxylate (16 g, 84% yield) as solid. LCMS (ESI): m/z [M+H] calc’d for C^HseNeO 433.1 ; found 434.0.

905

SUBSTITUTE SHEET (RULE 26) Step 2. To a stirred solution of benzyl (S)-4-(5-bromo-6-(1-methoxyethyl)pyridin-3-yl)piperazine- 1 -carboxylate (22.7 g, 52.26 mmol), bis(pinacolato)diboron (19.91 g, 78.4 mmol) in toluene (230 mL) at 0 °C, were added potassium acetate (12.82 g, 130.66 mmol) and Pd(dppf)Cl 2 -DCM (4.26 g, 5.23 mmol) in portions. The reaction mixture was stirred at 90 °C for 6 h under an argon atmosphere. The resulting 5 mixture was filtered and the filter cake was washed with EtOAc (200 mL x 3). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford benzyl (S)-4-(6-(1 -methoxyethyl)-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-3- yl)piperazine-1 -carboxylate (14.7 g, 58% yield) as solid. LCMS (ESI): m/z [M+H] calc’d for CaeHseBNsOs 481.3; found 482.3.

10 Step 3. To a stirred solution of 5-bromo-3-(3-((fe/t-butyldiphenylsilyl)oxy)-2,2-dimethylprop yl)-2- iodo-1 //-indole (17.46 g, 27 mmol) in 1 ,4-dioxane (150 mL) and H 2 O (30 mL) at 0 °C, were added potassium carbonate (9.33 g, 67.51 mmol) and Pd(dppf)Cl 2 DCM (2.2 g, 2.7 mmol) in portions, followed by benzyl (S)-4-(6-(1 -methoxyethyl)-5-(4, 4, 5, 5-tetramethyl-1 , 3, 2-dioxaborolan-2-yl) pyridin-3-yl) piperazine-1 -carboxylate (13 g, 27 mmol). The reaction mixture was stirred at 70 °C for 12 h under an 15 argon atmosphere. The resulting mixture was cooled to room temperature and quenched with H 2 O, then extracted with EtOAc (200 mL x 3). The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford benzyl (S)-4-(5-(5-bromo-3-(3-((fert-butyldiphenylsilyl)oxy)-2,2-di methylpropyl)-1 H- indol-2-yl)-6-(1 - methoxyethyl)pyridin-3-yl)piperazine-1 -carboxylate (20 g, 84.7% yield) as solid. LCMS (ESI): m/z [M+H] 20 calc’d for C49Hs7BN 4 04Si 873.2; found 873.3.

Step 4. To a mixture of benzyl (S)-4-(5-(5-bromo-3-(3-((tert-butyldiphenylsilyl)oxy)-2,2- dimethylpropyl)-1 //-indol-2-yl)-6-(1-methoxyethyl)pyridin-3-yl)piperazine-1 -carboxylate (19 g, 21.74 mmol) and CS2CO3 (49.58 g, 152.17 mmol) in DMF (190 mL) at 0 °C under argon atmosphere, was dropwise added 2,2,2-trifluoroethyl trifluoromethanesulfonate (50.46 g, 217.39 mmol). The reaction mixture was 25 stirred at room temperature for 12 h under an argon atmosphere, then quenched with H 2 O, extracted with EtOAc (200 mL x 3). The combined organic layers were washed with brine (200 mL x 3), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford benzyl (S)-4-(5-(5-bromo-3-(3-((tert-butyldiphenylsilyl)oxy)-2,2- dimethylpropyl)-1 -(2,2,2-trifluoroethyl)-1 H- indol-2-yl)-6-(1 -methoxyethyl)pyridin-3-yl)piperazine-1 - 30 carboxylate (17.6 g, 84.7% yield) as solid. LCMS (ESI): m/z [M+H] calc'd for CsiHseBF3N404Si 954.2; found 955.3.

Step 5. To a solution of benzyl (S)-4-(5-(5-bromo-3-(3-((terf-butyldiphenylsilyl)oxy)-2,2- dimethylpropyl)-1 -(2,2,2-trifluoroethyl)-1 H- indol-2-yl)-6-(1 -methoxyethyl)pyridin-3-yl)piperazine-1 - carboxylate (18 g, 18.83 mmol), was added TBAF in THF (180.0 mL) at 0 °C. The reaction mixture was

35 stirred at 40 °C for 12 h under an argon atmosphere, then quenched with cold H 2 O. The resulting mixture was extracted with EtOAc (200 mL x 3). The combined organic layers were washed with brine (200 mL x 3), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford benzyl (S)-4-(5-(5-bromo-3-(3-hydroxy-2,2- dimethylpropyl)-1 -(2,2,2-trifluoroethyl)-1 H- indol-2-yl)-6-(1 -methoxyethyl)pyridin-3-yl)piperazine-1 -

40 carboxylate (7.8 g, 57.7% yield) as solid. LCMS (ESI): m/z [M+H] calc’d for CssHwBrFaN^* 716.2; found 717.1.

906

SUBSTITUTE SHEET (RULE 26) Step 6. A solution of benzyl (S)-4-(5-(5-bromo-3-(3-hydroxy-2,2-dimethylpropyl)-1 -(2,2,2- trifluoroethyl)-1 A/-indol-2-yl)-6-(1 -methoxyethyl)pyridin-3-yl)piperazine-1 -carboxylate (1 g, 1.39 mmol) in 1 ,4-dioxane (10 mL) and H 2 O (2 mL), was added methyl (S)-1-((S)-2-((te/t-butoxycarbonyl)amino)-3-(3- (4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)propanoyl)hexahydropyridazine- 3-carboxylate (1.08 5 g, 2.09 mmol), potassium carbonate (481.47 mg, 3.48 mmol) and Pd(dtbpf)Cl 2 (181.64 mg, 0.28 mmol) in portions at 0 °C. The reaction mixture was stirred at 70 °C for 3 h under an argon atmosphere. The resulting mixture was cooled to room temperature, then quenched with H 2 O and extracted with EtOAc (50 mL x 3). The combined organic layers were washed with brine (20 mL x 3), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel chromatography 10 to afford methyl (S)-1 -((S)-3-(3-(2-(5-(4-((benzyloxy)carbonyl)piperazin-1 -yl)-2-((S)-1 - methoxyethyl)pyridin-3-yl)-3-(3-hydroxy-2,2-dimethylpropyl)- 1-(2,2,2-trifluoroethyl)-1 H-indol-5-yl)phenyl)- 2-((tert-butoxycarbonyl)amino)propanoyl)hexahydropyridazine- 3 -carboxylate (1.1 g, 77% yield) as solid. LCMS (ESI): m/z [M+H] calc’d for CssHeeFsNyOg 1027.5; found 1028.3.

Step 7. To a solution of methyl (S)-1-((S)-3-(3-(2-(5-(4-((benzyloxy)carbonyl)piperazin-1-yl )-2- 15 ((S)-1 -methoxyethyl)pyridin-3-yl)-3-(3-hydroxy-2,2-dimethylpropyl) -1 -(2,2,2-trifluoroethyl)-1 H-indol-5- yl)phenyl)-2-((fert-butoxycarbonyl)amino)propanoyl)hexahydro pyridazine-3-carboxylate (1.1 g, 1.07 mmol) in THE (8 mL) and H 2 O (2 mL) at 0 °C, was dropwise added LiOH (2.2 mL, 1 M aqueous) under an argon atmosphere. The reaction mixture was stirred for 2 h then concentrated under reduced pressure. The residue was acidified to pH 5 with citric acid (1 M) and extracted with EtOAc (20 mL x 3). The 20 combined organic layers were concentrated under reduced pressure. The residue was purified by reverse phase chromatography to afford (S)-1 -((S)-3-(3-(2-(5-(4-((benzyloxy)carbonyl)piperazin-1 -yl)-2-((S)-1 - methoxyethyl)pyridin-3-yl)-3-(3-hydroxy-2,2-dimethylpropyl)- 1-(2,2,2-trifluoroethyl)-1 H-indol-5-yl)phenyl)- 2-((ferf-butoxycarbonyl)amino)propanoyl)hexahydropyridazine- 3-carboxylic acid (750 mg, 69% yield) as solid. LCMS (ESI): m/z [M+H] calc’d for CS4H66F3N7O9 1013.5; found 1014.3.

25 Step 8. To a solution of (S)-1 -((S)-3-(3-(2-(5-(4-((benzyloxy)carbonyl)piperazin-1 -yl)-2-((S)-1 - methoxyethyl)pyridin-3-yl)-3-(3-hydroxy-2,2-dimethylpropyl)- 1-(2,2,2-trifluoroethyl)-1 H- indol-5-yl)phenyl)- 2-((tert-butoxycarbonyl)amino)propanoyl)hexahydropyridazine- 3-carboxylic acid (0.75 g, 0.74 mmol) in DCM (75 mL) at 0 °C, were added in portions HOST (0.5 g, 3.7 mmol), DIPEA (3.82 g, 29.58 mmol), and EDCI (4.25 g, 22.19 mmol) at 0 °C. The reaction mixture was stirred at room temperature for 12 h under 30 an argon atmosphere. The resulting mixture was concentrated under reduced pressure and extracted with EtOAc (100 mL x 3). The combined organic layers were washed with brine (50 mL x 3), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel chromatography to afford benzyl 4-(5-((6 3 S,4S)-4-((tert-butoxycarbonyl)amino)-10,10-dimethyl-5, 7- dioxo-1 1 -(2,2,2-trifluoroethyl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 /+8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- 35 benzenacycloundecaphane-1 2 -yl)-6-((S)-1 -methoxyethyl)pyridin-3-yl)piperazine-1 -carboxylate (0.5 g, 67.9% yield) as solid. LCMS (ESI): m/z [M+H] calc’d for CsAHetFaNTOe 995.5; found 996.3.

Step 9. To a mixture of benzyl 4-(5-((6 3 S,4S)-4-((tert-butoxycarbonyl)amino)-10,10-dimethyl-5, 7- dioxo-1 1 -(2,2,2-trifluoroethyl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 /+8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-1 2 -yl)-6-((S)-1 -methoxyethyl)pyridin-3-yl)piperazine-1 -carboxylate (500 mg,

40 0.5 mmol) in MeOH (15 mL) at 0 °C, was added paraformaldehyde (135.64 mg, 1.5 mmol), and Pd/C

(750 mg) in portions. The reaction mixture was stirred at room temperature for 12 h under a hydrogen atmosphere. The resulting mixture was filtered and the filter cake was washed with EtOAc (50 mL x 5).

907

SUBSTITUTE SHEET (RULE 26) The filtrate was concentrated under reduced pressure. The residue was purified by silica gel chromatography to afford tert-butyl ((6 3 S,4S)-1 2 -(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyr idin-

3-yl)-10,10-dimethyl-5,7-dioxo-1 ^(Z.S.Z-trifluoroethylJ-e 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (350 mg, 79.6% yield) as an

5 solid. LCMS (ESI): m/z [M+H] calc’d for CtyHeoFaNyOe 875.5; found 876.5.

Step 10. To a solution of tert-butyl ((6 3 S,4S)-1 2 -(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1- yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-1 , -(2,2,2-trifluoroethyl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 ' H- 8-oxa- 1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)carbamate (300 mg, 0.34 mmol) in DCM (2 mL) at 0 °C, was dropwise added HCI in 1 ,4-dioxane (1 mL, 4M, 4 mmol). The reaction mixture 10 was stirred at room temperature for 2 h, then concentrated under reduced pressure to give (6 3 S,4S)-4- amino-1 2 -(2-((S)-1 -methoxyethyl)-5-(4-methylpiperazin-1 -yl)pyridin-3-yl)-10,10-dimethyl-1 1 -(2,2,2- trifluoroethyl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 AA8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione hydrochloride (350 mg, crude) as solid. LCMS (ESI): m/z [M+H] calc’d for C42H52F3N7O4775.4; found 766.4.

15 Step 11. To a solution of (6 3 S,4S)-4-amino-1 2 -(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1- yl)pyridin-3-yl)-10,10-dimethyl-1 1 -(2,2,2-trifluoroethyl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-5,7-dione hydrochloride (150 mg, 0.19 mmol) and N- (1 -(4-(dimethylamino)-4-methylpent-2-ynoyl)-4-fluoropiperidine -4-carbonyl)-W-methyl-L-valine (154 mg, 0.39 mmol) in DMF (2 mL) at 0 °C, was dropwise added a mixture of DIPEA (1 g, 7.72 mmol) and HATU 20 (110 mg, 0.29 mmol) in DMF (0.2 mL). The reaction mixture was stirred at 0 °C for 2 h under an argon atmosphere, then quenched with H 2 O. The resulting mixture was extracted with EtOAc (20 mL x 3). The combined organic phase was washed with brine (10 mL x 3), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by reverse phase chromatography to afford 1 -(4-(dimethylamino)-4-methylpent-2-ynoyl)-4-fluoro-A/-((2S)- 1 -(((6 3 S,4S)-1 2 -(2-((S)-1 - 25 methoxyethyl)-5-(4-methylpiperazin-1 -yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-1 1 -(2,2,2-trifluoroethyl)- 6 1 ,6 2 , 6 3 ,6 4 ,6 5 ,6 e -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-

4-yl)amino)-3-methyl-1-oxobutan-2-yl)-N- methylpiperidine-4-carboxamide (39.5 mg, 17% yield) as solid.

1 H NMR (400 MHz, DMSO-dfe) δ 8.48 (d, J= 2.9 Hz, 1 H), 8.32 (t, J= 7.3 Hz, 1 H), 7.97 (s, 1 H), 7.82-7.69 (m, 3H), 7.66 (t, J= 7.8 Hz, 1H), 7.33-7.07 (m, 3H), 5.50 (dd, J= 16.7, 8.6 Hz, 1H), 5.33 (t, J= 9.2 Hz,

30 1 H), 5.16 (d, J = 12.2 Hz, 1 H), 4.94-4.80 (m, 1 H), 4.64 (d, J= 10.8 Hz, 1 H), 4.33-4.16(m, 3H), 4.12-4.02

(m, 2H), 3.71-3.50 (m, 3H), 3.25 (s, 3H), 3.21-3.16 (m, 3H), 3.14-3.05 (m, 1H), 2.96 (t, J= 4.7 Hz, 4H), 2.84 (s, 1H), 2.82-2.72 (m, 2H), 2.59-2.53 (m, 1H), 2.47-2.40 (m, 4H), 2.22 (d, J= 2.9 Hz, 9H), 2.18-2.12 (m, 2H), 2.11 -1 .99 (m, 3H), 1.87-1.78 (m, 1 H), 1.74-1 ,62(m, 1 H), 1 .59-1.48 (m, 1 H), 1.40-1 .32 (m, 9H), 1.01 (t, J= 7.7 Hz, 1H), 0.89 (s, 5H), 0.83 (d, J= 6.3 Hz, 1H), 0.77 (d, J= 6.6 Hz, 2H), 0.38 (s, 3H).

35 LCMS (ESI): m/z [M+H] calc’d for C^HseNeO? 1154.6; found 1155.7.

908

SUBSTITUTE SHEET (RULE 26) Example A735. Synthesis of 2-acryloyl-N-((2S)-1-(((6 3 S,4S v Z)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)pyrldin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)- thlazola-1 (5,3)-indola-6(1 ,3)-pyrldazlnacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)- N-methyl-5-oxa-2,9-dlazasplro[3.5]nonane-9-carboxamlde

5

Step 1. To a stirred mixture of BTC (425.2 mg, 1.448 mmol) in DCM (10 mL) was added dropwise pyridine (1.04 g, 13.16 mmol) and ferf-butyl 5-oxa-2,9-diazaspiro[3.5]nonane-2-carboxylate (1 g, 4.39 mmol), the reaction mixture was stirred at room temperature for 2 h. The resulting mixture was concentrated under reduced pressure to give crude ferf-butyl 9-(chlorocarbonyl)-5-oxa-2,9- 10 diazaspiro[3.5]nonane-2-carboxylate.

Step 2. To a stirred solution of ferf-butyl 9-(chlorocarbonyl)-5-oxa-2,9-diazaspiro[3.5]nonane-2- carboxylate (2.5 g, crude) in MeCN (20 mL) were added dropwise pyridine (1.04 g, 13.16 mmol) and benzyl (2S)-3-methyl-2-(methylamino)butanoate (970.66 mg, 4.38 mmol) at room temperature. The reaction mixture was stirred at 80 °C for 12 h and concentrated under reduced pressure. The residue was 15 purified by silica gel column chromatography to afford ferf-butyl (S)-9-((1 -(benzyloxy)-3-methyl-1 - oxobutan-2-yl)(methyl)carbamoyl)-5-oxa-2,9-diazaspiro[3.5]no nane-2-carboxylate (783 mg, 37.6% yield, two steps) as an oil. LCMS (ESI): m/z [M+H] calc’d for C25H37N3O6475.3; found 476.3.

Step 3. A solution of ferf-butyl (S)-9-((1 -(benzyloxy)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamoyl)-5-oxa-2,9-diazaspiro[3.5]nonane-2-carb oxylate (783 mg, 1.65 mmol) and 10 wt% 20 palladium on carbon (226.29 mg) in THF (10 mL) was stirred for 2 h at 50 °C under a hydrogen atmosphere. The resulting mixture was cooled to room temperature, filtered and the filter cake was washed with MeCN (10 mL x 3). The filtrate was concentrated under reduced pressure to give N-{2-(tert- butoxycarbonyl)-5-oxa-2,9-diazaspiro[3.5]nonane-9-carbonyl)- N-methyl-L-valine (591 mg, 98.8% yield) as solid. LCMS (ESI): m/z [M+H] calc’d for CieHsiNsOe 385.2; found 386.3.

909

SUBSTITUTE SHEET (RULE 26) Step 4. To a stirred solution of intermediate 2 (731 mg, 1.16 mmol) and DIPEA (2.25 g, 17.38 mmol) in MeCN (50 mL) was added GIF (644.31 mg, 2.32 mmol) and A/-(2-(fert-butoxycarbonyl)-5-oxa- 2,9-diazaspiro[3.5]nonane-9-carbonyl)-N-methyl-L-valine (446.68 mg, 1.16 mmol) at room temperature. The reaction mixture was stirred for 2 h then concentrated under reduced pressure. The residue was 5 purified by silica gel column chromatography to afford tert-butyl 9-(((2S)-1 -(((6 3 S,4S,Z)-1 1 -ethyl-1 2 -(2-((S)- 1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-2(4,2)- thiazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamoyl)-5-oxa-2,9-diazaspiro[3.5]nonane-2-carb oxylate (752 mg, 65% yield) as solid.

LCMS (ESI): m/z [M+H] calc’d for C52H71N9O9S 997.5; found 996.6.

10 Step 5. To a stirred solution of tert-butyl 9-(((2S)-1 -(((6 3 S,4S,2)-1 1 -ethyl-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-2(4,2)- thiazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamoyl)-5-oxa-2,9-diazaspiro[3.5]nonane-2-carb oxylate (752 mg, 0.75 mmol) in DCM (40 mL) was added TEA (10 mL) in portions at room temperature. The reaction mixture was stirred for 2 h,

15 then concentrated under reduced pressure. To the residue was added saturated aqueous sodium bicarbonate (100 mL) and DCM (100 mL). The aqueous layer was separated and extracted with DCM (100 mL x 2). The combined organic phase was dried over anhydrous sodium sulfate, filtrated and concentrated under reduced pressure to afford N-((2S)-1-(((6 3 S,4S,Z)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-2(4,2)- 20 thiazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-yl)-N- methyl-5-oxa-2,9-diazaspiro[3.5]nonane-9-carboxamide (587 mg, 87% yield) as solid. LCMS (ESI): m/z [M+H] calc’d for C47H63N9O7S 897.5; found 898.4.

Step 6. A stirred solution of N-((2S)-1 -(((6 3 S,4S,Z)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3- yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-2(4,2)-thiazola-1 (5,3)-indola-6(1 ,3)- 25 pyridazinacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2 -yl)-N-methyl-5-oxa-2,9- diazaspiro[3.5]nonane-9-carboxamide (586 mg, 0.65 mmol) in MeCN (10 mL) was added acrylic acid (47 mg, 0.65 mmol), DIPEA (421 mg, 3.26 mmol), CIP (362 mg, 1.3 mmol). The reaction mixture was stirred for 12 h and concentrated under reduced pressure. The residue was purified by reverse phase chromatography to afford 2-acryloyl-N-((2S)-1 -(((6 3 S,4S,Z)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3- 30 yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 ' H- 8-oxa-2(4,2)-thiazola-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2 -yl)-N-methyl-5-oxa-2,9- diazaspiro[3.5]nonane-9-carboxamide (194 mg, 27% yield) as a solid. Ή NMR (400 MHz, DMSO-db) 6 8.78 (dd, J= 4.8, 1.7 Hz, 1 H), 8.48 (d, J = 1.6 Hz, 2H), 7.85-7.65 (m, 3H), 7.65-7.42(m,2H), 6.30 (mm,1H), 6.10 (m, J= 17.0, 1H), 5.78-5.50 (m, J= 10.3, 2H), 5.10 (dd, 1 H), 4.40-3.80 (m, 14H), 3.60 - 35 3.10 (m, 10H), 2.94 (d, J = 14.5 Hz, 1 H), 2.85 (s, 4H), 2.42 (dd, 1 H), 2.07 (dd, 2H), 1.80 (s, 2H), 1 .55 (s,

3H), 1.32 (d, 3H), 0.95 - 0.75 (m, 12H), 0.33 (s, 3H). LCMS (ESI): m/z [M+H] calc'd for C50H65N9O8S 951.5; found 952.6.

910

SUBSTITUTE SHEET (RULE 26) Example A720. Synthesis of (2fl)-2-(((1-(4-(dimethylamlno)-4-methylpent-2-ynoyl)azetldl n- 3-yl)oxy)methyl)-N- ((6 3 S,4¾2)-1 1 -ethyH 2 -(2-((S)-1 -met hoxyethyl)pyridln-3-yl)-2 1 , 10,10-trlmethyl· 5,7-dtoxo-e 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H, 2 1 H- 8-oxa-1 (5,3)-lndola-6(1 ,3)-pyridazina-2(5,3)- trlazolacyck>undecaphane-4-yl)-3-methylbutanamlde

5

Step 1. To a stirred solution of methyl 1 -methyl-1 ,2,4-triazole-3-carboxylate (7.0 g, 49.60 mmol) in CCU (70. mL) was added NBS (13.24 g, 74.40 mmol) and AIBN (11.40 g, 69.44 mmol) in portions at 25 °C under an argon atmosphere. The resulting mixture was stirred for 24 h at 80 °C. The resulting mixture was filtered, the filtrate was cooled to 20 °C and kept at 20 °C for 30 min. The resulting mixture was

10 filtered. The filter cake was washed with H 2 O (3 x 50 mL) and pet. ether (3 x 100 mL). The filter cake was dried under reduced pressure. This resulted in methyl 5-bromo-1 -methyl-1 ,2,4-triazole-3-carboxylate (10 g, crude) as a light yellow solid. LCMS (ESI): m/z [M+H] calc’d for CsHeBrNaOa 219.0; found 219.9.

Step 2. To a stirred solution of methyl 5-bromo-1 -methyl-1 ,2,4-triazole-3-carboxylate (10.0 g, 45.50 mmol) in MeOH (150.0 mL) and H 2 O (30.0 mL) was added NaBhU (6.88 g, 181.80 mmol) in

15 portions at -5 °C under a nitrogen atmosphere. The resulting mixture was stirred for 2 h at 0~10 °C. Desired product could be detected by LCMS. The reaction was quenched with brine (100 mL) at 0 °C.

The resulting mixture was extracted with pet. ether (100 mL). The aqueous layer was separated and filtered. The filter cake was washed with MeOH (2 x 50 mL). The filtrate was concentrated under reduced pressure to afford (5-bromo-1 -methyl-1 ,2,4-triazol-3-yl)methanol (6 g, crude) as a light yellow solid. LCMS

20 (ESI): m/z [M+H] calc’d for CAHeBrNaO 191.98; found 192.0.

Step 3. A solution of (5-bromo-1 -methyl-1 ,2,4-triazol-3-yl)methanol (6.0 g) and HBr in AcOH (144.0 mL) was stirred for overnight at 80 °C .The mixture was neutralized to pH 9 with saturated NaHCOs (aq.). The resulting mixture was extracted with EtOAc (3 x 60 mL). The combined organic layers were washed with brine (3x100 mL), dried over anhydrous NazSOA. After filtration, the filtrate was

911

SUBSTITUTE SHEET (RULE 26) concentrated under reduced pressure. This resulted in 5-bromo-3-(bromomethyl)-1 -methyl-1 ,2,4-triazole (6 g, crude) as a white solid. LCMS (ESI): m/z [M+H] calc’d for CAHsBraNs 253.89; found 253.8.

Step 4. To a stirred mixture of 5-bromo-3-(bromomethyl)-1 -methyl-1 ,2,4-triazole (6.0 g, 23.54 mmol) and fert-butyl 2-[(diphenylmethylidene)amino]acetate (6.95 g, 23.54 mmol) in toluene (42 mL) and

5 DCM (18.0 mL) was added (2f?,4/ : 7,5S)-1-(anthracen-9-ylmethyl)-5-ethenyl-2-[(S)-(prop- 2-en-1- yloxy)(quinolin-4-yl)methyl]-1 -azabicyclo[2.2.2]octan-1 -ium bromide (1.43 g, 2.35 mmol) in portions at 0 °C under argon atmosphere. The resulting mixture was stirred and KOH (60 mL) in H 2 O was added. The resulting mixture was stirred for 24 h at -10 °C under an argon atmosphere. Desired product could be detected by LCMS. The reaction was quenched with sat. NHXI (aq.) at 0 °C. The resulting mixture was

10 extracted with EtOAc (3 x 10OmL). The combined organic layers were washed with brine (1 x 200 mL), dried over anhydrous NaaSO*. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC to afford tort-butyl (2S)-3-(5-bromo-1 -methyl-1 ,2,4-triazol-3-yl)-2- [(diphenylmethylidene)amino]propanoate (5 g, 38.6% yield) as a yellow oil. LCMS (ESI): m/z [M+H] calc’d for CaiHaiBrN40a 469.12; found 469.1.

15 Step 5. To a stirred solution of tort-butyl (2S)-3-(5-bromo-1 -methyl-1 ,2,4-triazol-3-yl)-2- [(diphenylmethylidene)amino]propanoate (5.0 g, 10.65 mmol) in DCM (50.0 mL) was added TEA (25.0 mL) dropwise at 0 °C under argon atmosphere. The resulting mixture was stirred for 16 h at room temperature under an argon atmosphere. The resulting mixture was concentrated under reduced pressure to afford (2S)-2-amino-3-(5-bromo-1 -methyl-1 ,2,4-triazol-3-yl)propanoic acid (6 g, crude) as a

20 brown oil. LCMS (ESI): m/z [M+H] calc'd for CeHgBrNtOa 249.00; found 249.0.

Step 6. To a stirred solution of (2S)-2-amino-3-(5-bromo-1 -methyl-1 ,2,4-triazol-3-yl)propanoic acid (6.0 g, 24.09 mmol) in THE (36.0 mL) was added NaHCOs (10.14 g, 120.69 mmol), B0C2O (7.89 g, 36.14 mmol) in portions at 0 °C under an argon atmosphere. The resulting mixture was stirred for 16 h at room temperature. Desired product could be detected by LCMS. The resulting mixture was concentrated

25 under reduced pressure. The mixture was purified by reverse phase chromatography to afford (2S)-3-(5- bromo-1 -methyl-1 ,2,4-triazol-3-yl)-2-[(tert-butoxycarbonyl)amino]propanoic acid (3 g, 33.9% yield) as a white solid. LCMS (ESI): m/z [M+H] calc'd for CuHizBrN^ 349.05; found 349.0.

Step 7. To a stirred solution of 2-[[(2Af)-1 -ethyl-2-[2-[(1 S)-1 -methoxyethyl]pyridin-3-yl]-5-(4, 4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)indol-3-yl]methyl]-2-methylpropyl (3S)-1 ,2-diazinane-3-carboxylate

30 (1.0 g, 1.65 mmol) in DMF (10.0 mL) was added DIPEA (4.28 g, 33.08 mmol), (2S)-3-(5-bromo-1 -methyl- 1 ,2,4-triazol-3-yl)-2-[(terf-butoxycarbonyl)amino]propanoic acid (0.69 g, 1.98 mmol) and HATU (0.75 g,

1.99 mmol) in portions at 0 °C. The resulting mixture was stirred for 2 h at 20 °C under an argon atmosphere. Desired product could be detected by LCMS. The resulting mixture was quenched with H 2 O (100 mL) and extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine

35 (3x100 mL), dried over anhydrous NaaSO*. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse phase chromatography to afford 2-[[(2M)-1 -ethyl-2-[2-[(1 S)- 1-methoxyethyl]pyridin-3-yl]-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)indol-3-yl]methyl]-2- methylpropyl (3S)-1 -[(2S)-3-(5-bromo-1 -methyl-1 ,2,4-triazol-3-yl)-2-[(tert- butoxycarbonyl)amino]propanoyl]-1 ,2-diazinane-3-carboxylate (800 mg, 46.5% yield) as a light yellow

40 solid. LCMS (ESI): m/z [M+H] calc’d for C4sHe4BBrNeOe 935.42; found 935.2.

Step 8. To a stirred solution of 2-[[(2M)-1 -ethyl-2-[2-[(1 S)-1 -methoxyethyl]pyridin-3-yl]-5-(4, 4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)indol-3-yl]methyl]-2-methylpropyl (3S)-1 -[(2S)-3-(5-bromo-1 -methyl-

912

SUBSTITUTE SHEET (RULE 26) 1 ,2,4-triazol-3-yl)-2-[(fert-butoxycarbonyl)amino]propanoyl]- 1 ,2-diazinane-3-carboxylate (800.0 mg, 0.86 mmol) in dioxane (10.0 mL) were added K3PO4 (0.45 g, 2.12 mmol), XPhos (122.26 mg, 0.27 mmol), XPhos Pd G3 (0.22 g, 0.27 mmol) and H 2 O (2.0 mL) at room temperature. The resulting mixture was stirred for 3 h at 75 °C under an argon atmosphere. Desired product could be detected by LCMS. The 5 resulting mixture was extracted with EtOAc (3 x 100 mL). The combined organic layers were washed with brine (3 x 60 mL), dried over anhydrous NaaS04. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse phase chromatography to afford fe/t-butyl ((6 3 S,4S,Z)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-2 1 ,10,10-trimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 1 H,2'H-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(5,3)-triazolacycloundecaphane-4- 10 yl)carbamate (400 mg, 56.8% yield) as a light yellow solid. LCMS (ESI): m/z [M+H] calc’d for CagHszNeOe 729.41 ; found 729.3.

Step 9. To a solution of fe/t-butyl ((6 3 S,4S,2)-1 '-ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)- 2 1 ,10,10-trimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H,2 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina- 2(5,3)-triazolacycloundecaphane-4-yl)carbamate (400.0 mg, 0.56 mmol) in DCM (1 mL) was added TFA 15 (0.5 mL). The reaction was stirred for 1 h at room temperature under an argon atmosphere. After concentration, the mixture was neutralized to pH 8 with saturated NaHCOa (aq., 20 mL). The mixture was extracted with DCM (3 x 20 mL). The organic layers were dried over Na 2 SO 4 and concentrated to afford (6 3 S,4S,Z)-4-amino-1 ’-ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-2 1 ,10,10-tri methyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 1 H,2 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(5,3)-triazolacycloundecaphane-5,7-dione 20 (500 mg, crude) as a light yellow solid. ESI-MS m/z = 629.3 [M+H] 4 ; Calculated MW628.3. LCMS (ESI): m/z [M+H] calc’d for C34H44N8O4629.36; found 629.3.

Step 10. To a stirred solution of (6 3 S,4S,Z)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3- yl)-2 1 ,10,10-trimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H, 2 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(5,3)- triazolacycloundecaphane-5,7-dione (170.0 mg, 0.27 mmol) and (fl)-2-(((1 -benzhydrylazetidin-3- 25 yl)oxy)methyl)-3-methylbutanoic acid (114.68 mg, 0.32 mmol) in DMF (5 mL) were added DIPEA (698.86 mg, 5.41 mmol) and HATU (123.36 mg, 0.32 mmol) dropwise at 0 °C under an air atmosphere. The resulting mixture was stirred for 2 h at 0 °C. The resulting mixture was diluted with 25 mL H 2 O. The resulting mixture was extracted with EtOAc (3 x 25 mL). The combined organic layers were washed with brine (3 x 25 mL), dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under 30 reduced pressure to afford (2/7)-2-(((1 -benzhydrylazetidin-3-yl)oxy)methyl)-N-((6 3 S,4S,Z)-1 1 -ethyl-1 2 -(2- ((S)-1 -methoxyethyl)pyridin-3-yl)-2 1 ,10,10-trimethyl-5,7-dioxo-6' ,δ^,δ^,δ 6 - hexahydro-1 ' H, 2' H- 8-oxa- 1 (5,3)-indola-6(1 ,3)-pyridazina-2(5,3)-triazolacycloundecaphane-4-yl)-3-methy lbutanamide (180 mg, crude) as an off-white oil. LCMS (ESI): m/z [M+H] calc’d for CseHegNgOe 964.54; found 964.4.

Step 11. To a stirred solution of (2f7)-2-(((1-benzhydrylazetidin-3-yl)oxy)methyl)-AA((6 3 S,4S,Z)- 35 1 '-ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-2 1 ,10,10-trimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-

1 ' H,2' H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(5,3)-triazolacycloundecaphane-4-yl)-3-methy lbutanamide (180.0 mg, 0.19 mmol) and Pd/C (90.0 mg, 0.85 mmol) in MeOH(10 mL) was added BoczO (81.48 mg, 0.37 mmol) at room temperature under a hydrogen atmosphere. The resulting mixture was stirred overnight at room temperature. The resulting mixture was filtered, the filter cake was washed with MeOH 40 (3x10 mL). The filtrate was concentrated under reduced pressure to afford tert-butyl 3-((2fl)-2-

(((6 3 S,4S,2)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-2' ,10,10-trimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 ® - hexahydro-1 Ή.2' H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(5,3)-triazolacycloundecaphane-4-

913

SUBSTITUTE SHEET (RULE 26) yl)carbamoyl)-3-methylbutoxy)azetidine-1-carboxylate (80 mg, 47.7% yield) as an off-white solid. LCMS (ESI): m/z [M+H] calc’d for 04βΗβ7Ν90β 898.52; found 898.4.

Step 12. To a stirred solution of fert-butyl 3-((2fl)-2-(((6 3 S,4S,Z)-1 '-ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-2' ,10,10-trimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 1 H,2' H- 8-oxa-1 (5,3)-

5 indola-6(1 ,3)-pyridazina-2(5,3)-triazolacycloundecaphane-4-yl)carbamoy l)-3-methylbutoxy)azetidine-1- carboxylate in DCM (2 mL) was added TFA (1.0 mL) dropwise at 0 °C under an air atmosphere. The resulting mixture was stirred for 1 h at 0 °C. The resulting mixture was concentrated under reduced pressure to afford (2fl)-2-((azetidin-3-yloxy)methyl)-N-((6 3 S,4S,2)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-2' ,10,10-trimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 1 H, 2' H -8-oxa-

10 1 (5,3)-indola-6(1 ,3)-pyridazina-2(5,3)-triazolacycloundecaphane-4-yl)-3-methy lbutanamide (85 mg, crude) as a yellow green oil.

Step 13. To a stirred solution of (2fl)-2-((azetidin-3-yloxy)methyl)-N-((6 3 S,4S,Z)-1 ' -ethyl-1 2 -(2- ((S)-1 -methoxyethyl)pyridin-3-yl)-2’ ,10,10-trimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 1 H, 2 1 H- 8-oxa- 1 (5,3)-indola-6(1 ,3)-pyridazina-2(5,3)-triazolacycloundecaphane-4-yl)-3-methy lbutanamide (80.0 mg, 0.10

15 mmol) and 4-(dimethylamino)-4-methylpent-2-ynoic acid (38.90 mg, 0.25 mmol) in DMF (2 mL) were added DIPEA (518.27 mg, 4.01 mmol) and COMU (51 .52 mg, 0.12 mmol) in portions at 0 °C The reaction mixture was stirred under an air atmosphere for 2 h. The crude product (150 mg) was purified by reverse phase chromatography to afford (2fl)-2-(((1-(4-(dimethylamino)-4-methylpent-2-ynoyl)azetidi n-3- yl)oxy)methyl)-W-((6 3 S,4S,Z)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-2’ ,10,10-trimethyl-5,7-dioxo-

20 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H, 2 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(5,3)- triazolacycloundecaphane-4-yl)-3-methylbutanamide (15.3 mg, 16.3% yield) as an off-white solid. Ή NMR (400 MHz, DMSO-cfe) δ 8.77 (dd, J= 4.8, 1.7 Hz, 1H), 8.15 (d, J= 1.7 Hz, 1H), 8.07 (d, J= 8.1 Hz,

1 H), 7.85 - 7.78 (m, 1 H), 7.70 (d, J = 8.6 Hz, 1 H), 7.58 - 7.48 (m, 2H), 5.82 (s, 1 H), 4.95 (d, J = 11.7 Hz, 1H), 4.41 - 4.30 (m, 5H), 4.30 (d, J= 8.2 Hz, 2H), 4.25 (d, J= 5.6 Hz, 4H), 4.10 (td, J= 17.1 , 16.1 , 9.1

25 Hz, 2H), 3.99 - 3.82 (m, 3H), 3.71 - 3.60 (m, 1 H), 3.54 - 3.43 (m, 3H), 3.39 (s, 2H), 3.22 (d, J = 1.6 Hz, 1H), 2.92 (d, J= 13.6 Hz, 1H), 2.86 -2.77 (m, 2H), 2.45 (s, 6H), 2.37 (q, J= 7.7 Hz, 1H), 2.17 (d, J= 6.6 Hz, 2H), 2.03 (d, J = 10.2 Hz, 2H), 1.78 - 1.66 (m, 3H), 1.47 (t, J = 10.9 Hz, 6H), 1.35 - 1.28 (m, 12H), 0.32 (s, 3H). LCMS (ESI): m/z [M+H] calc'd for C51H70N10O7935.55; found 935.3.

914

SUBSTITUTE SHEET (RULE 26) Example A692. Synthesis of (3S)-1-(4-(dlmethylamlno)-4-methylpent-2-ynoyl)-N-((2S)-1- (((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyrldin-3-yl)-10,10-dimethyFSJ-dioxo-e 1 ,6 2 ,6 3 ,6 4 ,6 S ,6 6 - hexahydro-1 1 H-8-oxa-2(5,2)-oxazola-1(5,3)-indola-6(1,3)-pyrldazlna cycloundecaphane-4-yl)amino)- 3-methyl-1-oxobutan-2-yl)-A#-methylpyrrolldine-3-cartooxamld e

Step 1. To a solution of 1 ,3-oxazol-2-ylmethanol (5.0 g, 50.46 mmol) in THF (75 mL), were added imidazole (8.59 mg, 0.13 mmol), and TBSCI (11.41 mg, 0.08 mmol) at 0 °C. The resulting solution was stirred for 5 h then concentrated under reduced pressure. The crude material was purified by silica gel column chromatography to afford 2-[[(tert-butyldimethylsilyl)oxy]methyl]-1 ,3-oxazole (10 g, 92.8%

10 yield) as colorless oil. LCMS (ESI): m/z [M+H] calc'd for CioHigNOaSi 214.13; found 214.3.

Step 2. To a solution of 2-[[(fert-butyldimethylsilyl)oxy]methyl]-1 ,3-oxazole (10.0 g, 46.87 mmol) in THF (150.0 mL, 1851.45 mmol) at -78 °C was added n-BuLi (22.4 mL, 56.25 mmol) over 10 min and stirred for 30 min at -78 °C under an argon atmosphere. Then the solution of Bra (3.6 mL, 70.31 mmol) in THF (10 mL) was added over 10 min to the solution at -78 °C. The resulting solution was slowly warmed 15 to room temperature and stirred for 2 h. The resulting mixture was diluted with NH4CI/H 2 O (100 mL) and

915

SUBSTITUTE SHEET (RULE 26) extracted with EtOAc (3 x 100 mL). The combined organic layers were dried over anhydrous NaaSO*. After filtration, the filtrate was concentrated under reduced pressure and purified by silica gel column chromatography to afford 5-bromo-2-[[(tert-butyldimethylsilyl)oxy]methyl]-1 ,3-oxazole (5.3 g, 38.6% yield) as yellow oil. LCMS (ESI): m/z [M+H] calc'd for CioHteBrNOaSi 292.04; found 292.0.

5 Step 3. To a solution of 5-bromo-2-[[(fert-butyldimethylsilyl)oxy]methyl]-1 ,3-oxazole (4.0 g, 13.69 mmol) in DCM (60.0 mL) was added PBrs (7.41 g, 27.37 mmol) at 0 °C under an argon atmosphere. The resulting solution was stirred for 4 h then diluted with NaHCOa/H 2 O (30mL). The mixture was extracted with EtOAc (3 x 40 mL). The organic layers were concentrated under reduced pressure and purified by silica gel column chromatography to afford 5-bromo-2-(bromomethyl)-1 ,3-oxazole (2.5 g, 75.7% yield) as

10 yellow oil. LCMS (ESI): m/z [M+H] calc’d for CAHaBraNO 239.87; found 241.9.

Step 4. A mixture of 5-bromo-2-(bromomethyl)-1 ,3-oxazole (9.0 g, 37.36 mmol), Cat:200132-54- 3 (2.26 g, 3.74 mmol), DCM (45.0 mL), toluene (90.0 mL), KOH (20.96 g, 373.63 mmol), H 2 O (42 mL),and fert-butyl 2-[(diphenylmethylidene)amino]acetate (13.24 g, 44.82 mmol) at 0 °C was stirred for 4 h then diluted with H 2 O (30 mL). The mixture was extracted with DCM (3 x 40 mL). The organic layers were

15 concentrated under reduced pressure and purified by reverse phase column chromatography to afford fert-butyl (2S)-3-(5-bromo-1 ,3-oxazol-2-yl)-2-[(diphenylmethylidene)amino]propanoate (4.8 g, 28.2% yield) as a yellow solid. LCMS (ESI): m/z [M+H] calc'd for CaaHaaBrNaOa 455.10; found 457.1.

Step 5. A mixture of fert-butyl (2S)-3-(5-bromo-1 ,3-oxazol-2-yl)-2- [(diphenylmethylidene)amino]propanoate (1.20 g, 2.64 mmol), DCM (10.0 mL, 157.30 mmol), and TEA

20 (5.0 mL, 67.32 mmol) at 0 °C was stirred for 2 h then concentrated under reduced pressure to afford (S)- 3-(5-bromooxazol-2-yl)-2-((2,2,2-trifluoroacetyl)-l4-azaneyl )propanoic acid (0.5 g, 81.3% yield) as a yellow solid. LCMS (ESI): m/z [M+H] calc'd for CeHyBrNaOa 234.97; found 237.0.

Step 6. A mixture of (S)-3-(5-bromooxazol-2-yl)-2-((2,2,2-trifluoroacetyl)-l4-aza neyl)propanoic acid (500.0 mg, 2.13 mmol), BocaO (928.56 mg, 4.26 mmol), dioxane (2.50 mL), H 2 O (2.50 mL), and

25 NaHCOa (714.84 mg, 8.51 mmol) at 0 °C was stirred for 3 h. The resulting solution was purified by reverse phase column chromatography to afford (2S)-3-(5-bromo-1 ,3-oxazol-2-yl)-2-[(fert- butoxycarbonyl)amino]propanoic acid (0.65 g, 91.1% yield) as a yellow solid. LCMS (ESI): m/z [M+H] calc’d for CnHisBrNaOs 335.02; found 334.8.

Step 7. To a solution of (2S)-3-(5-bromo-1 ,3-oxazol-2-yl)-2-[(fert-

30 butoxycarbonyl)amino]propanoic acid (500.0 mg, 1.49 mmol) and 3-[(2Af)-1-ethyl-2-[2-[(1 S)-1- methoxyethyl]pyridin-3-yl]-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)indol-3-yl]-2,2-dimethylpropan-1- ol (808.16 mg, 1.64 mmol) in DMF(5.0 mL) and H 2 O(1.0 mL) were added K3PO4 (791.67 mg, 3.73 mmol) and Pd(dppf)Cla(109.16 mg, 0.15 mmol). The resulting mixture was stirred for 2 h at 70 °C under an argon atmosphere. The mixture was purified by reverse phase column chromatography to afford (2S)-2-

35 [(fert-butoxycarbonyl)amino]-3-[5-[(2M)-1 -ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-[2-[(1 S)-1 - methoxyethyl]pyridin-3-yl]indol-5-yl]-1 ,3-oxazol-2-yl]propanoic acid (600 mg, 64.79% yield) as a light brown solid. LCMS (ESI): m/z [M+H] calc'd for C34H44N4O7621.33; found 621 .3.

Step 8. To a stirred mixture of methyl (3S)-1 ,2-diazinane-3-carboxylate(627.10 mg, 4.350 mmol) and (2S)-2-[(fert-butoxycarbonyl)amino]-3-[5-[(2Af)-1 -ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-[2-[(1 S)-1 -

40 methoxyethyl]pyridin-3-yl]indol-5-yl]-1 ,3-oxazol-2-yl]propanoic acid (900.0 mg, 1.45 mmol) in DCM (10.0 mL) were added HATU (661.54 mg, 1.74 mmol) and DIPEA (3747.71 mg, 29.00 mmol) at 0 °C. The resulting mixture was stirred for 2 h. Desired product could be detected by LCMS. The resulting mixture

916

SUBSTITUTE SHEET (RULE 26) was concentrated under reduced pressure and the crude material was purified by silica gel column chromatography to afford methyl (3S)-1 -[(2S)-2-[(te/t-butoxycarbonyl)amino]-3-[5-[(2/Vf)-1 -ethyl-3-(3- hydroxy-2,2-dimethylpropyl)-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]indol-5-yl]-1 ,3-oxazol-2-yl]propanoyl]- 1 ,2-diazinane-3-carboxylate (900mg,83.11%) as a brown yellow solid. LCMS (ESI): m/z [M+H] calc’d for 5 CwHwNeOe 747.41 ; found 747.2.

Step 9. To a stirred mixture of methyl (3S)-1 -[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[5-[(2M)-1 - ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-[2-[( 1 S)-1 -methoxyethyl]pyridin-3-yl]indol-5-yl]-1 ,3-oxazol-2- yl]propanoyl]-1 ,2-diazinane-3-carboxylate (2000.0 mg, 2.68 mmol) in THF (18. mL) and hfeO (6.0 mL) was added LIOH.HzO (337.10 mg, 8.03 mmol) at 0 °C. The resulting mixture was stirred for 2 h. Desired 10 product could be detected by LCMS. The reaction was quenched with H 2 O at 0 °C and adjusted to pH 6 with 1 N HCI solution. The resulting mixture was extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (1x10 mL), dried over anhydrous NaaSO*. After filtration, the filtrate was concentrated under reduced pressure to afford (3S)-1-[(2S)-2-[(terf-butoxycarbonyl)amino]-3-[5-[(2M)-1- ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-[2-[(1 S)-1 -methoxyethyl]pyridin-3-yl]indol-5-yl]-1 ,3-oxazol-2- 15 yl]propanoyl]-1 ,2-diazinane-3-carboxylic acid (1300 mg, 66.2% yield) as a yellow solid. LCMS (ESI): m/z [M+H] calc’d for CagHszNeOe 733.39; found 733.3.

Step 10. To a stirred mixture of (3S)-1-[(2S)-2-[(fert-butoxycarbonyl)amino]-3-[5-[(2M)-1-eth yl-3- (3-hydroxy-2,2-dimethylpropyl)-2-[2-[(1 S)-1 -methoxyethyl]pyridin-3-yl]indol-5-yl]-1 ,3-oxazol-2- yl]propanoyl]-1 ,2-diazinane-3-carboxylic acid (1.2 g, 1.64 mmol) and DIPEA (8.5 g, 65.50 mmol) in DCM 20 (120.0 mL) were added HOST (1.8 g, 13.10 mmol) and EDCI (7.8 g, 40.93 mmol) at 0 °C. The resulting mixture was stirred for 2 h. Desired product could be detected by LCMS. The mixture was concentrated under reduced pressure and purified by silica gel column chromatography to afford tert-butyl ((6 3 S,4S)-1 1 - ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8- oxa-2(5,2)-oxazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)carbamate (660 mg, 56.4%

25 yield) as a brown yellow solid. LCMS (ESI): m/z [M+H] calc’d for CsgHsoNeO? 715.38; found 715.3.

Step 11. A mixture of tert-butyl ((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-2(5,2)-oxazola-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-4-yl)carbamate (20.0 mg, 0.028 mmol) and TEA (3.0 mL) in DCM (6.0 mL) at 0 °C was stirred for 2 h. Desired product could be detected by LCMS. The resulting mixture was 30 concentrated under reduced pressure to afford (6 3 S,4S)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(5,2)-oxazola-1 (5,3)- indola-6(1 ,3)-pyridazinacycloundecaphane-5,7-dione (160mg, crude) as a yellow green solid. LCMS (ESI): m/z [M+H] calc’d for C34H42N6O5615.33; found 615.2.

Step 12. To a stirred mixture of (6 3 S,4S)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3- 35 yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 8 -hexahydro-1 1 /+8-oxa-2(5,2)-oxazola- 1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-5,7-dione (180.0 mg, 0.293 mmol) and (2S)-2-[(fert- butoxycarbonyl)(methyl)amino]-3-methylbutanoic acid (135.45 mg, 0.59 mmol) in DMF (2.0 mL) were added HATU (133.60 mg, 0.35 mmol) and DIPEA (756.86 mg, 5.86 mmol) at 0 °C. The resulting mixture was stirred for 2 h. Desired product could be detected by LCMS. The mixture was purified by reverse 40 phase column chromatography to afford fert-butyl ((2S)-1 -(((6 3 S,4S)-1 '-ethyl-1 2 -(2-((S)-1 - methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 ' H- 8-oxa-2(5,2)- oxazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2-

917

SUBSTITUTE SHEET (RULE 26) yl)(methyl)carbamate (160 mg, 66% yield) as a brown yellow solid. LCMS (ESI): m/z [M+H] calc’d for C45H61 Ντθβ 828.47; found 828.4.

Step 13. To a stirred mixture of fert-butyl ((2S)-1-(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-2(5,2)- 5 oxazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)amino)-3-methyl-1 -oxobutan-2- yl)(methyl)carbamate (160.0 mg, 0.19 mmol) in DCM (2.0 mL) was added TFA (1.0 mL) at 0 °C. The resulting mixture was stirred for 2 h. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure to afford (2S)-N-((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-2(5,2)- 10 oxazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)-3-methyl-2-(methylamino )butanamide (130 mg, crude) as a yellow solid. LCMS (ESI): m/z [M+H] calc'd for C40H53N7O6728.41 ; found 728.5.

Step 14. To a stirred mixture of ((2S)-N-((6 3 S,4S)-1 '-ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3- yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(5,2)-oxazola-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-4-yl)-3-methyl-2-(methylamino)but anamide (130.0 mg, 0.18 mmol) and 15 (3S)-1 -[4-(dimethylamino)-4-methylpent-2-ynoyl]pyrrolidine-3-carbo xylic acid (180.25 mg, 0.72 mmol) in

DMF (2.0 mL) were added DIPEA (461.64 mg, 3.57 mmol) and HATU (135.81 mg, 0.36 mmol) at 0 °C. The resulting mixture was stirred for 2 h. Desired product could be detected by LCMS. The mixture was purified by reverse phase column chromatography to afford (3S)-1-(4-(dimethylamino)-4-methylpent-2- ynoyl)-N-((2S)-1 -(((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1 -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo- 20 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(5,2)-oxazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-

4-yl)amino)-3-methyl-1-oxobutan-2-yl)-W-methylpyrrolidine -3-carboxamide (55.3 mg, 31.3% yield) as a solid. 1 H NMR (400 MHz, DMSO-de) 68.77 (dd, J = 4.8, 1.7 Hz, 1 H), 8.09 - 8.00 (m, 1 H), 7.92 (s, 1 H), 7.88 - 7.80 (m, 1 H), 7.62 (d, J = 8.7 Hz, 1 H), 7.59 - 7.49 (m, 2H), 7.39 - 7.30 (m, 1 H), 5.69 (p, J = 8.8 Hz, 1H), 5.44 (d, J= 12.1 Hz, 1H), 4.67 (d, J= 10.7 Hz, 1H), 4.30 - 4.15 (m, 3H), 3.99 (dt, J= 13.2, 6.4 25 Hz, 3H), 3.89 - 3.79 (m, 1 H), 3.62 (ddd, J = 30.5, 18.6, 11.4 Hz, 5H), 3.39 (dd, J = 9.4, 3.8 Hz, 2H), 3.21 -3.13 (m, 1H), 3.08 (d, J= 15.0 Hz, 3H), 2.99 - 2.74 (m, 6H), 2.26 - 2.18 (m, 5H), 2.16 (s, 2H), 2.14 - 1.94 (m, 3H), 1.86 - 1.68 (m, 2H), 1.57 (q, J = 9.2, 5.8 Hz, 1 H), 1.44 - 1.27 (m, 9H), 0.94 (d, J = 6.6 Hz, 4H), 0.89 (dd, J= 6.5, 2.5 Hz, 2H), 0.80 (d, J= 6.3 Hz, 2H), 0.77 - 0.69 (m, 4H), 0.58 (d, J= 20.4 Hz, 3H). LCMS (ESI): m/z [M+H] calc’d for CsaHyiNgOe 962.55; found 962.5.

30

918

SUBSTITUTE SHEET (RULE 26) Example A675. Synthesis of {2i¾-2 {((T(4 {dimethylam!no) 4”methyl pent-2-ynoyi)azetidin- 3” l)oxy)met yl)”N-({b 3 S,4S,Z5-1 1 -ethyl -1 ^ ”(2 {(S)-1-mbt ox eί yl)p r! !n-3-yl) 1b,10”dimet l·5,7- dioxo-6 1 ! 6 2 ,6 3 ,6 4 ! 8 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(2 ! 4)-oxa2:oia-1{5,3)dridoia-6(1 ! 3)- pyridaz!nacycioyndecap ane-4 yi)-3-methylbi!tanamide

Step 1. A mixture of 2-bromo-4-(ethoxycarbonyl)-1 ,3-oxazoi-5-ylium (6.83 g, 31 .19 mmol), EtOH (100.0 rrsL) and NaBH 4 (4.72 g, 124.76 mmol) at 0 °C was stirred for 6 h at 0 °C under an air atmosphere. The reaction was quenched with H 2 O at 0 °C The resulting mixture was extracted with EtOAc (3 x 100 ml). The combined organic layers were dried over anhydrous Na 2 SO 4, After filtration, the filtrate was concentrated under reduced pressure to afford (2-bromo-1 ,3-oxaz.ol-4-yl)methanol (4.122 g, 74.3% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 4 H 4 BrN0 2 177.95; found 178.0

Step 2. A mixture of (2-hromo-1 ,3-oxazol-4-yl)methanol (4.30 g, 24.16 mmol), DCM (50 L) and phosphorus tribromide (9809.39 mg, 36.24 mmol) at 0 °C was stirred overnight at 0 °C under an air atmosphere. The reaction was quenched by the addition of NaHCO 3 (aq.) at 0 °C. The resulting mixture was extracted with EtOAc (3 x 100 mL). The combined organic layers were dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure to afford 2-bromo-4- (bromomethyl)-1 ,3-oxazoie (3.28 g, 56.4% yield) as a liquid. LCMS (ESI): m/z [M+H] calc’d for C 4 H 3 Br 2 NO 239.87; found 239.9. Step 3, A mixture of 2-bromo-4-(bromomethyl)-1 ,3-oxazole(3280.0 mg, 13.62 mmol), KOH (9M, 10 rnL), 30 ml mixture of toiuene/DCM (7/3) and tert- butyl 2-[(diphenylmethylidene)amino]acetate (5228.74 mg, 17.70 mmol) at -16 °C was stirred overnight under an air atmosphere. The resulting mixture was extracted with EtOAc (3 x 50 rnL). The combined organic layers were dried over anhydrous Na^SCU. After filtration, the filtrate was concentrated under reduced pressure to afford fe/Tbutyl (28)--3-(2--bromo- 1 ,3-oxazol-4-yl)-2-[(diphenylmethylidene)amino]propanoate (8.33 g, 80.6% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for CjaH 2 jBrNaO 3 455.10; found 455.1 .

Step 4, A mixture of tert- butyl (2S)-3-(2-bromo-1 ,3-Oxazol-4-yl)-2-[(diphenylmethylidene)amino] propanoate (4100.0 mg, 9.0 mmol) and citric acid (1 N) (40.0 mL, 0.21 mmol), in THE (40 L) at room temperature was stirred overnight under an air atmosphere. The reaction was quenched by the addition of HCI (aq.) (100 mL) at 0 °C. The aqueous layer was extracted with EtOAc (3 x 100 L). K2CO 3 (aq.) (200 L) was added to the resulting mixture and extracted with EtOAc (3 x 100 mL). The organic layer was concentrated under reduced pressure to afford tert-buiyi (28)-2-amino-3-(2-bromo-1 ,3-oxazoi-4- yi)propanoate (1730 g, 66% yield) as a dark yellow solid. LCMS (ESI): m/z [M+H] cale’d for C 1 oHisBrNaO 3 291 .03; found 291 .0.

Step 5, A mixture of terf-butyl (2S)-2-amino-3-(2-bromo-1 ,3-oxazol-4-yl)propanoate (1780.0 mg, 6.11 mmol), TFA (10.0 mL) and DCM (10.0 l.) at 0 °C was stirred for overnight under an air atmosphere. The resuiting mixture was concentrated under reduced pressure to afford (2S)-2-amino-3-(2- bromo-1 ,3-oxazoi-4-yl)propanoic acid (1250 mg, 87% yield) as a dark yeilow solid. LCMS (ESI): m/z [M+H] calc’d for C 6 HyBrts Oa 234.97; found 234 9.

Step 6, A mixture of dl-tert-butyl dicarbonate(4178.58 g, 19.15 mmol), THF (10 mL), H 2 O (10 iTiL),(2S)-2-amino-3-(2-broiTio-1 ,3-oxazol-4-yl)propanoic acid (1500.0 mg, 6.38 rrsmol) and NaHCO 3 (3216.74 g, 38.29 mmol) at room temperature was stirred overnight under an air atmosphere. The reaction was quenched with H 2 O at room temperature. The resulting mixture was concentrated under reduced pressure. The resulting mixture was extracted with EtOAc (3 x 100 mL). The aqueous layer was acidified to pH 6 with 1 M HCi (aq.). The resulting mixture was extracted with EtOAc (3 x 100 mL). The combined organic layers were dried over anhydrous NA 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure to afford (2S)-3-(2-bromo-1 ,3-oxazoi-4-yl)-2-[(ierf- butoxycarbonyl)amino]propanoic acid (920 mg, 43.0% yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for C 1 IHI 5 BGN 2 0 5 335.02; found 335.0.

Step 7. A mixture of 3-(2-bromo-1 ,3-oxazoi-4-yl)-2-[(terf-butoxycarbonyl)amino]propanaie acid (850.0 mg, 2.54 mmol), methyl 1 ,2-dlazinane-3-carboxylate (1.88 g, 13.04 mmol), DIPEA (1966.68 g, 15.22 mmol), DCM (30.0 mL) and HATU (1446.48 mg, 3.80 mmol) at 0 °C was stirred for 3 h under an air atmosphere. The resulting mixture was extracted with DCM (3 x 50 L). The combined organic layers were dried over anhydrous NaaSCb. After filtration, the filtrate was concentrated under reduced pressure. The resulting mixture was purified by reverse flash chromatography to afford methyl 1-[3-(2-bromo-1 ,3- oxazol-4-yl)-2-[(ie/i-butoxycarbonyl)amino]propanoyl]-1 ,2--diazinane--3-carboxylate (610 mg, 52.1% yield) as a solid. LCMS (ESI): m/z [M+H] cale’d for C 1 7H 2 sBrN40t5461 .10; found 461 .0.

Step 8. A mixture of methyl 1-[3-(2-brorno-1 ,3-oxazol-4-yl)-2-[(terf- butoxycarbonyl)amino]propanoyi]-1 ,2-diazinane-3-carboxylate (570.0 mg, 1.24 mmol), LIOH (2.0 L, 1 M aq.) and THF (2.0 mL) at 0 c C was stirred for 3 h under an air atmosphere. The resulting mixture was concentrated under reduced pressure. The resulting mixture was extracted with EtOAc (3 x 50 mL). The combined aqueous layers were acidified to pH 5 with 1 N HCi (aq.). The aqueous phase was extracted with EtOAc (3 x 50 rnL). The combined organic layers were dried over anhydrous NaaSCXt. After filtration, the filtrate was concentrated under reduced pressure to afford 1-[3-(2-bromo-1 ,3-oxazol-4-yl)-2-i(ferf- butoxycarbonyl)amino]propanoyl]-1 ,2-diazinane-3-carboxylic acid (500 mg, 90.5% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 1 eTfeBrN^Oe 447.09; found 446.8.

Step 9, A mixture of 1-[3-(2-bromo-1 ,3-oxazoi-4-yl)-2-[(tert-butoxycarbonyl)amino]propanoyl]-

1 .2-diazinane-3-carboxylic acid (450.0 mg, 1 .01 mmol), 3-[(2/W)-1-ethyl-2-[2-[(1 S)-1-methoxyethyl]pyridin- 3-yl]-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaboroian-2-yl)indol-3-yl3-2,2-dimethylpropan-1-oi (743.19 mg, 1 .51 mmol), DMAP (24.58 mg, 0.20 mmol), DCM (15.0 mL) and DCC (311 .37 mg, 1 .51 mmol) at 0 °C was stirred for 3 h under an air atmosphere. The resulting mixture was extracted with EtOAc (3 x 20 mL). The combined organic layers were dried over anhydrous NazSCk. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC to afford 3-(1-ethyl-2-(2-((S)-1- methoxyethyl)pyridin-3-yl)-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaboroian-2-yl)-1H-indol-3-yl)-2,2- dimethylpropyl(S)-1-((S)-3-(2-bromooxazol-4-yl)-2-((tert- butoxycarbonyl)amino)propanoyi)hexahydropyridazine-3-carboxy late (330 mg, 35.8% yield) as a white solid. LCMS (ESI): m/z [M+H] calc d for C 4 5Hs2BBrN60« 921 39; found 921 .4.

Step 10, A mixture of 3-(1-ethyl-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-5-(4,4,5,5 -tetramethyl-

1 .3.2-dioxaboroian-2-yl)-1H-indol-3-yl)-2,2-dimethylpropyl(S) -1-((S)-3-(2-bromooxazoi-4-yl)-2-((feri- butoxycarbonyl)amino)propanoyi)hexahydropyridazine-3-carboxy late (290.0 mg, 0.33 mmol), K3PO4 (206.64 mg, 0.97 mmol), X-Phos (30.94 mg, 0.07 mmol), XPhos Pd G3 (54.93 g, 0.07 m ol), dioxane (5. mL) and H 2 O (1 .0 L) at 70 °C was stirred for 4 h under an argon atmosphere. The resulting mixture was extracted with EtOAc (3 x 20 mL). The combined organic layers were dried over anhydrous NA 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep- TLC to afford tert-butyl ((6 3 S,4S,Z)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5 ,7- dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 W-8-oxa-2(2,4)-oxazoia-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-4-yl)carbamate (130 mg, 56.0% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for CasHsoNeO 7 5.38; found 715.3.

Step 11. A mixture of tert-butyl ((6 3 S,4S,Z)-1 1 -ethyl-1 2 -(2-((S)-1-mefhoxyethyl)pyridin-3-yl)-10,1 Q- dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(2,4)-oxazoia-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-4-yl)carbamate (120.0 mg), DCM (2.0 mL) and TFA (0.2 mL) at room temperature was stirred for 6 h under an air atmosphere. The resulting mixture was concentrated under reduced pressure to afford (6 3 S,4S,Z)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10- dimethyl-6 1 ,6 2 ,8 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 W-8-oxa-2(2,4)-oxazola-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-5,7-dione (90 mg, 87.2% yield) as a solid. LCMS (ESI): m/z [M+H] calc ' d for C 3 4H42N6O5 615.33; found 615.3.

Step 12. A mixture of (6 3 S,4S,Z)-4-amino-1 1 -ethyl-1 2 -(2-((S)-l -me†hoxyethyl)pyridin-3-yl)-l 0,10- dimethyl-6 1 ,6 2 ,8 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 W-8-oxa-2(2,4)-oxazoia-1 (5,3)-indola-6(1 ,3) - pyridazinacycloundecaphane-5,7-dione (200.0 g, 0.33 mmol), (2R)-2-([[1-(diphenylmethyl)aze†idin-3- yl]oxy]methyl)-3-methylbutanoic acid (172.49 g, 0.49 mmol), DIPEA (420.48 mg, 3.25 mmol), DMF (3.0 mL) and HATU (148.44 g, 0.39 mmol) at 0 °C was stirred for 3 h under an air atmosphere. The resulting mixture was extracted with EtOAc (3 x 20 L). The combined organic layers were dried over anhydrous NaaSCX. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC to afford (2fl)-2-(((1-benzhydrylazetidin-3-yl)oxy)methyl)-N -((6 3 S,4S,Z)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 l 6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(2,4)- oxazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)-3-methylbutanamide (154 mg, 77.0% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 5 eHerNrO? 950.52; found 950.6.

Step 13, A mixture of (2f?)-2-(((1-benzhydrylazetidin-3-yl)oxy)methyl)-N- ((6 3 S,4S,Z)-1 1 -ethyl-1 2 - (2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-djmethyl-5,7-diox o-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 [ H~ 8-oxa- 2(2,4)-oxazola-1 {5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)-3-methylbutanamide (240.0 mg, 0.25 mmol), (Boo)kO (165.37 mg, 0.76 mmol), MeOH (5.0 mL) and Pd(OI ~ l) 2 (72.0 g, 0.51 mmol) at room temperature was stirred overnight under an Hj atmosphere. The resulting mixture was filtered, the filter cake was washed with MeOH (3 x 5 mL). The filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC to afford tert-butyl 3-((2f?)-2-(((6 3 S,4S,Z)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 W-8-oxa-2{2,4)- oxazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)carbamoyl)-3-methylbutox y)azetidine-1- carboxylate (150 mg, 67.2% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 4 8HS5N7O9 884.49; found 884.2.

Step 14, A mixture of tert-butyl 3-((2/?)-2-(((6 3 S,4S,2l)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin- 3-yl)-10,10-dimethyl-S -dioxo-B 1 ,6 2 ,6 3 ,6 4 ,6 5 .6 6 -hexahydro-1 1 W-8-oxa-2(2,4)-oxazola-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-4-yl)carbamoyi)-3-methylbutoxy)az etidine-1-carboxylate (150.0 mg), DCM (2.0 ml.) and TFA (0.40 mL) at 0 °C was stirred for 3 h under an air atmosphere. The resuiting mixture was concentrated under reduced pressure to afford (2ff)-2-((azotidin-3-yloxy)methyl)-fV-((6 3 S,4S,Z)-1 ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-S -dioxo-O 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hoxahydro-1 1 H- 8- oxa-2(2,4)-oxazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)-3-methylbutanamide (120 g, 90.2% yield) as a solid. LCMS (ESI): m/z [M+H] ealc’d for C 4 3H57N7O7784.44: found 784.2.

Step 15. A mixture of (2/?)-2-((azetidin-3-yloxy)methyl)-W-((6 3 S,4S,Z)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(2,4)- oxazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)-3-methylbutanamide (130.0 mg, 0.17 mmol), sodium 4-(dimethylamino)-4-methylpenf-2-ynoate (44.07 mg, 0.25 mmol), DMF (3.0 mL), DIPEA (64.29 mg, 0.50 mmol) and COMU (106.46 mg, 0.25 mmol) at 0 °C was stirred for 3 h under an air atmosphere. The resulting mixture was extracted with EtOAc (3 x 20 mL). The combined organic layers were dried over anhydrous NA 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by reverse phase chromatography to afford (2fl)-2-{((1-(4- (dimethylamino)-4-methylpent-2-ynoyi)azet!din-3-yl)oxy)methy l)-W-((6 3 S,4S,Z)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-2(2,4)- oxazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)-3-methylbutanamide (25 mg, 16.4% yield) as a solid. 1 H NMR (400 MHz, DMSO-dfe) d 8.77 (dd, J= 4.8, 1 .8 Hz, 1H), 8.55 (s, 1H), 8.14 (dd, J= 8.3, 2.9 Hz, 1H), 7.82 (d, J = 7.6 Hz, 1H), 7.76 - 7.65 (m, 3H), 7.54 (dd, J = 7.7, 4.7 Hz, 1H), 7.54 - 7.02 (m, 1H), 5.72 (td, J = 7.4, 3.4 Hz, 1H), 4.97 (d, J = 11 .9 Hz, 1H), 4.46 - 4.24 (m, 6H), 4.18 - 4.04 (m, 2H), 3.94 (dd, J = 32.9, 7.8 Hz, 1H), 3.77 - 3.63 (m, 2H), 3.57 (s, 1H), 3.49 (s, 2H), 3.21 (s, 3H), 2.90 (d, J = 14.6 Hz, 1H), 2.87 - 2.79 (m, 1H), 2.72 (td, J= 15.5, 14.6, 3.1 Hz, 2H), 2.46 (s, 1H), 2.43 - 2.26 (m, 6H), 2.11 - 1 99 (m, 1H), 1 82 - 1.66 (m, 2H), 1.56 - 1.37 (m, 11H), 0.89 (dt, J- 12,3, 7.7 Hz, 12H), 0.35 (s, 3H). LCMS (ESI): m/z [M+H] calc'd for C 5 iHeaNsOs 921 .52; found 921 .5. Example A607. The synthesis of C 2 f¾-2-C(C1-(4-( imethyl amino)-4”!inethylper!t-2- nhogl)0;zbMϊh-3-n1)o c n^qίHgl)-M-({2¾,6¾435-1 1 -b^M 2 -(2-((5)-1^6¾Ho c nqIHnl)rnp !h-3^l)- 1Q,10-dimet yl·5,7 !O o-8^8 2 ,6 3 ,6^6 5 ,8 6 - exa y !Ό-1 1 H-8-oxa-1(5,3) !ndoia-6{1,3)-p r! a2:lna 2{1,3)-p!perid!nacycioundecaphane-4-yl)-3"meihyl butariamide

Step 1. A mixture of Zn (44.18 g, 675.41 mmol) and la (8.58 g, 33.77 mmol) in DMF (120 mL) was stirred for 30 min at 50 °C under an argon atmosphere, followed by the addition of methyl (2R}-2- [(tert-bufoxycarbonyl) amino]-3-iodopropanoate (72 24 g, 219.51 mmol) in DMF (200 mL). The reaction mixture was stirred at 50 °C for 2 h under an argon atmosphere. Then a mixture of 2,6-dibromo-pyridine (40 g, 168.85 mmol) and PdfPPhs (39.02 g, 33.77 mmol) in DMF (200 mL) was added. The resulting mixture was stirred at 75 °C for 2 h, then cooled down to room temperature and extracted with EtOAc (1 L x 3). The combined organic layers were washed with FLO (1 L x 3), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford methyl (2S)-3-(6-bromopyridin-2-yl)-2-[(tert-butoxycarbonyl) amino] propanoate (41 g, 67% yield) as oil. LCMS (ESI): m/z [M+H] calc’d for CuH^BrNaC 358.1 ; found 359.1 .

Step 2. To a solution of 3-[(2M)-2-[2-[(1 S)-1-methoxyethyl] pyridin-3-yl]-5-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)-1-(2,2,2-trif luoroethyl) indol-3-yl]-2,2-dimethylpropan-1-ol (45.0 g, 82.35 mmol) in dioxane (400 mL) and H 2 O (80 mL), were added potassium carbonate (28.45 g, 205.88 mmol), methyl (2S)-3-(6-brGmopyridin-2-yl)-2-[(te/t-bu†oxycarbanyl) amino] propanoate (35.5 g, 98.8 mmol), Pd(dtbpf)Cl2 (5.37 g, 8.24 mmol) at room temperature. The reaction mixture was stirred at 70 °C for 2 h under a nitrogen atmosphere. The resulting mixture was extracted with EtOAc (500 L x 3). The combined organic layers were washed with H 2 O (300 mL x 3), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography to afford methyl (S)-2-((fefi-butoxycarbonyl)amino)-3-(6-(3-(3-hydroxy-2,2-di methylpropyl)-2-(2-{(S)-1- methoxyethyl)pyridin-3-yl)-1-{2 2,2-trifiuoroethyl)-1H-indol-5-yl)pyridin-2-yl)propanoate (48 g, 83% yield) as solid. LCMS (ESI): m/z [M+H] calc ' d for C 3 7H45F3N4O6898.3; found 699.4.

Step 3, A solution of methyl (S)-2-((ie/†-butoxycarbonyl)amino)-3-(6-(3-(3-hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1-(2, 2,2-trifluoroethyl)-1H-indcl-5-yl)pyridin-2- yl)propanoate (52 g, 74.42 mmol) in THF (520 mL), was added LiOH (74.41 mL, 223.23 mmol) at 0 °C. The reaction mixture was stirred at room temperature for 3 h. The resulting mixture was acidified to pH 5 with HCl (aq.) and extracted with EtOAc (1 L x 3). The combined organic layers were washed with HjO (1 L x 3), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford (S)-2-((tert-butoxycarbonyl)amino)-3-(6-(3-(3-hydroxy-2 > 2-dimethylpropyl)-2-(2-((S)-1- methoxyethyl)pyridin-3-yl)-1-(2,2,2-trifiiioroethyl)-1H-indo l-5-yl)pyridin-2-yl)propanoic acid (50 g, 98% yield) as solid. LCMS (ESI): m/z [M+H] calc’d for C 3 6H43F3N4OS 884.3; found 685.1

Step 4. To a solution of (S)-2-((tert-butoxycarbonyl)amino)-3-(6-(3-(3-hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1-(2, 2,2-trifiuoroethyl)-1H-indol-5-yl)pyridin-2- yi)propanoic acid (55 g, 80.32 mmol) in DCM (600 L), were added DIPEA (415.23 g, 3212.82 mmol), and HATH (45.81 g, 120.48 mol) at 0 °C. The reaction mixture was stirred at room temperature for 12 h and then quenched with H 2 O. The resulting mixture was extracted with EtOAc (1 L x 3). The combined organic layers were washed with H 2 O (1 L), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gei column chromatography to afford methyl 1-((S)-2-((tert-butoxycarbonyl)amino)-3-(6-(3-(3-hydroxy-2,2 -dimethylpropyl)-2-(2-((S)-1- methoxyethyl)pyridin-3-yl)-1-(2,2,2-trifluoroethyl)-1H-indol -5-yl)pyridin-2- yl)propanoyl)hexahydropyridazine-3-carboxylate (63 g, 96% yield) as solid. LCMS (ESI): m/z [M+H] calc'd for C 4 2H53F3N6O7810.4; found 811 .3.

Step 5. A solution of methyl 1-((S)-2-((terTbutoxyearbonyl)amino)-3-(6-(3-(3-hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1-(2, 2,2-trifluoroethyl)-1H-indol-5-yl)pyridin-2- yl)propanoyl)hexahydropyridazine-3-carboxylate (50 g, 61.66 rnmol) in THF (500 mL) and 3M LiOH (61 .66 mL, 184.980 mmol) at 0 °C was stirred at room temperature for 3 h, then acidified to pH 5 with HGI (aq.). The resulting mixture was extracted with EtOAc (800 mL x 3). The combined organic layers were washed with H 2 O (800 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford 1-((S)-2-((tert-butoxycarbonyl)amino)-3-(6-(3-(3-hydroxy-2,2 -dimethylpropyl)- 2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1-(2,2,2-trifiuoroeth yl)-l H-indol-5-yl)pyridin-2- yl)propanoyi)hexahydropyridazine-3-carboxylic acid (48 g, 97% yield) as solid. LCMS (ESI): m/z [M+H] calc ' d for C^HsiFaNeO? 796.3; found 797.1 .

Step 8. To a solution of l-((S)-2-((ie/†-butoxycarbonyl)amino)-3-(6-(3-(3-hydroxy-2 ,2- dimeihylpropyl)-2-(2-((S)-1-meihoxyethyl)pyridin-3-yl)-1-(2, 2,2-trifluoroeihyl)-1H-indol-5-yl)pyridin-2- yl)propanoyl)hexahydropyridazine-3-carboxylic acid (50 g, 62.74 mmol) In DCM (10 L) at 0 °C, were added DiPEA (243.28 g, 1882.32 mmol), EDCi (360.84 g, 1882.32 mmol) and HOST (84.78 g, 627.44 mmol). The reaction mixture was stirred at room temperature for 3 h, quenched with H 2 O and concentrated under reduced pressure. The residue was extracted with EtOAc (2 L x 3) The combined organic layers were washed with H 2 O (2 L x 3), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford tert-butyl ((4S)-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5 ,7-dioxo-1 1 -(2,2,2- trifluoroethyl)-6 1 ,8 2 ,6 3 ,6 4 ,6 5 ,8 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(2,6)- pyridinacycloundecaphane-4-yl)carbamate (43.6 g, 89% yield) as solid. LCMS (ESI): m/z [M+H] calc’d for C 4 1H49F3N6O6 778.3; found 779.3.

Step 7. To a solution of tert- butyl ((4S)-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl- 5,7-dioxo-1 L (2,2,2-trlfluoroethyl)--6 1 ,6 2 ,6 3 ,6 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-l (5,3)-indola-6(1 ,3)-pyridazlna- 2(2,6)-pyridinacycloundecaphane-4-yl)carbamate (300 g) in DCM (10 mL), was added TEA (3 mL) at 0 °C. The reaction mixture was stirred at room temperature for 1 h. The resulting mixture was diluted with toluene (10 L) and concentrated under reduced pressure three times to afford (4S)-4-amlno-1 2 -(2-((S)-

I -methoxyethyl)pyridin-3-yl)-10,10-dimethyl-1 1 -(2,2,2-trifluoroethyl)-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8- oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(2,8)-pyridinacycloundecaphane-5,7-dione (280 g, crude) as oil. LCMS (ESI): /z [M+H] calc’d for GaeE iFsNeC 679.2; found 678.3.

Step 8. To a solution of (4S)-4-amino-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-1 ^ (2 ,2 ,2-trif I uoroethyl)-6 1 ,6 2 ,6 3 1 6 4 ,6 5 ,6 6 -hexahydro-1 1 W-8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(2,6)- pyridinacycloundecaphane-5,7-dione (140 mg, 0.21 mmol) in MeCN (2 mL), were added DIPEA (268.58 g, 2.06 m ol), W-(4-(tert-butoxycarbonyl)-1-oxa-4,9-diazaspiro[5 5]undecane-9-carbonyl)-W-methyl-L- valine (127.94 g, 0.31 mmol) and CIP (114.68 mg, 0.41 mmol) at 0 °C. The reaction mixture was stirred at room temperature for 1 h. The resulting mixture was concentrated under reduced pressure. The residue was extracted with EtOAc (10 mL. x 3). The combined organic layers were washed with H 2 O (10 mL x 3), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by siiica gei chromatography to afford tert-butyl 9-(((2S)-1-(((8 3 S,4S)-1 2 -(2-((S)-1- methoxyethyl) pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-1 1 -(2,2,2-trifiuoroethyl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-

I I H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(2,6)-pyridinacycloundecaphane-4-yl) amino)-3-methyl-1- oxobutan-2-yl) (methyl)carbamoyi)-1-oxa-4,9-diazaspiro [5.5] undecane-4-carboxylate (170 mg, 76% yield) as solid. LCMS (ESI): m/z [M+H] calc’d for C S 6H74F3N9O9 1073.5; found 1074.6.

Step 9. To a solution of tert- butyl 9-(((2S)-1-(((6 3 S,4S)-1 2 -(2-((S)-1-methoxyethyl) pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-1 1 -(2,2,2-trif luoroethyl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 W-8-oxa-1 (5,3)-indola- 6(1 ,3)-pyridazina-2(2,6)-pyridinacycloundecaphane-4-yl) amino)-3-methyl-1-oxobutan-2-yl) (methyl)carbamoyl)-1-oxa-4,9-diazaspiro [5.5] undecane-4-carboxylate (160 mg, 0.15 mmol) in DCM (5 mL) at 0 °C, was dropwise added TEA (1 .5 mL). The reaction mixture was stirred at 0 °C for 1 h. The resulting mixture was dlluted with toluene (10 L) and concentrated under reduced pressure three times to afford N-({2S)-1 -(((6 3 S,4S)-1 2 -(2-((S)-1-methoxyethyl) pyridin -3-yl)-10,10-dimethyl-5,7-dioxo-1 1 -(2,2,2- trifluoroeihyl)-6 1 ,6 2 ,6 3 ,6 4 ,8 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-lndola-6(1 ,3)-pyridazina-2(2,6)- pyridinacycloundecaphane-4-yl) amino)-3-methyl-1-oxobutan-2-yl)- ( V-methyl-1-oxa-4,9-diazaspiro [5.5] undecane-9-carboxamide (150 mg, crude) as oil. LCMS (ESI): m z [M+H] calc’d for CS1H65F3N O7973.5; found 974.4.

Step 10. To a solution of N- ((2S)-1-(((6 3 S,4S)-l 2 -(2-((S)-1-methoxyethyl) pyridin-3-yl)-10,10- dlmethyl-5,7-dioxo-1 1 -(2,2,2-trifluoroethyl)-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(2,6)-pyridinacycloundecaphane-4-yl) amino)-3-methyl-1-oxobutan-2-yl)-N-methyl-1-oxa-4,9- diazaspiro [5.5] undecane-9-carboxamide (150 g, 0.15 m ol) in DMF (3 mL), were added DlPEA (199.01 mg, 1 .54 mmol), acrylic acid (16.64 mg, 0.23 mmol) and COMU (98.39 mg, 0.23 mmol) at 0 °C. The reaction mixture was stirred at room temperature for 1 h and concentrated under reduced pressure. The residue was extracted with EtOAc (10 mL x 3). The combined organic layers were washed with h½0 (10 mL x 3), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography and reverse phase chromatography to afford 4-acryloyl- N-({2S)-1 -(((6 3 S,4S)-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,1 Q-dimethyl-5,7-dioxo-1 1 -(2,2,2- trifiuoroethyl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro--1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(2,6)- pyridinacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-y l)-N -methyl-1-oxa-4,9- diazaspiro[5.5]undecane-9-carboxamide (53mg, 33% yield) as solid. 1 H NMR (400 MHz, DMSO-de) d 8.79 (dd, J = 4.8, 1 .9 Hz, 2H), 8.09 (d, J = 31 .4 Hz, 1H), 7.96 (d, J = 8.9 Hz, 1H), 7.90 - 7.72 (m, 3H), 7.64 (t, J = 7.8 Hz, 1H), 7.56 (dd, J = 7.8, 4.7 Hz, 1H), 7.03 (s, 1H), 6.86 (dd, J = 16.6, 10.4 Hz, 1H), 8.20

(d, J = 14.0 Hz, 1H), 5.73 (d, J = 12.2 Hz, 2H), 5.47 (s, 1H), 5.35 (d, J = 11 .8 Hz, 1H), 4.68 (s, 1H), 4.28

(d, J - 12.5 Hz, 1H), 4.13 (d, J = 6.4 Hz, 1H), 3.92 (s, 1H), 3.84 - 3.64 (m, 6H), 3.59 (d, J - 14.0 Hz, 3H),

3.50 (s, 3H), 3.10 (s, 5H), 3.02 (d, J = 13.3 Hz, 2H), 2.85 (d, J = 12.1 Hz, 3H), 2.08 - 1.89 (m, 2H), 1.81 (s, 1H), 1 .75 - 1 51 (m, 5H), 1 39 (d, J - 6.1 Hz, 4H), 1 .24 (s, OH), 0.91 - 0.66 (m, 10H), 0.53 (s, 3H). LCMS (ESI): m/z [M+H] calc’d for G54HS8F3N9O8 1027.5; found 1028.1.

Example A590. The synthesis of (2i¾-2 (((1-(4-(dirriethylamlRo) 4”methylpent-2 ynoyl)azetid -3-yl)Gxy)meihyl)-W”(C6 3 S,4S,2)-1 s -(2-({S)-1-methoxyeihyl)p r! -3-yl)-1Q,10” dimethyl-5 J dioxO”1 1 -(2, 2 ,2-friflyoroethyl)-6 1 ,6 ¾ ,6 3 ,8 4 , 6 s , 6 6 -hexahydro-1 1 if-8-oxa-2(5,3Hhiadiazola- 1(5,3)-ir«dGia-6(1,3)”pyrldazirsacycloi decapharie-4”yl)-3-methyl bytariam!de

Step 1, To a mixture of ethyl 2-ethoxy-2-iminoacetate (25.0 g, 172.23 mmol) and EtOH (250.0 ml) at 0 °C was added ammonium chloride (9.21 g, 172 23 mmol) in portions then stirred for 4 h at room temperature under an argon atmosphere. The resulting mixture was concentrated under reduced pressure and washed with EtaO (3 x 200 ml.). The organic layers were combined and concentrated under reduced pressure. This resulted in ethyl 2-amino-2-iminoacetate hydrochloride (20g, crude) as a light yellow solid. LCMS (ESI): m/z [M+H] calc’d for C 4 H8N 2 O 2 117.07; found 116.9.

Step 2. To a mixture of ethyl 2-amino-2-iminoacetate hydrochloride (13.30 g, 87.17 mmol), H 2 O (50.0 mL) and Et2O (100.0 mL) at 0 °C was added sodium hypochlorite pentahydrate (7.79 g, 104.60 mmol) dropwise. The resulting mixture was stirred tor 3 h under an argon atmosphere. The mixture was extracted with EtaO ( 3x 200 rnL). The resulting solution was washed with brine (3 x 100 mL). The organic phase was dried over anhydrous NA 2 SO 4 and concentrated under reduced pressure to ai!ord ethyl (2)-2- amino-2-(chloroimino) acetate (7 g, crude) as a light yellow solid. LCMS (ESI): m/z [M+H] calc’d tor C 4 H7CIN 2 O 2 151 .03; found 150.8.

Step 3, To a solution of ethyl (Z)-2-amino-2-(chloroimino) acetate (8.40 g, 55.792 m ol) and MeOH (130.0 L) at 0 °C was added potassium thiocyanate (5.42 g. 55.79 mmol) in portions. The resulting mixture was stirred for 4 h at room temperature under an argon atmosphere. The reaction was quenched with HjO/ice. The mixture was extracted with EtOAc (5 x 100 mL). The resulting organic phase was dried over anhydrous NazSCk and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford ethyl 5-amino-1 , 2, 4-thiadiazoie-3-carboxylate (2.3 g, 23.8% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for G5H7N3O 2 S 174.03; found 173.8.

Step 4, To a solution of ethyl 5-amino-1 , 2, 4-thiadiazole-3-earboxylate (5.80 g, 33.49 mmol), MeCN (90.0 mL) and CuBrz (11 .22 g, 50.23 mmol) at 0 °C was added 2-methyl-2-propylnitrit (6.91 g, 86.98 mmol) dropwise under an argon atmosphere. The mixture was stirred for 30 min. The mixture was then stirred for 4 h at 50 c C. The mixture was cooled to 0 °C and quenched with H 2 O/lee. The mixture was extracted with EtOAc (3x100 mL). The resulting organic phase was dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure. The residue was purified by silica gel chromatography to afford ethyl 5-bromo-1 , 2, 4-thiadiazoie-3-carboxylate (6.2 g, 78.1% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for GsHsBrISbCteS 236.93; found 237.1 .

Step 5. To a solution of (S)-3-(5-bromo-2-(2-(1-methoxyethyl)pyridin-3-yl)-1-(2,2,2- trifiuoroethyl)-1H-indol-3-yl)-2,2-dimethylpropan-1-ol (18.60 g, 33.24 mmol), DCM (170.0 mL) and imidazole (5.66 g, 83.10 mol) at 0 °C was added ierf-butyl-chlorodiphenylsilane (11 .88 g, 43.21 mol) dropwise. The resulting mixture was stirred for 3 h at room temperature under an argon atmosphere. The reaction was quenched with H 2 O/lce. The mixture was extracted with EtOAc (3x 200 mL). The organic layer was washed with brine (3x100 mL), dried over anhydrous NA 2 SO 4 and concentrated under reduced pressure. The residue was purified fay silica gel column chromatography to afford (S)-5-bromc-3-(3-((iert- butyldiphenylsllyl)oxy)-2,2-dimethylpropyl)-2-(2-(1-methoxye thyl)pyridin-3-yl)-1-(2,2,2-trifluoroethyl)-1 H- indole (22 g, 89.7% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for CasHwBrFs^OiSi 737.24; found 737.0.

Step 6. To a solution of (S)-5-bromo-3-(3-((iefi-butyldiphenylsllyl)oxy)-2,2-dimethyl propyl)-2-(2- (1-methoxyethyl)pyridin-3 -yl)-1-{2,2,2 trifiuoroethyl) -1 // indole (28.0 g, 37.95 mmol), toluene (270.0 L), KOAc (9.31 g, 94.88 mmol) and bis(pinacolato)diboron (19.27 g, 75.90 mmol) at 0 °C was added Pd(dppf)Cl2.CH?Cl2 (6.18 g, 7.59 mmol) in portions. The resulting mixture was stirred for 3 h at 90 “C under an argon atmosphere. The mixture was cooled to room temperature and quenched with LEG/lce. The mixture was extracted with EtOAc (3 x 200 mL). The resulting organic phase was dried over anhydrous NA 2 SO 4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford (S)-3-(3-((fe/i-butyldiphenylsllyl)oxy)-2,2-dimethylpropyl)- 2-(2-(1- methoxyethyl)pyridin-3-yl)-5-(4,4,5,5-tetramethyl-l ,3,2-dioxaborolan-2-yl)-1-(2,2,2-trifluoroethyl)-1H-indole (28.2 g, 94.7% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for C 4 5H56BF3N 2 O4S1 785.41 ; found 785.4.

Step 7. To a solution of (S)-3-(3-((ie/T-butyldiphenylsilyl)oxy)-2,2-dimethylpropyl)- 2-(2-(1- methoxyethyl)pyridin-3-yl)-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-1-(2,2,2-trifluoroethyl)-1 //-indole ( 9.60 g, 24.97 mmol), ,4-dioxane (200 ml), H 2 O (40 ml), ethyl 5-bromo-1 , 2, 4-thiadiazoie-3- carboxylate (5.92 g, 24.97 mmol) and K3PO4 (13.25 g, 62.43 mmol) at 0 °C was added Pd(dtbpf)Cl2 (1 .63 g, 2.50 mmol) in portions. The resulting mixture was stirred for 1 .5 h at 75 °C under an argon atmosphere. The mixture was cooled to 0 °C and quenched with h O/Ice and extracted with EtOAc (3x200 rnL). The resulting organic phase was dried over anhydrous Na2SO 4 and concentrated under reduced pressure.

The residue was purified by silica gel column chromatography to afford ethyl (S)-5-(3-(3-((tert·· biityldiphenylsllyl)oxy)-2,2-dlmeihylpropyl)-2-(2-(1-methoxy ethyl)pyridin-3-yl)-1-(2,2,2-trifluoroethyl)-1 H- indol-5-yl)-1 ,2,4-thiadiazoie-3-carboxylate (14 g, 68.8% yield) as a yellow solid. LCMS (ESI): /z [M+H] calc’d for C 4 4H49F3N4O4SS1 815.33; found 815.2.

Step 8, To a solution of ethyl (S)-5-(3-(3-((tert-butyldiphenylsllyi)oxy)-2,2-dimethylpropy l)-2-(2-(1- methoxyethyl)pyridin-3-yl)-1-(2,2,2-trifluoroethyl)-1 W-indol-5-yl)-1 ,2,4-thiadiazole-3-carboxylate (13.60 g, 16.69 mmol) and EtOH (140.0 mL) at 0 °C was added NaBhU (3 16 g, 83.43 mmol) in portions. The resulting mixture was stirred for 3 h then quenched with hbO/lce. The resulting mixture was washed with brine (3 x 100 mL) and extracted with EtOAc (3 x 200 mL) The combined organic layers were dried over anhydrous NasSOi and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford (S)-5-(3-(3-((ieri-butyldiphenylsllyl)oxy)-2,2-dimethylpropy l)-2-(2-(1- methoxyethyl)pyridin-3-yl)-1-(2.2,2-trifiuoroethyl)-1H-indol -5-yl)-1 ,2,4-thiadiazol-3-ol (9.7 g, 75.2% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 4 2H47F3N4O 3 SSi 773.32; found 773.3.

Step 9, To a solution of (S)-5-(3-(3-((ieri-butyldiphenylsllyi)oxy)-2,2-dimethylpropy l)-2-(2-(1- methoxyethyl)pyridin-3-yl)-1-(2.2,2-trifiuoroethyl)-1H-indol -5-yl)-1 ,2,4-thiadiazol-3-ol (9.70 g, 12.55 mmol), DCM (100.0 mL) and CB^ (8.32 g, 25.10 mmol) at 0 °C was added PPli3 (6.58 g, 25.10 mmol) in DCM (20.0 mL) dropwise. The resulting mixture was stirred for 2 h under an argon atmosphere then quenched with h O/ice. The mixture was extracted with DCM (3 x 200 ml.). The resulting organic phase was dried over anhydrous NaaSCTi and concentrated under reduced pressure. The residue was purified by reverse flash chromatography to afford (S)-3-(bromomethyl)-5-(3-(3-((/e/7-butyldiphenylsilyl)oxy)-2 ,2- dimethylpropyl)-2-(2-(1-mefhoxyethyl)pyridin-3-yl)-1-(2,2,2- trifluoroethyl)-1H-indol-5-yl)-1 ,2,4-thiadiazoie (9.5 g, 90.6% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 42 H eBrF 3 N 4 0 2 SSi 835.23; found 834.9.

Step 10. To a stirred solution of (S)-3-(bromomethyl)-5-(3-(3-((feff-butyldiphenylsllyl)oxy)-2 ,2- dimethylpropyl)-2-(2-(1-methoxyethyl)pyridin-3-yl)-1-(2,2,2- triiiuoroethyl)-1H-indol-5-yl)-1 ,2,4-thiadiazoie (9.40 g, 11 .25 mmol), toluene (84.0 mL), DCM (36.0 mL), tert-butyl 2- [(diphenylmethylidene)amino]acetate (3.32 g, 11 .25 mmol) and O-Allyi-N- (3- anthracenylmethyl)cinchonidinium bromide (0.68 g, 1.13 mmol) at 0 °C was added 9M KOH aqueous (94.0 mL) dropwise. The resulting mixture was stirred overnight under an argon atmosphere. The mixture was extracted with EtOAc (3 x 200 mL) and the organic phase was concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford tert-butyl (S)-3-(5-(3-(3-((te/T butyldiphenylsllyl)oxy)-2,2-dimethylpropyl)-2-(2-((S)-T-meth oxyeihyl)pyridin-3-yl)-T-(2,2,2-trifluoroethyl)·· 1H-indol-5-yl)-1 ,2,4-thiadiazol-3-yl)-2-((diphenylmethylene)amino)propanoate (9 g, 78.2% yield)as a solid. LCMS (ESI): m/z [M+H] calc’d for C 6 iHeeFsNsO+SSi 1050.46; found 1050.8.

Step 11. To a solution of te/t-butyl (S)-3-(5-(3-(3-((tert-butyldiphenylsllyl)oxy)-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1-(2, 2,2-trifiuoroethyl)-1H-indol-5-yl)-1 ,2.4- thiadiazoi-3-yl)-2-((diphenylmethylene)amino)propanoate (8.0 g, 7 62 mmol) and DCM (40.0 mL) at 0 °C solution was added TFA (40.0 rnL) dropwise. The resulting mixture was stirred overnight at room temperature then concentrated under reduced pressure. The residue was basified to pH 8 with NaHCO 3 . The mixture was extracted with EtOAc (3 x 200 ml_). The organic phase was concentrated under reduced pressure. The residue was purified by reverse flash chromatography to afford (S)-2-amino-3-{5-(3-(3- ((fe/†-butyldiphenylsilyl)oxy)-2,2-dimethylpropyl)-2-(2-(( S)-1-methoxyethyl)pyridin-3-yl)-1-(2,2,2- trifluoroethyl)-1H-indol-5-yl)-1 ,2,4-thiadiazol-3-yl)propanoic acid (5 g, 79.1% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C^HsoFsNsG^SSi 830.34; found 830.2.

Step 12, To a solution of (S)-2-amino-3-(5-(3-(3-((te/t-butyldiphenylsllyl)oxy)-2,2-di methylpropyl)·· 2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1-(2,2,2-trlfluoroeth yl)-1H-indol-5-yl)-1 ,2,4-thiadiazol-3-yl)propanoic acid (4.70 g, 5.66 mmol), DCM (50.0 mL) and EtaN (2.86 g, 28.31 mmol) at 0 “C was added (Boc)aO (1 .36 g, 6.23 mmol) dropwise. The resulting mixture was stirred for 3 h at room temperature under an argon atmosphere then concentrated under reduced pressure and purified by reverse flash chromatography to afford (¾-2-((ierf-butoxycarbonyl)amino)-3-(5-(3-(3-((iert-butyldi phenylsllyi)oxy)-2,2-dimethylpropyl)-2-(2- ((S)-1-methoxyethyl)pyridin-3-yl)-1-(2,2,2-trifluoroethyl)-1 H-indol-5-yl)-1 ,2,4-thiadiazoi-3-yl)propanoic acid (5 g, 94.9% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 4 3H58F3N5O6SSI 930.39; found 930 3.

Step 13, To a mixture of (S)-2-((tert-butoxycarbonyl)amino)-3-(5-(3-(3-((iert- buiyidiphenylsllyi)oxy)-2,2-dimethylpropyl)-2-(2-((S)-1-meth oxyethyl)pyridin-3-yl)-1-(2,2,2-trifiuoroethyl)- 1H-indol-5-yl)-1 ,2,4-thiadiazol-3-yl)propanoic acid (5.30 g, 5.70 mmol), DMF (60.0 L), methyl 1 ,2- diazinane-3-carboxylate (1 .64 g, 11 .40 mmol) and DiPEA (22.09 g, 170.94 mmol) at 0 °C was added HATU (2 82 g, 7.41 mmol) in DMF (5 ml.) dropwise. The resulting mixture was stirred for 3 h at room temperature under an argon atmosphere. The reaction was then quenched with H 2 O/ice. The mixture was extracted with EtOAc (3 x 100 mL) and the organic phase was washed with brine (3 x 100 mL). The resulting mixture was dried over anhydrous teSC and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford methyl (5)-1-((S)-2-((/erf- butoxycarbonyl)amino)-3-(5-(3-(3-((/ert-butyldiphenylsilyl)o xy)-2,2-dimethylpropyl)-2-(2-((S)-1- methoxyethyl)pyridin-3-yl)-1-(2,2,2-trifluoroethyl)-1H-indol -5-yl)-1 ,2,4-thiadiazol-3- yl)propanoyi)hexahydropyridazine-3-carboxylate (5.6 g) as a solid. LCMS (ESI): rn/z [M+H] calc’d for C55H68F3N7G7SS! 1056.47; found 1056.2.

Step 14. A mixture of methyl (S)-1-((S)-2-((/er/-butoxycarbonyl)aminc)-3-(5-(3-(3-((tert- butyldiphenylsllyl)oxy)-2,2-dimethylpropyl)-2-(2-((S)-1-meth oxyethyl)pyridin-3-yl)-1-{2,2,2-trifluoroethyl)- 1H-indol-5-yl)-1 ,2,4-thiadiazol-3-yl)propanoyi)hexahydropyridazine-3-carboxy late (5.60 g, 5.30 mmol) and TBAF In THF (56.0 mL) was stirred overnight at 40 °C under an argon atmosphere. The reaction was quenched with sat. NH4CI (aq.). The mixture was extracted with EtOAc (3 x 100 L) and the organic phase was concentrated under reduced pressure. The residue was purified by reverse flash chromatography to afford (S)-1-((S)-2-((fe/i-butoxycarbonyl)amino)-3-(5-(3-(3-hydroxy -2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1-(2, 2,2-trifluoroethyl)-1H-indol-5-yl)-1 ,2,4- thiadiazol-3-yl)propanoyl)hexahydropyridazine-3-carboxylie acid (4.1 g, 96.2% yield) as a solid. LCMS (ESI): m/z [M+H] calc ' d for C 3 8H48F3N7O7S 804.34; found 804.3.

Step 15. To a solution of (S)-1-((S)-2-((iert-butoxycarbonyl)amino)-3-(5-(3-(3-hydroxy -2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1-(2, 2,2-trifiuoroethyl)-1H-indol-5-yl)-1 ,2,4- thiadiazoi-3-yl)propanoyl)hexahydropyridazine-3-carboxylic acid (4.0 g, 5.0 mmol) and DCM (450.0 mL) at 0 c 'C were added DIPEA (51 .45 g, 398.08 mmol), HOBt (6 72 g, 49.76 mmol) and EDCI (57.23 g, 298.55 mmol) in portions. The resulting mixture was stirred for 16 h at room temperature under an argon atmosphere. The reaction was quenched with hhO/lce and extracted with EtOAc (3 x 30 rriL). The organic phase was concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford ierf-butyl ((6 3 S,4S,2)-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,1 Q-dimethyl-5,7- dioxo-1 1 -(2,2,2 trifluoroethyl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 l H-8-oxa-2(5,3)-ihiadiazola-1 (5,3)-indola- 6(1 3)-pyridazinacycloundecaphane-4-yl)carbamate (1 .7 g, 43.5% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for Cssf-beFsIM/OsS 788.33: found 786.3.

Step 16, To a solution of ((6 3 S,4S,Z)-l 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5 ,7- dioxo-1 L -(2,2,2 trifluoroethyl)-6',6 2 ,6 3 ,8 4 ,6 5 ,6 6 -hexahydro-l 1 W-8-oxa-2(5,3)-thiadiazola-1 (5,3)-indola- 6(1 .3)-pyridazinacycloundecaphane-4-yl)carbamate (300.0 g, 0.38 mmol) and DCM (2.0 mL) at 0 c C was added TFA (1 .0 L) dropwise. The resulting mixture was stirred for 2 h at room temperature then concentrated under reduced pressure. The residue was basified to pH 8 with saturated NaHGOa (aq.).

The mixture was extracted with EtOAc (3 x 20 mL). The organic phase was concentrated under reduced pressure to afford (6 3 S,4S,2)-4-amino-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-1 1 -(2,2,2- trifiuoroethyl)-6 1 ,6 2 , 6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 W-8-oxa-2(5,3)-thiadiazola-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-5,7-dione (270 mg, crude) as a solid. LCMS (ESI): /z [M+H] calc’d for C 3 3H 3 8F3N7O4S 686.27; found 686.1 .

Step 17, To a solution of (6 3 S,4S,2)-4-amino-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10- dimethyl-1 1 -(2,2,2-trifluoroethyl)-6’ ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 W-8-oxa-2(5,3)-thiadiazola-1 (5,3)-indola- 6(1 ,3)-pyridazinacycloundecaphane-5,7-dione (160.0 g, 0.23 mmol), (R)-2-(((1-benzhydrylazetidin-3- yi)oxy)methyl)-3-methylbutanoic acid (123.70 mg, 0.35 mmol) and DMF (2.0 mL) at 0 °C were added DIPEA (603.09 mg, 4 660 mmol) and COMU (119.91 mg, 0.28 mmol) in DMF (0.5 mL). The resulting mixture was stirred for 2 h at room temperature under an argon atmosphere. The reaction was quenched with H 2 O/lce and extracted with EtOAc (3 x 20 mL). The organic phase was washed with brine (3 x 10 mL) and concentrated under reduced pressure. The residue was purified by Prep-TLC to afford (2R}-2- (((1-benzhydrylazetidin-3-yl)oxy)methyl)-N- ((6 3 S,4S,Z)-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-1 1 -(2,2,2-triliuoroethyl)-6 1 ,6 2 ,6 3 , 6 4 ,6 5 ,6 6 -hexahydro-1 1 t-8-oxa-2(5,3)-thiadiazola- 1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)-3-methylbutanamide (160 mg, 67.2% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 5 sHesFaNeOeS 1021 .46; found 102 .4.

Step 18. To a solution of (2R)-2-(((1-benzhydrylazetidin-3-yl)oxy)methyl)-N -((6 3 S,4S,2)-1 2 -(2- ((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-1 1 -(2,2,2-trifluoroeihyl)-6 1 ,6 2 , 6 3 6 4 ,6 5 ,6 6 - hexahydro-1 1 H-8-oxa-2(5,3)-thiadiazoia-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)-3- methylbutanamide (160.0 g, 0.16 mmol) and MeOH (5.0 mL) at 0 °C was added (BOC)KO (85.49 mg, 0.39 mmol) dropwise followed by Pd/C (320.0 mg) in portions. The resulting mixture was stirred overnight at room temperature under a hydrogen atmosphere. The resulting mixture was filtered and the filter cake was washed with EtOAc (3 x 20 mL). The filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC to afford tert-butyl 3-((2F?)-2-(((6 3 S,4S,2)-1 2 -(2-((S)-1-methoxyethyl)pyridin-3- yl)-10,10-dimethyl-5,7-dioxo-1 1 -(2,2,2-trifiuoroethyl)-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-2(5,3)- thladlazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)carbamoyl)-3--methylbuto xy)azetldine-· 1-earboxylate (80 mg, 53.5% yield) as a solid. LCMS (ESI): m/z [M+H] cale’d for C 4 7H61F3N8O8S 955.44; found 955.2. Step 19, To a solution of tert-butyl 3-((2 ?)-2-(((6 3 S,4S,Z)-1 2 -(2-((S)-1-methGxyethyl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-1 1 -(2,2,2-trifluoroethyl)-6 1 ,6 2 ,6 3 ,6 4 l 6 5 ,6 6 -hexahydro-1 1 W-8-oxa-2(5,3)-thiadiazola-

I (5 s 3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)carbamoyl)-3-methylbutox y)azetidine-1- carboxylate (120.0 mg, 0. 3 mmol) and DCM (0.80 mL) at 0 °C was added TFA (0.4 mL) dropwise and the resulting mixture was stirred for 2 h at room temperature. The mixture was basified to pH 8 with saturated NaHCO 3 (aq.). The mixture was extracted with EtOAc (3 x 10 mL) and concentrated under reduced pressure. The residue was purified by reverse fiash chromatography to afford (2/ : ?)-2-((azetidin- 3-yloxy)methyl)-N- ((6 3 S,4S,Z)-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5 ,7-dioxo-1 '-(2,2,2- trifiuoroethyl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 ' H-8-oxa-2(5,3)-thiadiazola-1 (5,3) -indola- 6(1 ,3) - pyridazinacycloundecaphane-4-yl)-3-methylbutanamide (40 g, 37.2% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C^HssFsNeOeS 855.38: found 855.3.

Step 20, To a solution of (2fl)-2-((azetidin-3-yloxy)methyl)-N- ((6 3 S,4S,2)-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-1 '-(2,2,2-trifiuoroethyl)-8 1 ,8 2 ,6 3 ,6 4 ,6 5 ,8 6 -hexahydro-

I I H-8-oxa-2(5,3)-thiadiazola-1 (5,3)-indola-8(1 ,3)-pyridazinacycloundecaphane-4-yl)-3-methylbutanamide (32.0 mg, 0.037 mmol), 4-(dimethylamino)-4-methylpent-2-ynoic acid (11.62 mg, 0.074 mmol) and DMF (0.50 mL) at 0 °C were added DIPEA (19349 mg, 1 .48 mmol) and COMU (19.23 mg, 0,044 mmol) in DMF (0.1 mL) dropwise. The resulting mixture was stirred for 2 h at room temperature under an argon atmosphere. The reaction was quenched with H 2 0/lce and extracted with EtOAc (3 x 20 mL). The organic phase was washed with brine (3 x 10 ml) and concentrated under reduced pressure. The crude product (60 mg) was purified by reverse phase chromatography to afford (2ff)-2-(((1-(4-(dimethylamino)-4- methylpent-2-ynoyi)azetidin-3-yl)oxy)methyl)-N -((6 3 S,4S,Z^-1 2 -(2-((S)-1-mcthoxyethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-1 1 -(2,2,2-trifiuoroethyl)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydiO-1 1 F/-8-oxa-2(5,3)-thiadiaz.oia-

1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)-3-methylbutanamide (11 .7 g, 30.9% yield) as a solid. 1 H NMR (400 MHz, DMSO-cfe) 58.80 (dd, J = 4.7, 1.8 Hz, 1H), 8.58 (s, 1H), 8.30 (d, J = 8.9 Hz, 1H), 7.92 (d, J = 8.6 Hz, 1H), 7.84-7.69 (m, 2H), 7.56 (dd, J = 7.8, 4.8 Hz, 1H), 5.78 (t, J = 8.6 Hz, 2H), 5.10 (d, J = 12.1 Hz, 1H), 4.91 (dd, J= 16.9, 8.8 Hz, 1H), 4.31 (d, J = 6.6 Hz, 6H), 4.05 (dd, J = 16.3, 6.6 Hz, 2H), 3.87 (d, J = 6.3 Hz, 1H), 3.75 (d, J = 11.3 Hz, 1H), 3.61 (d, J = 11.0 Hz, 1H), 3.53 (d, J= 9.8 Hz, 1H), 3.45 (s, 3H), 3.25 (s, 1H), 3.09 (d, J = 10.4 Hz, 1H), 3.01 (d, J = 14.5 Hz, 1H), 2.79 (s, 1H), 2.45-2.35 (m, 2H), 2.20 (d, J = 5.4 Hz, 6H), 2.15 (d, J = 12.0 Hz, 1H), 1.81 (d, 2H), 1.74-1.65 (m, 1H), 1.54 (s, 1H), 1.41-1.30 (m, 9H), 1.24 (s, 1H), 0.94 (s, 3H), 0.90-0.81 (m, 5H), 0.29 (s, 3H). LCMS (ESI): m/z [M+H] calc’d for CSOHMFSNSOVS 992.47; found 992.5.

The following table of compounds (Table 3) were prepared using the aforementioned methods or variations thereof, as is known to those of skill in the art.

Table 3: Exemplary Compounds Prepared by Methods of the Present Invention

Blank = not determined fetched Pair Analysis FIGs. 1A-1B compare the potency in two different cell-based assays of compounds of Formula

BB of the present invention (points on the right) and corresponding compounds of Formula AA (points on the left) wherein a H is replaced with (S) Mo. The y axes represent pERK EC50 (FIG. 1A) or CTG IC50 (FIG. I B) as measured in an H358 cell line. Assay protocols are below. The linked points represent a matched pair that differs only between H and (8)Me substitution. Each compound of Formula BB demonstrated reduced potency in cell assays compared to the corresponding compound of Formula AA.

Biological Assays Potency assay: pERK

The purpose of this assay is to measure the ability of test compounds to inhibit K-Ras in cells. Activated K-Ras induces increased phosphorylation of ERK at Threonine 202 and Tyrosine 204 (pERK). This procedure measures a decrease in cellular pERK in response to test compounds. The procedure described below in NGI-H358 cells is applicable to K-Ras G12C.

Note: This protocol may be executed substituting other cell lines to characterize inhibitors of other RAS variants, including, for example, AsPC-1 (K-Ras G12D), Capan-1 (K-Ras G12V), or NCI- H1355 (K-Ras G13C).

NCI-H358 cells were grown and maintained using media and procedures recommended by the ATCC. On the day prior to compound addition, cells were plated in 384-well cell culture plates (40 mI/weli) and grown overnight in a 37°G, 5% C02 incubator. Test compounds were prepared in 10, 3-fold dilutions in DMSG, with a high concentration of 10 mM. On the day of assay, 40 nL of test compound was added to each well of cell culture plate using an Echo55Q liquid handler (LabCyte©). Concentrations of test compound were tested in duplicate. After compound addition, cells were incubated 4 hours at 37°C, 5% C02. Following incubation, culture medium was removed and cells were washed once with phosphate buffered saline.

In some experiments, cellular pERK level was determined using the AiphaLISA SureFire Ultra p- ERK1/2 Assay Kit (PerkinElmer). Cells were lysed in 25 pL lysis buffer, with shaking at 800 RPM at room temperature. Lysate (10 pL) was transferred to a 384-well Opti-plate (PerkinElmer) and 5 pL acceptor mix was added. After a 2-hour Incubation in the dark, 5 pL donor mix was added, plate was sealed, and incubated 2 hours at room temperature. Signal was read on an Envision plate reader (PerkinElmer) using standard AiphaLISA settings. Analysis of raw data was carried out in Excel (Microsoft) and Prism (GraphPad) Signal was plotted vs. the decadal logarithm of compound concentration, and IC 5 o was determined by fitting a 4-parameter sigmoidal concentration response model.

In other experiments, cellular pERK was determined by In-Cell Western Following compound treatment, cells were washed twice with 200 mΐ. tris buffered saline (TBS) and fixed for 15 minutes with 150 μL 4% paraformaldehyde in TBS. Fixed cells were washed 4 times for 5 minutes with TBS containing 0.1% Triton X-100 (TBST) and then blocked with 100 pL Odyssey blocking buffer (LI-COR) for 60 minutes at room temperature. Primary antibody (pERK, GST-4370, Cell Signaling Technology) was diluted 1 :200 in blocking buffer, and 50 pL was added to each well and incubated overnight at 4°C. Cells were washed 4 times for 5 minutes with TBST. Secondary antibody (IR-800CW rabbit, LI-GOR, diluted 1 :800) and DNA stain DRAQ5 (LI-GOR, diluted 1 :2000) were added and incubated 1-2 hours at room temperature. Cells were washed 4 times for 5 minutes with TBST. Plates were scanned on a Li-COR Odyssey CLx Imager. Analysis of raw data was carried out in Excel (Microsoft) and Prism (GraphPad). Signal was plotted vs. the decadal logarithm of compound concentration, and lC 5 o was determined by fitting a 4-parameter sigmoidal concentration response model.

The following compounds exhibited a pERK EC50 of under 5 uM (H358 KRAS G12C):

A48,A15,A272,A174, A163,A453,A447,A279,A240,A214,A225,A136,A226,A219,A228,A21 ,A12,A78,A424 ,A219,A378,A224,A4,A53,A187.A218,A213,A314,A220,A208,A24,A9, A126,A345,A48,A203,A210,A184, A 469,A388,A113,A328,A893,A639,A364,A1 QG,A249,A486,A307,A347,A33,A210,A192,A285,A488,A185, A 612,A109,A284,A200,A2,A6,A606,A325,A139,A498,A393,A561 ,A125,A494,A547,A215,A258,A195,A259, A212,A637,A53,A83,A68,A178, A189,A205,A78,A254,A690,A583,A14,A19,A92,A576,A278,A331 ,A42,A6 7,A2G9,A350,A582,A652,A703,A623,A191 ,A241 ,A199, A193,A478,A251 ,A177,A222,A23,A59,A26,A21 1 , A108,A279,A120,A7,A134.A521 ,A116,A467,A834,A729,A151 ,A110,A277,A340,A221 ,A723,A13,A442,A6 11 ,A50,A190,A553,A698,A211 .A303.A813.A37.A146,A866,A688,A216,A390,A548,A238,A160, A183, A 16 4,4451 ,A4S1 ,4524,41 ,4186, A37, 4635, 471 ,4269,4289,4489,4400,4731 ,A497, 4568, 4274,4253,4471 ,A 720,4241 ,A179, A180,A426,A117,A363,A716,A423,A217,A708, A227,A3,A12,A8,A381 ,A84,A408,A85,A1 71 ,A263,A473,A258,A564,A118,A103.A565.A641 ,A655,A47,A11 .A392.A169,A487,A640,A206,A449,A35 8,4192, A148,A4,A41 ,A5,A18,4301 , A10.A65.A554, A159.A264.A99, A79.A142,4143,A25,A98,A80, A101 ,A 730, A212,A359,A61 ,A441 ,A283,A413.A717,4145, A182,A82,A181 ,4233, 4232, A634,A495, 434, A251 ,A53 9,A632,A54,A327,A37,A198, 4807, 4645, 435, 4214, A225, 4838, A4Q,A52, 4288, A448, 4575, 4176,A593,A1 5,A17,A94,A170,4713,A93,A402,A64,A281 ,A399,A422,A214,4225,4625,431 ,4119,A135,4281 ,4676,47 09.A81 ,A32,A833,A39, 4646, 4662, A124, 4732, 4320, A81 ,A187,A354,A45,A570,A165,A68,A20,A455,A43 1 ,A270,A250,A457,A153.A404.A710,4541 ,4127, A373,A369, 4557, A349, 4598, A618,A60,A636,A499,A87, 4156, 4680, A477.A406, A330, 4202, 4535, A617,A737,A201 ,A302,A722,A209,A374,A631 ,A29,A555,A420, A380,A111 ,A306,A173,A628,A672,A51 ,A167,A588,A512,A194,A282,A412,4701 ,A583, 4396, 4678, A649, 427, 4204, A626, 4257, A614,A409,A172,A372,A353,A58,A728,A74,A619,4144, A183, A538, 4445, 4531 ,43 60,4361 .4459.A536, 4344, A267, 4574, 4677, A530, 4415,430,A73,A152,A490,A702,A714,A483,A567,A43, 4310,4319,486,4321 ,A656,A739,A115,A130,4155,A608,A648,A168,A485,A738,A129,A650 ,A715.A488, 4147,4121 ,4470,4115,A133,4510,4421 ,A309,A335,A387, 4386, A734,A95, 4430, A604, 4458, 4592, A384, 4664, A197, 4725, 489, 483, A586, 4622, A305, 4498, A668, 4427, 4630, A158, 4644, A735,A70, 4683, A352, 43 41 ,4719,A674,A70,444,A501 ,A438,A698,A377,A417,A154,A433,A104, A184,A603,A280,A712,A237,A10 5, A394, 4605,4517,A704,A566,A77,A356,A454,A600,A643,A112,A569,A529,A 247,A463,A437,A718,A47 2,A461 ,4558, 448, A671 ,A395,A670,A681 ,A687,A382,A82,A686,A342,A436,A296,A16,A545,A533,A416, 4149,4207,4371 , A596,A675,A132,A419,A56,A579,A733,A573,A707,A597,A697,A75,A6 53,A362,A615,4 332, 469, A162, A128, 4432, A654,A22, 4397, A526, 4582, A41 S.A91 ,A260,A97,A191 ,A55,A581 ,A375,A522, 4108,4367,4610.A552.A571 ,A57,A543,A661 ,A138, A196, 4246, 4337, A446, 4265, 496, A509, 4123,A627,A 651 ,4682,4157,A572,A624,A691 ,4532, 4462, A580, 4695, 4186,4316, 4540, A590, 4665, 4244,4166.A587, 4629, 4595, 4518.A519,A131 ,4502, A726,A452,A141 ,A181 ,4262, 4338, A155,A389,A124,A275,A414.A546 ,A679,A425, 4669, A2S,A520, 488, A131 ,4589,4621 ,4182,4297,4594,4283,4194, 4250, 4336, 4706, 4252, 4440,4107, A724, 4525,4388, 4175,4300,4333,4659,4346,4150, 4476, 4368, A528, 4503, 4504,4505,4684 ,476,4736,4551 ,4383,4491 , 4492, A493, 4410,4316,4295,4559,4511 ,438,4140, 4663, 4334, 4700, 4692, A348,A584,A513, 4657, 4328, 4515,4317,4135,4660,4351 ,4544, 4281 ,4685,A602,A556,A385,A326,A464 ,4465,4403, 4133, 4299, 4667, 4255, 4334, 4256, 4585, 4642, 4133, 4443, 4435, 4560, 4444, 4439, 4324, 412 0, 4407, 4527, 4245, 4370, A537, 4247, 4474, 4475, 4705, A323, 4112, 4298,4609,4673, A292, 4599, 4132, 41 45,4266,4601 ,4466,4549, 4379, 4727, 4167,4711 ,A75, 476,4121 ,4357, 4620, 4316,4479,4290,4339,432 2,4376,4456,4391 ,4291 ,4550, 4343, 4721 ,A689,A411 ,4578,4618, 4534, A365, 4858, A699, 4577, 4647, 45 91 , 4542, A279, 4294.

Determination of Ceii Viability in HAS Mutant Cancer Ceii Lines Protocol: CeliTiter-Glo® Cell Viability Assay

Note The following protocol describes a procedure for monitoring ceii viability of K-Ras mutant cancer ceii lines in response to a compound of the invention. Other RAS isoforms may be employed, though the number of cells to be seeded will vary based on ceil line used.

The purpose of this cellular assay was to determine the effects of test compounds on the proliferation of three human cancer cell lines (NCI-H358 (K-Ras G12C), AsPC-1 (K-Ras G12D), and Capan-1 (K-Ras G12V)) over a 5-day treatment period by quantifying the amount of ATP present at endpoint using the CeiiTiter-Gio® 2.0 Reagent (Promega).

Cells were seeded at 250 cells/well in 40 pL of growth medium in 384-weli assay plates and incubated overnight in a humidified atmosphere of 5% CO 2 at 37°C. On the day of the assay, 10 mM stock solutions of test compounds were first diluted into 3 mM solutions with 100% DMSO. Well-mixed compound solutions (15 pL) were transferred to the next wells containing 30 pL of 100% DMSO, and repeated until a 9-concentration 3-fold serial dilution was made (starting assay concentration of 10 μM). Test compounds (132.5 nL) were directly dispensed into the assay plates containing cells. The plates were shaken for 15 seconds at 300 rpm, centrifuged, and Incubated in a humidified atmosphere of 5% CO? at 37 °C for 5 days. On day 5, assay plates and their contents were equilibrated to room temperature for approximately 30 minutes. CelITiter-Glo® 2.0 Reagent (25 mI_) was added, and plate contents were mixed for 2 minutes on an orbital shaker before incubation at room temperature for 10 minutes. Luminescence was measured using the PerkinElmer Enspire. Data were normalized by the following: (Sample signal/Avg. DMSO) * 100. The data were fit using a four-parameter logistic fit.

The purpose of this biochemical assay was to measure the ability of test compounds to facilitate ternary complex formation between a nucleotide-loaded K-Ras isoform and Cyclophilin A; the resulting ternary complex disrupts binding to a BRAF RBD construct, inhibiting K-Ras signaling through a RAF effector. Data is reported as IC50 values.

In assay buffer containing 25 M HEPES pH 7.3, 0.002% Tween20, 0.1% BSA, 00 mM NaCI and 5 mM MgGL, tagless Cyclophilin A, His6-K-Ras-GMPPNP, and GST-BRAF RBD were combined in a 384-weli assay plate at final concentrations of 25 μM, 12.5 nM and 50 nM, respectively. Compound was present in plate wells as a 10-point 3-fold dilution series starting at a final concentration of 30 μM. After incubation at 25°C for 3 hours, a mixture of Anti-His Eu-W1024 and anti-GST ailophycocyanin was then added to assay sample wells at final concentrations of 10 nM and 50 nM, respectively, and the reaction incubated for an additional 1 .5 hours. TR-FRET signal was road on a microplate reader (Ex 320 nm, Em

665/615 nrn). Compounds that facilitate disruption of a K-Ras:RAF complex were identified as those eliciting a decrease in the TR-FRET ratio rolative to DMSO control wells.

Table 4; Biological Assay Data for Representative Compounds of the Present invention

Table 5, Additional H358 Ceil Viability assay data (K-Ras G12C, lC50, uM): * Key: ++: IC50 > 1 uM +++: 1 uM > IC50 > 0.1 uM ++: 0.1 uM > iC50 > 0.01 uM

4: IC50 < 0.01 uM

Additional Ras-Raf disrupisorj/FRET/IVlOA assay data (IC5Q, uM)·, * Key:

+++++: IC50 ³ 10 uM ++++: 10 uM > IC50 > 1 uM 1 uM > IC50 > 0.1 uM ++: 0.1 uM > IC50 > 0.01 uM +: IC50 < 0.01 uM Table 6. KRAS G12S FRET data

Table 7, KRAS G12D FRET data

Table 8. KRAS G13C FRET data

Table 9, KRAS G12V FRET data

Table 10, KRAS WT FRET data

Table 11, KRAS G12C FRET data

Table 12. KRAS G13D FRET data

Table 13, KRAS G61H FRET data

Table 14. MBAS G12C FRET data

Table 15. NBAS 061 R FRET data

Table 16. NBAS 061 K FRET data

Table 17, MR AS WT FRET data

In vitro Cell Proliferation Panels Potency for inhibition of cell growth was assessed at CrownBio using standard methods. Briefly, cell lines were cultured in appropriate medium, and then piated in 3D methyleeilulose. Inhibition of celi growth was determined by CelITiter-Glo® after 5 days of culture with increasing concentrations of compounds. Compound potency was reported as the 50% inhibition concentration (absolute IC50).

The assay took place over 7 days. On day 1 , cells In 2D culture were harvested during logarithmic growth and suspended in culture medium at 1x105 cells/ml. Higher or lower cell densities were used for some cell lines based on prior optimization 3.5 ml of cell suspension was mixed with 6.5% growth medium with 1% methyleeliuiose, resulting in a cell suspension in 0.65% methylceilulose. 90 mI of this suspension was distributed in the wells of 296-weli plates. One plate was used for day 0 reading and 1 plate was used for the end-point experiment. Plates were incubated overnight at 37 C with 5% GO2. On day 2, one plate (for to reading) was removed and 10 ml growth medium plus 100 mI GelITiter-Gio® Reagent was added to each well. After mixing and a 10 minute incubation, luminescence was recorded on an EnVision Multi-Label Reader (Perkin Elmer). Compounds in DMSO were diluted in growth medium such that the final, maximum concentration of compound was 10 μM, and serial 4 -fold dilutions were performed to generate a 9-point concentration series. 10 ml of compound solution at 10 times final concentration was added to weils of the second plate. Plate was then incubated for 120 hours at 37C and 5% C02. On day 7 the plates were removed, 100 ml GellTiier-Glo© Reagent was added to each well, and after mixing and a 10 minute incubation, luminescence was recorded on an EnVision Multi- Label Reader (Perkin Elmer). Data was exported to GeneData Screener and modeled with a sigmoidal concentration response model in order to determine the IC50 for compound response.

Not ail cell lines with a given RAS mutation may be equally sensitive to a RAS inhibitor targeting that mutation, due to differential expression of efflux transporters, varying dependencies on RAS pathway activation for growth, or other reasons. This has been exemplified by the cell line KYSE-410 which, despiteB having a KRAS G12G mutation, is insensitive to the KRAS G12G (OFF) Inhibitor MRTX-849 (Hallin et al., Cancer Discovery 10:54-71 (2020)), and the cell line SW1573, which is insensitive to the KRAS G12C (OFF) inhibitor AMG510 (Canon et al., Nature 575:217-223 (2019)).

Table 18: C50 values for various cancer cell lines with Compound B

* Key: low sensitivity: IC50 > 1 uM moderately sensitive: 1 uM > IC5G > 0.1 uM very sensitive: IC50 < 0.1 uM

Table 19: Summary of IC50 results for various cancer cell Sines with several compounds of the present invention (Compounds B and E-M)

‘Key:

(L) low sensitivity: IC50 > 1 uM (M) moderately sensitive: 1 uM > IC50 > 0.1 uM

(V) very sensitive: IC50 < 0.1 uM in vivo NSCLC K-Ras 012C Xenograft Mo els Compound A: Methods: The effects of a compound of the present invention, Compound A (H358 pERK K-Ras G12G EC50: 0.001 uM), on tumor cell growth in vivo were evaluated in the human non-smaii cell lung cancer NCi-H35S KRASG12C xenograft model using female BALB/c nude mice (6-8 weeks old). Mice were implanted with NCI-H358 tumor cells in 50% Matrigel (5 x 106 cells/mouse) subcutaneously in the flank.

At the indicated tumor volume (dotted line, FIG. 2A), mice were randomized to treatment groups to start the administration of test articles or vehicle. Compound A was administered by oral gavage dally at the dose of 100 g/kg. Body weight and tumor volume (using calipers) was measured twice weekly until study endpoints. Spaghetti plot (FIG. 2B) shows the tumor volume change in individual tumors during the course of treatment.

Results:

FIG. 2A shows Compound A dosed at 100 g/kg by dally oral gavage led to tumor regression in NCI-H358 KRASG12C xenograft model, which is a sensitive model to KRASG12C inhibition alone. The spaghetti titer plot (FIG. 2B) displaying individual tumor growth is shown next to the tumor volume plot (FIG. 2A). Over the treatment course of 28 days, Compound A drove tumor regression in all 10 animals bearing NCI-H358 KRASG12C tumors.

Compound B;

Methods:

The combinatorial effect of a compound of the present invention, Compound B (H358 pERK K- Ras G12C EC50: 0.003 uM), with cobimetinib on tumor cell growth In vivo were evaluated In the human non-small cell lung cancer NGI-H358 KRASG12C xenograft model using female BALB/c nude mice (6-8 weeks old). Mice were implanted with NCI-H358 tumor cells in 50% Matrigel (5 x 106 cells/mouse) subcutaneously in the flank. At indicated tumor volume (dotted line, FIG. 3A), mice were randomized to treatment groups to start the administration of test articles or vehicle. Compound B was administered by intermittent (twice weekly) intravenous injection at the dose of 50 mg/kg. Cobimetinib was administered by dally oral gavage at 2.5 mg/kg. The combination of Compound B and cobimetinib at their respective single-agent dose and regimen was also tested. Body weight and tumor volume (using calipers) was measured twice weekly until study endpoints. End of study responses in individual tumors were plotted as a waterfall plot (FIG. 3B), and the numbers indicate number of tumor regression in each group. Tumor regression is defined as greater than 10% reduction of tumor volume at the end of study relative to initial volume.

Results:

FIG. 3A shows the combination of intermitent intravenous administration of Compound B at 50 mg/kg plus dally oral administration of cobimetinib at 2.5 mg/kg drove tumor regression, whereas each single agent led to tumor growth inhibition. End of study responses were shown as waterfall plois (FIG. 3B), which indicate 6 out 10 mice had tumor regression in the combination group, whereas no tumor regressions recorded in each single agent group.

Compound C:

Methods:

The combinatorial effect of a compound of the present invention, Compound C (H358 pERK K- Ras G12C EC50: 0.007 uM), with a SHP2 inhibitor, RMC-4550, on tumor cell growth in vivo were evaluated in the human non-smail cell lung cancer NCI-H358 KRASG12C xenograft model using female BALB/c nude mice (6-8 weeks old). Mice were implanted with NCI-H358 tumor cells in 50% Matrigei (5 x 106 celis/mouse) subcutaneously in the flank. At indicated tumor volume (dotted line, FIG. 4A), mice were randomized to treatment groups to start the administration of test articles or vehicle. Compound C was administered by once weekly intravenous injection at the dose of 60 mg/kg. SHP2 inhibitor was administered by dally oral gavage at 30 mg/kg. The combination of Compound C and SHP2 inhibitor at their respective single-agent dose and regimen was also tested. Body weight and tumor volume (using calipers) was measured twice weekly until study endpoints. End of study responses in Individual tumors were plotted as a waterfall plot (FlG. 4B), and the numbers indicate number of tumor regression in each group. Tumor regression is defined as greater than 10% reduction of tumor volume at the end of study relative to initial volume.

Results:

In FIG 4A, the combinatorial activity of once weekly intravenous administration of Compound C at 60 mg/kg plus dally oral administration of SHP2 inhibitor at 30 mg/kg is shown. The combination treatment had similar anti-tumor activity as the single agent SHP2 inhibitor, but the combination treatment ied to 8 out of 10 mice with tumor regression, whereas single agent SHP2 inhibitor led to 5 out of 10 mice with tumor regressions. Single agent Compound C administered once weekly via intravenous injection led to tumor growth inhibition with one tumor regression.

CelS Proliferation Assay Methods:

NCI-H358 cells were plated in 12-well tissue culture plates at a density of 100,000 cells/well in RPMI 1640 (10% FBS, 1% PenStrep) and cultured overnight at 37°C, 5% C02. The following day, cells were treated with either trametinib (10 nM) or a compound of the present invention, Compound D (H358 pERK K-Ras G12C EC50: 0.024 uM), (17 nM). These concentrations represent the EC50 values from a 72-hour proliferation assay using the CeilTiter-Glo® reagent (Promega). Additionally, cells were treated with the combination of trametinib and Compound D at the above indicated concentrations. The plate was placed in the Incucyte S3 live cell analysis system (37°C, 5% C02) and confluence was measured by recording images at 6-hour intervals for a maximum of 40 days, or until wells reached maximal confluence. Media and drug were replaced at 3-4 day intervals. Data are plotted as % confluence over the time course of the experiment for each single agent and respective combination (FIG. 5).

Results:

As shown in FIG. 5, treatment of NCI-H358 cells with submaximal (EC5Q) concentrations of Compound D or MEK inhibitor results in a short period of growth inhibition, followed by proliferation. Cells reach maximal confluence in -10 days after the addition of drug. The combination of the MEK inhibitor, trametinib, with Compound D resulted in complete and sustained inhibition of cell growth throughout the duration of the assay.

While the invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure come within known or customary practice within the art to which the invention pertains and may be applied to the essential features set forth herein. All publications, patents and patent applications are herein Incorporated by reference in their entirety to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety.

Appendix C-1

RAS fNHfBlTGRS

Background

The vast majority of small molecule drugs act by binding a functionally Important pocket on a target protein, thereby modulating the activity of that protein. For example, cholesterol-lowering drugs known as statins bind the enzyme active site of HMG-CoA reductase, thus preventing the enzyme from engaging with its substrates. The fact that many such drug/target Interacting pairs are known may have misled some into believing that a small molecule modulator could be discovered for most, if not ail, proteins provided a reasonable amount of time, effort, and resources. This is far from the case. Current estimates are that only about 10% of all human proteins are targetable by small molecules. Bojadzlc and Buchwald, Curr Top Med Chem 18: 674-699 (2019). The other 90% are currently considered refractory or intractable toward above-mentioned small molecule drug discovery. Such targets are commonly referred to as “undruggable.” These undruggable targets include a vast and largely untapped reservoir of medically important human proteins. Thus, there exists a great deal of Interest in discovering new molecular modalities capable of modulating the function of such undruggabie targets.

It has been well established in literature that Ras proteins (K-Ras, H-Ras and N-Ras) play an essential role in various human cancers and are therefore appropriate targets for anticancer therapy indeed, mutations In Ras proteins account for approximately 30% of ail human cancers in the United States, many of which are fatal. Dysregulation of Ras proteins by activating mutations, overexpression or upstream activation is common in human tumors, and activating mutations in Ras are frequently found in human cancer. For example, activating mutations at codon 12 in Ras proteins function by inhibiting both GTPase-activating protein (GAP)-dependent and intrinsic hydrolysis rates of GTP, significantly skewing the population of Ras mutant proteins to the “on” (GTP-bound) state (Ras(ON)), leading to oncogenic MARK signaling. Notably, Ras exhibits a picomolar affinity for GTP, enabling Ras to be activated even in the presence of low concentrations of this nucleotide. Mutations at codons 13 (e.g., G13D) and 61 (e.g., G61 K) of Ras are also responsible for oncogenic activity in some cancers.

Despite extensive drug discovery efforts against Ras during the last several decades, a drug directly targeting Ras is still not approved. Additional efforts are needed to uncover additional medicines for cancers driven by the various Ras mutations.

Summary

Provided herein are Ras inhibitors. The approach described herein entaiis formation of a high affinity three-component complex between a synthetic ligand and two intracellular proteins which do not interact under normal physiological conditions: the target protein of interest (e.g., Ras), and a widely expressed cytosolic chaperone (presenter protein) in the cell (e.g., cyclophilm A). More specifically, In some embodiments, the inhibitors of Ras described herein induce a new binding pocket in Ras by driving formation of a high affinity tri-complex between the Ras protein and the widely expressed cytosolic chaperone, cyclophiiin A (CYRA). Without being bound by theory, the inventors believe that one way the inhibitor effect on Ras is effected by compounds of the invention and the complexes the form is b steric occlusion of the interaction site between Ras and downstream effector molecules, such as RAF and PI3K, which are required for propagating the oncogenic signal.

As such, in some embodiments, the disclosure features a compound, or pharmaceutically acceptable salt thereof, of structural Formula I: wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH{R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 10- membered heteroarylene;

B is absent, -CH(R 9 )-, or >C=CR 9 R 9' where the carbon is bound to the carbonyl carbon of - N(R 11 )C(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C 1 -C 4 alkylene, optionally substituted Gi-O. alkenylene, optionally substituted C 1 -C 4 heteroalkylene, -C(O)O-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(O)NH-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C 1 -C 4 heteroalkylene, or 3 to 8-membered heteroarylene;

L is absent or a linker;

W is hydrogen, cyano, S(O) 2 R’, optionally substituted amino, optionally substituted arnldo, optionally substituted C 1 -CA alkoxy, optionally substituted G1-C 4 hydroxyalkyl, optionally substituted G1-C 4 aminoalkyl, optionally substituted C 1 -C 4 haloalkyl, optionally substituted G1-C 4 alkyl, optionally substituted GI-G 4 guanidinoalkyl, C 0 -C 4 alkyl optionally substituted 3 to 11-membered heterocycloalkyl, optionally substituted 3 to 8-membered cycloalkyl, or optionally substituted 3 to 8-membered heteroaryl;

X 1 is optionally substituted C 1 -C 2 alkylene, NR, O, or S(G) r< ;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2; R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R', C(O)OR’, C(O)N(R’) 2 , S(O)R’, S(O) 2 R’, or S(O) 2 N(R') 2 ; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CM, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 is CH, CH 2 , or N;

Y 6 is C(O), CH, CH 2 , or N; R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl, or R 1 and R 2 combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 2 is absent, hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl;

R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl ;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C 1 -C 3 alkyl), C=O, C=S, C==NH, optionally substituted 3 to 6-membered eydoalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7a and R 8a are, independently, hydrogen, halo, optionally substituted C 1 -C 3 alkyl, or combine with the carbon to which they are attached to form a carbonyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl; R 9 is hydrogen, F, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-merrsbered heterocycloalky I, or R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14- embered heterocycloalkyl;

R 9' is hydrogen or optionally substituted C 1 -C 6 alkyl; R 10 is hydrogen, halo, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl; R 10a is hydrogen or halo; R 11 is hydrogen or C 1 -C 3 alkyl. Also provided are pharmaceutical compositions comprising a compound 0! Formula I, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient; and R 16 is hydrogen or C 1 -C 3 alkyl (e.g., methyl).

Also provided is a method of treating cancer In a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof.

In some embodiments, a method is provided of treating a Ras protein-related disorder in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof.

Further provided is a method of inhibiting a Ras protein in a cell, the method comprising contacting the cell with an effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof.

It is specifically contemplated that any limitation discussed with respect to one embodiment of the invention may apply to any other embodiment of the invention. Furthermore, any compound or composition of the invention may be used in any method of the invention, and any method of the invention may be used to produce or to utilize any compound or composition of the invention.

Brief Description of the Figures

FIG. 1A: A compound of the present invention, Compound A, exhibits PK-dependent RAS pathway modulation in a Capan-2 CDX model (PDAC, KRAS G12V7WT). Single dose compared to twice administered PK/PD measurement of Compound A. Second dose of Compound A delivered 8 hours following first dose, depicted by black arrow. All dose levels well tolerated. Tumor DUSP6 mRNA expression as percent of control graphed as bars on left y-axis. Dotted line indicates return to control level of DUSP6. Unbound plasma PK (nM) graphed as lines, plotted in Log 10 scale on right y-axis. N = 3/time point. Error bars represent standard error of the mean.

FIG. 1B: Combinatorial anti-tumor activity with a compound of the present invention, Compound A, and upstream SHP2 inhibition in a Capan-2 CDX model (PDAC, KRAS G12V/WT). Capan-2 cells were implanted in 50% Matrigel. Animals were randomized and treatment was initiated at average tumor volume of ~18Qmm3. Animals were dosed with SHP2 inhibitor RMC-4550 20 mg/kg po q2d, Compound A 100 mg/kg po bid, combination RMC-4550 and Compound A, or Control for 40 days. All dose levels were tolerated n = 10/group (n = 9 in Combination arm). Ms = no significance; " * p<0.001 by one-way ANOVA. Definitions and Chemical Terms

In this application, unless otherwise clear from context, (i) the term “a" moans "one or more"; (ii) the term "or" is used to mean "and/or" unless expiicitiy indicated to refer to alternatives only or the alternative are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and "and/or”; (ili) the terms "comprising” and "including" are understood to encompass itemized components or steps whether presented by themselves or together with one or more additional components or steps; and (Iv) where ranges are provided, endpoints are included.

As used herein, the term “about” is used to indicate that a value includes the standard deviation of error for the device or method being employed to determine the value. In certain embodiments, the term “about” refers to a range of values that fall within 25%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less in either direction (greater than or less than) of a stated value, unless otherwise stated or otherwise evident from the context (e.g., where such number would exceed 100% of a possible value).

As used herein, the term “adjacent” in the context of describing adjacent atoms refers to bivalent atoms that are directly connected by a covalent bond.

A “compound of the present invention” and similar terms as used herein, whether explicitly noted or not, refers to Ras inhibitors described herein, including compounds of Formula I and subformula thereof, and compounds of Table 1 and Table 2, as well as salts (e.g., pharmaceutically acceptable salts), solvates, hydrates, stereoisomers (Including atropisomers), and tautomers thereof.

The term “wild-type” refers to an entity having a structure or activity as found In nature in a “nor al” (as contrasted with mutant, diseased, altered, etc) state or context. Those of ordinary skill in the art will appreciate that wild-type genes and polypeptides often exist in multiple different forms (e.g , alleles).

Those skilled in the art will appreciate that certain compounds described herein can exist in one or more different isomeric (e.g., stereoisomers, geometric isomers, atropisomers, tautomers) or isotopic (e.g., in which one or more atoms has been substituted with a different isotope of the atom, such as hydrogen substituted for deuterium) forms. Unless otherwise indicated or clear from context, a depicted structure can be understood to represent any such isomeric or isotopic form, individually or in combination.

Compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated. Compounds of the present disclosure that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically active starting materials are known In the art, such as by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of olefins, C=N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present disclosure. Cls and trans geometric isomers of the compounds of the present disclosure are described and may be isolated as a mixture of isomers or as separated isomeric forms.

In some embodiments, one or more compounds depicted herein may exist In different tautomeric forms. As will be clear from context, unless explicitly excluded, references to such compounds encompass all such tautomeric forms. In some embodiments, tautomeric forms result from the swapping of a single bond with an adjacent double bond and the concomitant migration of a proton. In certain embodiments, a tautomeric form may be a prototropic tautomer, which is an isomeric protonation states having the same empirical formula and total charge as a reference form. Examples of moieties with prototropic tautomeric forms are ketone - enoi pairs, amide - imidic acid pairs, lactam - iactim pairs, amide - imidic acid pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, such as, 1H- and 3H-imidazoie, 1H-, 2H- and 4H-1 ,2,4-triazole, 1H- and 2H- isoindole, and 1H- and 2H-pyrazole. In some embodiments, tautomeric forms can be In equilibrium or sterically locked into one form by appropriate substitution. In certain embodiments, tautomeric forms result from acetal interconversion.

Unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. Exemplary isotopes that can be incorporated into compounds of the present invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, and iodine, such as 2 H, 3 H, "C, i3 C, 14 C, i3 N, 15 N, 15 G, 17 O, 13 O, 32 P, 33 P, 35 S, 18 F, 36 CI, 123 l and i25 l. lsotopieally-labeled compounds (e.g., those labeled with 3 H and 14 C) can be useful in compound or substrate tissue distribution assays. Tritiated (i.e., 3 H) and carbon-14 (l.e., 14 G) isotopes can be useful for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements). In some embodiments, one or more hydrogen atoms are replaced by 2 H or 3 H, or one or more carbon atoms are replaced by 13 C- or l4 C-enriched carbon. Positron emitting isotopes such as 1s O, 13 N, n C, and 18 F are useful for positron emission tomography (PET) studies to examine substrate receptor occupancy. Preparations of isotopically labelled compounds are known to those of skill in the art. For example, isotopicaliy labeled compounds can generally be prepared by following procedures analogous to those disclosed for compounds of the present invention described herein, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.

As is known in the art, many chemical entities can adopt a variety of different solid forms such as, for example, amorphous forms or crystalline forms (e.g., polymorphs, hydrates, solvate). In some embodiments, compounds of the present Invention may be utilized in any such form, including in any solid form. In some embodiments, compounds described or depicted herein may be provided or utilized in hydrate or soivate form.

At various places In the present specification, substituents of compounds of the present disclosure are disclosed in groups or in ranges. It is specifically intended that the present disclosure include each and every individual subcombination of the members of such groups and ranges. For example, the term "C 1 -C 6 alkyl” is specifically Intended to individually disclose methyl, ethyl, Cs alkyl, C 4 alkyl, C 5 alkyl, and C 6 alkyl. Furthermore, where a compound includes a plurality of positions at which substituents are disclosed in groups or In ranges, unless otherwise Indicated, the present disclosure is intended to cover individual compounds and groups of compounds (e.g., genera and subgenera) containing each and every individual subcombination of members at each position.

The term “optionally substituted X” (e.g., “optionally substituted alkyl”) is intended to be equivalent to “X, wherein X is optionally substituted” (e.g., “alkyl, wherein said alkyl is optionally substituted”) it is not intended to mean that the feature “X” (e.g., alkyl) per se is optional. As described herein, certain compounds of interest may contain one or more “optionally substituted" moieties. In general, the term “substituted”, whether preceded by the term “optionally” or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent, e.g., any of the substituents or groups described herein. Unless otherwise indicated, an "optionally substituted” group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. For example, in the term "optionally substituted C 1 -C 6 alkyl-C 2 -C9 heteroaryl,” the alkyl portion, the heteroaryl portion, or both, may be optionally substituted. Combinations of substituents envisioned by the present disclosure are preferably those that result in the formation of stable or chemically feasible compounds. The term “stable”, as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein.

Suitable monovalent substituents on a substitutable carbon atom of an "optionally substituted” group may be, independently, deuterium; halogen; -(CH 2 )o- 4 R°; -(CH 2 )O- 4 OR°; -O(CH 2 ) 0 - 4 R°; -O-(CH 2 ) 0 - 4 C(O)OR° ; -(CH 2 )O CH(OR°) 2 ; -(CH 2 ) 0 - 4 SR°; -(CH 2 )o- 4 Ph. which may be substituted with R°; -(CH 2 )o- 4 O(CH 2 )o-;Ph which may be substituted with R°; -CH==CHPh, which may be substituted with R°; -(CH 2 ) 0 - 4 O(CH 2 )o-i-pyridyi which may be substituted with R°; 4-8 membered saturated or unsafurafed heterocycloalkyl (e.g., pyridyi); 3-8 membered saturated or unsaturated eycloalkyl (e.g., cyclopropyl, cyclobutyl, or cyclopentyl); -NO 2 ; -CN; -N 3 ; -(CH 2 ) 0 - 4 N(R°) 2 ; -(CH 2 )o- 4 N(R°)C(O)R°; -N(R°)C(S)R°; -(CH 2 ) 0 - 4 N(R°)C(O)NR°2; -N(R°)C(S)NR°2; -(CH 2 ) 0 - 4 N(R°)C(O)OR°; - N(R°)N(R°)C(O)R°; -N(R°)N(R°)C(O )NR° 2 ; -N(R°)N(R°)C(O)OR°; -(CH 2 ) 0 - 4 C(O)R°; -C(S)R°; -(CH 2 ) 0 - 4 C(O)OR°; -(CH 2 ) 0 - 4 -C(O)-N(R°) 2 ; -(CH 2 )O -4-C(O)-N(R°)-S(O) 2 -R°; -C(NCN)NR° 2 ; -(CH 2 ) 0 - 4 C(O)SR°; -(CH 2 ) 0 - 4 C(O)OSiR° 3 ; -(CH 2 ) 0 - 4 OC(O)R°; -OC( 0)(CH 2 )O- 4 SR°; -SC(S)SR°; -(CH 2 ) 0 - 4 SC(O)R°; -(CH 2 ) 0 - 4 C(O)NR° 2 ; -C(S)NR c 2 ; -C(S)SR°; -(CH 2 ) 0 - 4 OC(O) NR° 2 ; -C(O)N(OR°)R°; -C(O)C(O)R°; -C(O)CH 2 C(O)R°; -C(NOR°)R°; -(CH 2 )O- 4 SSR°; -(CH 2 ) 0 - 4 S(O) 2 R°; -( CH 2 )O- 4 S(O) 2 OR°; -(CH 2 )O- 4 OS(O) 2 R°; -S(O) 2 NR° 2 ; -(CH 2 )O- 4 S(O)R°; -N(R°)S(O) 2 NR° 2 ; -N(R°)S(O) 2 R°; -N( OR°)R°; -C(NOR°)NR° 2 ; -C(NH)NR° 2 ; -P(O) 2 R°; -P(O)R° 2 ; -P(O)(OR°) 2 ; -OP(O)R° 2 ; -OP(O)(OR°) 2 ; -OP(O )(OR°)R°, -SiR°s; -(Cl 4 straight or branched alkylene)O-N(R°) 2 ; or -(C 1 -4 straight or branched alkylene)C(O)O-N(R°) 2 , wherein each R° may be substituted as defined below and is independently hydrogen, -C 1 -6 aliphatic, -CH 2 Ph, -O(CH 2 ) 0 - 1 Ph, -CH 2 -(5-6 membered heteroaryl ring), or a 3-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R°, taken together with their intervening ato (s), form a 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, which may be substituted as defined below.

Suitable monovalent substituents on R° (or the ring formed by taking two independent occurrences of R° together with their intervening atoms), may be, independently, halogen, -(CH 2 )o- 2 R*, O 2 , -SiR ® 3 , -OSIR ® 3, -C(O)SR·. -(C 1 -4 straight or branched alkylene)C(O)OR ® , or -SSR ® wherein each R ® is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently selected from C 1 -4 aliphatic, -CH 2 Ph, -O(CH 2 ) 0 - 1 Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents on a saturated carbon atom of R° include =O and =S.

Suitable divalent substituents on a saturated carbon atom of an “optionally substituted” group include the following: =O, =S, -NNR * 2 , =NNHC(O)R * , =NNHC(O)OR', =NNHS(O) 2 R * , -NR * , =NOR , -O(C(R * 2)) 2 -30, or -S(C(R * 2)) 2 -3S-, wherein each independent occurrence of R * is selected from hydrogen, C 1 -6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently seiected from nitrogen, oxygen, or sulfur. Suitable divalent substituents that are bound to vicinal substitutable carbons of an “optionally substituted" group include: -O(CR * 2) 2 -3O-, wherein each independent occurrence of R * is selected from hydrogen, C 1 -e aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.

Suitable substituents on the aliphatic group of R * include halogen, -R ® , -(haioR*), -OH, -OR ® , -O(haloR ® ), -CN, -C(O)OH, -C(O)OR ® , -NH 2 , -NHR ® , -NR , or -NO 2 , wherein each R ® is unsubstituted or where preceded by “halo" is substituted only with one or more halogens, and is independently C 1 -4 aliphatic, -CH 2 Ph, -O(CH 2 )o-:Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.

Suitable substituents on a substitutable nitrogen of an “optionally substituted" group include -FT, -NR 2 , -C<0)R , -C(O)OR , -C(O)C(O)R , -C<0)CH 2 C(O)R -S(O) 2 R , -S(O) 2 NR 2 , -C(S)NR , -C(NH)NR 2, or -N(R )S(O) 2 R ; wherein each R is independently hydrogen, C 1 -s aliphatic which may be substituted as defined below, unsubstituted -OPh, or an unsubstituted 3-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R , taken together with their intervening atom(s) form an unsubstituted 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyciie ring having 0-4 heteroatoms independently seiected from nitrogen, oxygen, or sulfur.

Suitable substituents on an aliphatic group of R are independently halogen, -R ® , -(haioR ® ), -OH, -OR ® , -O(haloR « ), -CN, -C(O)OH, -C(O)OR ® , -NH 2 , -NHR ® , -NR ® 2 , or -NO 2 , wherein each R* is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C 1 -4 aliphatic, -CH 2 Ph, -O(CH 2 ) 0 - 1 Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents on a saturated carbon atom of R include -O and -S

The term “acetyl,” as used herein, refers to the group -C(O)CH 3 .

The term “alkoxy,” as used herein, refers to a -O-C 1 -C 20 alkyl group, wherein the alkoxy group is attached to the remainder of the compound through an oxygen atom.

The term “alkyl," as used herein, refers to a saturated, straight or branched monovalent hydrocarbon group containing from 1 to 20 (e.g., from 1 to 10 or from 1 to 6) carbons. In some embodiments, an alkyl group is unbranched (i.e., is linear); In some embodiments, an alkyl group is branched. Alkyl groups are exemplified by, but not limited to, methyl, ethyl, n- and /so-propyl, n~, sec-, iso- and tert-butyl, and neopentyl.

The term “alkylene,” as used heroin, represents a saturated divalent hydrocarbon group derived from a straight or branched chain saturated hydrocarbon by the removal of two hydrogen atoms, and is exemplified by methylene, ethylene, isopropylene, and the like. The term “Cx-C y alkylene” represents alkvlene groups having between x and y carbons. Exemplary values for x are 1 , 2, 3, 4, 5, and 6, and exemplary values for y are 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, or 20 (e.g., C 1 -C 6 , CT-C 1 o, C 2 -C 20 , C 2 -C 6 , C 2 -C 10 , or C 2 -C 20 alkylene). In some embodiments, the alkylene can be further substituted with 1 , 2, 3, or 4 substituent groups as defined herein.

The term “alkenyl," as used herein, represents monovalent straight or branched chain groups of, unless otherwise specified, from 2 to 20 carbons (e.g., from 2 to 6 or from 2 to 10 carbons) containing one or more carbon-carbon double bonds and is exemplified by ethenyl, 1-propenyl, 2-propenyl, 2-methyl-1-propenyl, 1-butenyl, and 2-butenyl. Alkenyls Include both cis and trans isomers. The term “alkenylene,” as used herein, represents a divalent straight or branched chain groups of, unless otherwise specified, from 2 to 20 carbons (e.g., from 2 fo 6 or from 2 to 10 carbons) containing one or more carbon-carbon double bonds.

The term “alkynyl,” as used herein, represents monovalent straight or branched chain groups from 2 to 20 carbon atoms (e.g., from 2 to 4, from 2 to 6, or from 2 to 10 carbons) containing a carbon-carbon triple bond and is exemplified by ethynyl, and 1-propynyl.

The term “alkynyl sulfone,” as used herein, represents a group comprising the structure , wherein R is any chemically feasible substituent described herein.

The term “amino,” as used herein, represents -N(R ) 2 , e.g., -NH 2 and -N(CH 3 ) 2 .

The term “aminoalkyl,” as used herein, represents an alkyl moiety substituted on one or more carbon atoms with one or more amino moieties.

The term “amino acid,” as described herein, refers to a molecule having a side chain, an amino group, and an acid group (e.g., -CO 2 H or -SO 3 H), wherein the amino acid is attached to the parent molecular group by the side chain, amino group, or acid group (e.g., the side chain). As used herein, the term “amino acid” in its broadest sense, refers to any compound or substance that can be Incorporated into a polypeptide chain, e.g., through formation of one or more peptide bonds. In some embodiments, an amino acid has the general structure H 2 N-C(H)(R)-COOH In some embodiments, an amino acid is a naturally-occurring amino acid. In some embodiments, an amino acid is a synthetic amino acid; in some embodiments, an amino add is a D-amino acid; in some embodiments, an amino acid is an L-amino acid. “Standard amino acid” refers to any of the twenty standard L-amino acids commonly found in naturally occurring peptides. Exemplary amino acids include alanine, arginine, asparagine, aspartic acid, cysteine, glutarnic acid, glutamine, glycine, histidine, optionally substituted hydroxylnorvaline, isoleucine, leucine, lysine, methionine, norvaline, ornithine, phenylalanine, proline, pyrroiysine, selenocysteine, serine, taurine, threonine, tryptophan, tyrosine, and valine.

The term “aryl,” as used herein, represents a monovalent monocyclic, bicyclie, or muitlcyclic ring system formed by carbon atoms, wherein the ring attached to the pendant group is aromatic. Examples of aryl groups are phenyl, naphthyl, phenanthrenyl, and anthracenyl. An aryl ring can be attached to its pendant group at any heteroatom or carbon ring atom that results in a stable structure and any of the ring atoms can be optionally substituted unless otherwise specified.

The term “C 0 ,” as used herein, represents a bond. For example, part of the term -N(C(O)-(C 0 -C 5 alkylene-H)- includes -N(C(O)-(Go alkylene-H)-, which is also represented by -N(C(O)-H)-.

The terms “carbocyclic” and “carbocyclyl,” as used herein, refer to a monovalent, optionally substituted C 3 -C 1 2 monocyclic, bicyelic, or tricyclic ring structure, which may be bridged, fused or spirocyclic, in which ail the rings are formed by carbon atoms and at least one ring is non-aromatic. Carbocyclic structures inciude cycloalkyl, cycloalkenyl, and cycloalkynyl groups. Examples of carbocyclyl groups are cyclohexyl, cyclohexenyl, cyclooctynyl, 1 ,2-dihydronaphthyl, 1 ,2,3,4-tetrahydronaphthyl, fluorenyl, indenyl, indanyl, decalinyl, and the like. A carbocyclic ring can be attached to its pendant group at any ring atom that results in a stable structure and any of the ring atoms can be optionally substituted unless otherwise specified.

The term “carbonyl,” as used herein, represents a C(O) group, which can aiso be represented as

C=O.

The term “carboxyl," as used herein, means -CO 2 H, (C=O)(OH), COOH, or C(O)0H or the unprotonated counterparts.

The term “cyano,” as used herein, represents a -CM group.

The term “cycloalkyl,” as used herein, represents a monovalent saturated cyclic hydrocarbon group, which may bo bridged, fused or spirocyclic having from three to eight ring carbons, unless otherwise specified, and is exemplified by cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cycloheptyl.

The term “cycloalkenyl,” as used herein, represents a monovalent, non-aromatic, saturated cyclic hydrocarbon group, which may be bridged, fused or spirocyclic having from three to eight ring carbons, unless otherwise specified, and containing one or more carbon-carbon double bonds.

The term “diastereomer,” as used herein, means stereoisomers that are not mirror images of one another and are non-superimposabie on one another.

The term "enantiomer,” as used herein, means each individual optically active form of a compound of the invention, having an optical purity or enantiomeric excess (as determined by methods standard in the art) of at least 80% (l.e , at least 90% of one enantiomer and at most 10% of the other enantiomer), preferably at least 90% and more preferably at least 98%.

The term "guanidinyl,” refers to a group having the structure: , wherein each R is, independently, any any chemically feasible substituent described herein.

The term “guanidinoalkyl alkyl,” as used herein, represents an alkyl moiety substituted on one or more carbon atoms with one or more guanidinyl moieties.

The term “haloacetyl,” as used herein, refers to an acetyl group wherein at least one of the hydrogens has been replaced by a halogen.

The term “haioalkyl,” as used herein, represents an alkyl moiety substituted on one or more carbon atoms with one or more of the same of different halogen moieties.

The term "halogen,” as used herein, represents a halogen selected from bromine, chlorine, iodine, or fluorine.

The term "heteroalkyl," as used herein, refers to an "alkyl” group, as defined herein, in which at least one carbon atom has been replaced with a heteroatom (e.g., an O, N, or S atom). The heteroatom may appear in the middle or at the end of the radical.

The term "heteroaryl,” as used herein, represents a monovalent, monocyclic or polycyclic ring structure that contains at least one fully aromatic ring: l.e., they contain 4/7+2 pi electrons within the monocyclic or polycyclic ring system and contains at least one ring heteroatom selected from N, O, or S in that aromatic ring. Exemplary unsubstituted heteroaryl groups are of 1 to 12 (e.g., 1 to 11 , 1 to 10, 1 to 9, 2 to 12, 2 to 11 , 2 to 10, or 2 to 9) carbons. The term “heteroaryl" includes bicyciie, tricyclic, and tetracyclic groups in which any of the above heteroaromatic rings is fused to one or more, aryl or carbocyclic rings, e.g., a phenyl ring, or a cyclohexane ring. Examples of heteroaryl groups include, but are not limited to, pyridyi, pyrazolyl, benzooxazoiyl, benzoimidazoiyl, benzothiazolyl, imidazolyi, thiazoiyi, quinolinyl, tetrahydroquinolinyl, and 4-azaindolyl. A heteroaryl ring can be attached to its pendant group at any ring atom that results in a stable structure and any of the ring atoms can be optionally substituted unless otherwise specified. In some embodiment, the heteroaryl is substituted with 1 , 2, 3, or 4 substituents groups.

The term “heterocycloalkyl,” as used herein, represents a monovalent monocyclic, bieyciic or polycyclic ring system, which may be bridged, fused or spirocyclic, wherein at least one ring is non- aromatic and wherein the non-aromatic ring contains one, two, three, or four heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur. The 5-membered ring has zero to two double bonds, and the 6- and 7-membered rings have zero to three double bonds. Exemplary unsubstituted heterocycloalkyl groups are of 1 to 12 (e.g., 1 to 11 , 1 to 10, 1 to 9, 2 to 12, 2 to 11 , 2 to 10, or 2 to 9) carbons. The term “heterocycloalkyl” also represents a heterocyclic compound having a bridged multicycile structure In which one or more carbons or heteroatoms bridges two non-adjacent members of a monocyclic ring, e.g., a quinuciidinyl group. The term “heterocycloalkyl” includes bicyclic, tricyclic, and tetracyclic groups in which any of the above heterocyclic rings is fused to one or more aromatic, carbocyclic, heteroaromatic, or heterocyclic rings, e.g., an aryl ring, a cyclohexane ring, a cyclohexene ring, a cyclopentane ring, a cyclopentene ring, a pyridine ring, or a pyrrolidine ring.

Examples of heterocycloalkyl groups are pyrroiidinyl, piperidinyl, 1 ,2,3,4-fetrahydroquinoiinyl, decahydroquinolinyl, dihydropyrrolopyridine, and decahydronapthyridinyl. A heterocycloalkyl ring can be attached to its pendant group at any ring atom that results In a stable structure and any of the ring atoms can be optionally substituted unless otherwise specified.

The term "hydroxy,” as used herein, represents a -OH group.

The term "hydroxyalkyl,” as used herein, represents an alkyl moiety substituted on one or more carbon atoms with one or more -OH moieties.

The term “isomer,” as used herein, means any tautomer, stereoisomer, atropiosmer, enantiomer, or diasteroomer of any compound of the Invention. It is recognized that the compounds of the invention can have one or more chiral centers or double bonds and, therefore, exist as stereoisomers, such as double-bond isomers (l.e., geometric E/Z isomers) or diastereomers (e.g., enantiomers (i.e , (+) or (-)) or cis/trans isomers). According to the invention, the chemical structures depicted herein, and therefore the compounds of the Invention, encompass ali the corresponding stereoisomers, that is, both the stereomerically pure form (e.g., geometrically pure, enantiomerically pure, or diastereomerically pure) and enantiomeric and stereoisomeric mixtures, e.g., racemates. Enantiomeric and stereoisomeric mixtures of compounds of the invention can typically be resolved Into their component enantiomers or stereoisomers by well-known methods, such as chiral-phase gas chromatography, chiral-phase high performance liquid chromatography, crystallizing the compound as a chiral salt complex, or crystallizing the compound in a chiral solvent. Enantiomers and stereoisomers can also bo obtained from stereomerically or enantiomerically pure intermediates, reagents, and catalysts by well-known asymmetric synthetic methods.

As used herein, the term "linker” refers to a divalent organic moiety connecting moiety B to moiety W In a compound of Formula I, such that the resulting compound is capable of achieving an IC5G of 2 uM or less in the Ras-RAF disruption assay protocol provided In the Examples below, and provided here:

The purpose of this biochemical assay is to measure the ability of test compounds to facilitate ternary complex formation between a nucleotide-loaded Ras isoform and cyclophilin A; the resulting ternary complex disrupts binding to a BRAF RBD construct, inhibiting Ras signaling through a RAF effector.

In assay buffer containing 25 mM HEPES pH 7.3, 0.002% Tween20, 0.1% BSA, 100 mM NaCI and 5 M MgCl 2 , tagless Cyclophilin A, His6-K-Ras-GMPPNP (or other Ras variant), and GST-BRAF RBD are combined in a 384-well assay plate at final concentrations of 25 μM, 12.5 nM and 50 nM, respectively. Compound is present in plate wells as a 10-point 3-fold dilution series starting at a final concentration of 30 μM. After Incubation at 25°C for 3 hours, a mixture of Anti- His Eu-W1024 and anti-GST allophycocyanin is then added to assay sample wells at final concentrations of 10 nM and 50 nM, respectively, and the reaction incubated for an additional 1 .5 hours. TR-FRET signal is read on a micropiate reader (Ex 320 nm, Em 665/815 nm). Compounds that facilitate disruption of a Ras: RAF complex are identified as those eliciting a decrease in the TR-FRET ratio relative to DMSO control wells.

In some embodiments, the linker comprises 20 or fewer linear atoms. In some embodiments, the linker comprises 15 or fewer linear atoms. In some embodiments, the linker comprises 10 or fewer linear atoms. In some embodiments, the linker has a molecular weight of under 500 g/mol. In some embodiments, the linker has a molecular weight of under 400 g/mol. In some embodiments, the linker has a molecular weight of under 300 g/mol. in some embodiments, the linker has a molecular weight of under 200 g/mol. In some embodiments, the linker has a molecular weight of under 100 g/mol. In some embodiments, the linker has a molecular weight of under 50 g/mol.

As used herein, a “monovalent organic moiety” is less than 500 kDa. In some embodiments, a "monovalent organic moiety” is less than 400 kDa. In some embodiments, a “monovalent organic moiety” is less than 300 kDa. in some embodiments, a “monovalent organic moiety” is less than 200 kDa. In some embodiments, a “monovalent organic moiety” is less than 100 kDa. in some embodiments, a “monovalent organic moiety" is less than 50 kDa. In some embodiments, a “monovalent organic moiety” is less than 25 kDa. In some embodiments, a “monovalent organic moiety” is less than 20 kDa. In some embodiments, a “monovalent organic moiety” is less than 15 kDa. in some embodiments, a “monovalent organic moiety” is less than 10 kDa. In some embodiments, a “monovalent organic moiety” is less than 1 kDa. in some embodiments, a “monovalent organic moiety" is loss than 500 g/mol. In some embodiments, a “monovalent organic moiety” ranges between 500 g/mol and 500 kDa.

The term “stereoisomer,” as used herein, refers to all possible different isomeric as well as conformational forms which a compound may possess (e.g., a compound of any formula described herein), in particular ail possible stereochemically and conformationally isomeric forms, all diastereomers, enantiomers or conformers of the basic molecular structure, including atropisomers. Some compounds of the present invention may exist in different tautomeric forms, all of the latter being included within the scope of the present invention. The term “sulfonyl,” as used herein, represents an -S(O) 2 - group.

The term “thiocarbonyl,” as used herein, refers to a -C(S)- group.

The term “vinyl ketone," as used herein, refers to a group comprising a carbonyl group directly connected to a carbon-carbon double bond.

The term “vinyl sulfone,” as used herein, refers to a group comprising a sulfonyl group directed connected to a carbon-carbon double bond.

The term “ynone,” as used herein, refers to a group comprising the structure wherein R is any any chemically feasible substituent described herein.

Those of ordinary skill in the art, reading the present disclosure, will appreciate that certain compounds described herein may be provided or utilized in any of a variety of forms such as, for example, salt forms, protected forms, pro-drug forms, ester forms, isomeric forms (e.g., optical or structural isomers), isotopic forms, etc. in some embodiments, reference to a particular compound may relate to a specific form of that compound. In some embodiments, reference to a particular compound may relate to that compound in any form. In some embodiments, for example, a preparation of a single stereoisomer of a compound may be considered to be a different form of the compound than a racemic mixture of the compound; a particular salt of a compound may be considered to be a different form from another salt form of the compound; a preparation containing one conformational isomer ((Z) or (E)) of a double bond may be considered to be a different form from one containing ihe other conformational isomer ((E) or (Z)) of the double bond; a preparation in which one or more atoms is a different isotope than is present in a reference preparation may be considered to be a different form.

Detailed Description

Compounds

Provided herein are Ras Inhibitors. The approach described herein entails formation of a high affinity three-component complex between a synthetic ligand and two intracellular proteins which do not interact under normal physiological conditions: the target protein of interest (e.g., Ras), and a widely expressed cytosolic chaperone (presenter protein) in the cell (e.g., cyclophiiin A). More specifically, In some embodiments, the inhibitors of Ras described herein induce a new binding pocket in Ras by driving formation of a high affinity tn-complex between the Ras protein and the widely expressed cytosolic chaperone, cyclophilin A (CYRA). Without being bound by theory, the inventors believe that one way the inhibitory effect on Ras is effected by compounds of the invention and the complexes they form is by steric occlusion of the interaction site between Ras and downstream effector molecules, such as RAF, which are required for propagating the oncogenic signal.

Without being bound by theory, the inventors postulate that non-covaient interactions of a compound of the present invention with Ras and the chaperone protein (e.g., cyclophilin A) may contribute to the inhibition of Ras activity. For example, van dor Waals, hydrophobic, hydrophilic and hydrogen bond interactions, and combinations thereof, may contribute to the ability of the compounds of the present invention to form complexes and aci as Ras inhibitors. Accordingly, a variety of Ras proteins may be inhibited by compounds of the present invention (e.g., K-Ras, N-Ras, H-Ras, and mutants thereof at positions 12, 13 and 61 , such as G12C, G12D, G12V, G12S, G13C, G13D, and G61 L, and others described herein).

Accordingly, provided herein is a compound, or pharmaceutically acceptable salt thereof, having the structure of Formula 00:

Formula 00 wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 10- membered heteroarylene;

G is optionally substituted C 1 -C 4 alkylene, optionally substituted C 1 -C 4 alkenylene, optionally substituted C 1 -C 4 heteroalkylene, -C(O)O-CH(R 6 )-- where C is bound to -C(R 7 R 8 )-, -C(O)NH-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C 1 -C 4 heteroalkylene, or 3 to 8-membered heteroarylene: swlp (Switch l/P-loop) refers to an organic moiety that non-covaiently binds to both the Switch I binding pocket and residues 12 or 13 of the P-loop of a Ras protein (see, e.g., Johnson et al., 292:12981- 12993 (2017), incorporated herein by reference):

X 1 is optionally substituted C 1 -C 2 alkylene, NR, O, or S(O) n ;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R', C(O)OR', C(O)N(R’) 2 , S(O)R', S(O) 2 R’, or S(O) 2 N(R') 2 ; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y' ! , and Y 7 are, independently, or N C;

Y 5 is CH, CH 2 , or N;

Y 6 is C(O). CH, CH 2 , or N; R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl, or R 1 and R 2 combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl; R 2 is absent, hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R B is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C ^ N(OH), C ^ N(O-C 1 -C 3 alkyl), G=O, G=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7a and R 83 are, independently, hydrogen, halo, optionally substituted C 1 -C 3 alkyl, or combine with the carbon to which they are attached to form a carbonyl;

R 7' is hydrogen, halogen, or optionally substituted C1-C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-mcmbered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 10 is hydrogen, halo, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl;

R 10a is hydrogen or halo: and

R 16 is hydrogen or C 1 -C 3 alkyl (e.g., methyl). In some embodiments, the resulting compound is capable of achieving an IC50 of 2 uM or less (e.g., 1 .5 uM, 1 uM, 500 nM, or 100 nM or less) in the Ras- RAF disruption assay protocol described herein.

Accordingly, provided herein is a compound, or pharmaceutically acceptable salt thereof, having the structure of Formula I:

Formula I wherein the doited lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 10- membered heteroarylene;

B is absent, -CH(R 9 )-, or >C=CR S R 9 where the carbon is bound to the carbonyl carbon of - N(R 11 )C(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C 1 -C 4 alkylene, optionally substituted C 1 -C 4 alkenylene, optionally substituted C 1 -C 4 heteroalkylene, -C(O)O-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(Q)NH-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C 1 -C 4 heteroalkylene, or 3 to 8-membered heteroarylene;

L is absent or a linker;

W is hydrogen, cyano, S/OkR', optionally substituted amino, optionally substituted amldo, optionally substituted C 1 -C 4 alkoxy, optionally substituted C 1 -C 4 hydroxyalkyl, optionally substituted C 1 -C 4 aminoalkyl, optionally substituted C 1 -C 4 haloalkyl, optionally substituted C 1 -C 4 alkyl, optionally substituted C 1 -C 4 guanidinoalkyl, C0-C 4 alkyl optionally substituted 3 to 11-membered heterocycloalkyl, optionally substituted 3 to 8-membered cycloalkyl. or optionally substituted 3 to 8-membered heteroaryl;

X 1 is optionally substituted C 1 -C 2 alkylene, NR, G, or S(O) n ;

X 2 is O or NH:

X 3 is N or CH; n is 0, 1 , or 2; R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R’, C(O)OR’, C(O)N(R’) 2 , S(O)R’, SiQ^R’, or S(O) 2 N(R’) 2 ; each R' is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, or N C; R 1 is cyano, optionally substituted C 1 -C 6 alkyl , optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl, or R 1 and R 2 combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl; R 2 is absent, hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C8 alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or eyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R B is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), ON(O-C 1 -C 3 alkyl), G=O, G=S, C=NH, optionally substituted 3 to 6-membered eyeioalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 73 and R 83 are, independently, hydrogen, halo, optionally substituted C 1 -C 3 alkyl, or combine with the carbon to which they are attached to form a carbonyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl. optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is hydrogen, F, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl, or

R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl: R 9' is hydrogen or optionally substituted C 1 -C 6 alkyl; R 10 is hydrogen, halo, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl; R 10a is hydrogen or halo; R 11 is hydrogen or C 1 -C 3 alkyl; R 16 is hydrogen or C 1 -C 3 alkyl (e.g., methyl).

In some embodiments, the disclosure features a compound, or pharmaceutically acceptable salt thereof, of structural Formula la: wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 10- membered heteroarylene;

B is -CH(R 9 )- or >C=CR S R 9 where the carbon is bound to the carbonyl carbon of -N(R 11 )C(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C 1 -C 4 alkylene. optionally substituted C 1 -C 4 alkenylene, optionally substituted C 1 -C 4 heteroalkylene, -C(O)O-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(O)NH-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C 1 -C 4 heteroalkylene, or 3 to 8-membered heteroarylene;

L is absent or a linker;

W is hydrogen, optionally substituted amino, optionally substituted C 1 -C 4 alkoxy, optionally substituted C 1 -C 4 hydroxyalkyl, optionally substituted C 1 -C 4 aminoalkyl, optionally substituted C 1 -C 4 haloalkyl, optionally substituted C 1 -C 4 alkyl, optionally substituted C 1 -C 4 guanidinoalkyl, C0-C 4 alkyl optionally substituted 3 to 11-membered heterocycloalkyl, optionally substituted 3 to 8-membered cycloalkyl, or optionally substituted 3 to 8-membered heteroaryl;

X 1 is optionally substituted C 1 -C 2 alkylene, NR, G, or S(O) n ;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2; R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R', C(O)OR’, C(O)N(R’) 2 , S(O)R’, S(O) 2 R’, or S(O) 2 N(R') 2 ; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CM, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 is CH, CH 2 , or N;

Y 6 is C(O), CH, CH 2 , or N; R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl, or R 1 and R 2 combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 2 is absent, hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl;

R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl ;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C 1 -C 3 alkyl), C=O, C=S, C==NH, optionally substituted 3 to 6-membered eydoalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7a and R 8a are, independently, hydrogen, halo, optionally substituted C 1 -C 3 alkyl, or combine with the carbon to which they are attached to form a carbonyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl; R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl, or R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl; R 8' is hydrogen or optionally substituted C 1 -C 6 alkyl; R 10 is hydrogen, halo, hydroxy, C 1 -C 3 alkoxy, or C 3 alkyl; R 10a is hydrogen or halo; and R 11 is hydrogen or C 1 -C 3 alkyl.

In some embodiments, the disclosure features a compound, or pharmaceutically acceptable salt thereof, of structural Formula lb: wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CHslCiGHCH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -N(R 1 , )C(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C 1 -C 4 alkylene, optionally substituted C 1 -C 4 alkenylene, optionally substituted C 1 -C 4 heteroalkylene, -C(O)O-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(O)NH-€H(R 6 )- where C is bound to -C(R 7 R 8 )-, optionally substituted C 1 -C 4 heteroalkylene, or 3 to 8-membered heteroarylene;

L is absent or a linker;

W is hydrogen, optionally substituted amino, optionally substituted C 1 -C 4 alkoxy, optionally substituted C 1 -C 4 hydroxyalkyl, optionally substituted C 1 -C 4 aminoalkyl, optionally substituted C 1 -C 4 haloalkyl, optionally substituted C 1 -C 4 alkyl, optionally substituted C 1 -C 4 guanidinoalkyl, C0-C 4 alkyl optionally substituted 3 to 11-membered heterocycloalkyl, optionally substituted 3 to 8-membered cycloalkyl, or optionally substituted 3 to 8-membered heteroaryl;

X 1 is optionally substituted C 1 -C 2 alkylene, NR, O, or S(O) n ;

X 2 is O or NH; X 3 is N or CH; n is 0, 1 , or 2; R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R’, C(O)OR', C(O)N(R’) , S(O)R', S(O) 2 R’, or S(O) 2 N(R') 2 ; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 and Y 6 are, independently, CH or N; R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl; R 2 is hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or

R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobuiyi;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C -C 3 alkyl), C=O, C=S, C==NH, optionally substituted 3 to 6-membered eycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl. optionally substituted 3 to 8-membered eycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered eycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered eycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 10 is hydrogen, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl; and

R 11 is hydrogen or C 1 -C 3 alkyl. In some embodiments of compounds of the present invention, G is optionally substituted C 1 -C 4 heteroalkylene.

In some embodiments, a compound of the present Invention has the structure of Formula lc, or a pharmaceutically acceptable salt thereof:

Formula lc wherein the doted lines represent 2ero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R i0 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -N(R 11 )C(O)-. optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is hydrogen, optionally substituted amino, optionally substituted C 1 -C 4 alkoxy. optionally substituted C 1 -C 4 hydroxyalkyl, optionally substituted C 1 -C 4 aminoalkyl, optionally substituted C 1 -C 4 haloalkyl, optionally substituted C 1 -C 4 alkyl, optionally substituted C 1 -C 4 guanidinoalkyl. C0-C 4 alkyl optionally substituted 3 to 11-membered heterocycloalkyl, optionally substituted 3 to 8-membered cycloalkyl, or optionally substituted 3 to 8-membered heteroaryl;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(Q)R\ C(O)OR’, C(O)N(R') 2 , S(O)R’, S(O) 2 R’, or S(O) 2 N(R’) 2 ; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, independently, C or N;

Y 5 and Y 6 are, independently, CH or N;

R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl or optionally substituted 5 to 10-membered heteroaryl; R 2 is hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membored heterocycloalkyl optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl; R 4 is absent, hydrogen, haiogen, cyano, or methyl optionally substituted with 1 to 3 halogens; R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl; R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl; R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C 1 -C 3 alkyl), C=O, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -Ge alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is optionally substituted G·,- C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 10 is hydrogen, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl; and

R 11 is hydrogen or C 1 -C 3 alkyl.

In some embodiments of compounds of the present Invention, X 2 is NH In some embodiments, X 3 is CH.

In some embodiments of compounds of the present Invention, R 11 is hydrogen. In some embodiments, R 11 is C 1 -C 3 alkyl. In some embodiments, R 11 is methyl.

In some embodiments, a compound of the present Invention has the structure of Formula id, or a pharmaceutically acceptable salt thereof:

Formula Id wherein the doted lines represent 2ero, one, two, three, or four non-adjaeert double bonds;

A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- mernbered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is hydrogen, optionally substituted amino, optionally substituted C 1 -C 4 alkoxy, optionally substituted C 1 -C 4 hydroxyalkyl, optionally substituted C 1 -C 4 aminoalkyl, optionally substituted C 1 -C 4 haloalkyl, optionally substituted C 1 -C 4 alkyl, optionally substituted C 1 -C 4 guanidinoalkyl, G0-C 4 alkyl optionally substituted 3 to 11-membered heterocycloalkyl, optionally substituted 3 to 8-membered cycloalkyl, or optionally substituted 3 to 8-membered heteroaryl; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R’, C(O)OR’, C(O)N(R') 2 , S(O)R’, S(O) 2 R’, or S(O) 2 N{R’i2; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl ;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, Independently, C or N;

Y 5 and Y 6 are, independently, CH or N;

R 1 is cyano, optionally substituted Gi-C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-mombered heterocycloalkyl, optionally substituted 6 to 10-mombered aryl, or optionally substituted 5 to 10-mombered heteroaryl;

R 2 is hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted 3 to 6-mombered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl; R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted G1-C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C 1 -C 3 alkyl), C=G, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -G6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 6’ combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; and

R 10 is hydrogen, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl.

In some embodiments of compounds of the present Invention, X 1 is optionally substituted C 1 -C 2 alkylene. in some embodiments, X 1 is methylene. In some embodiments, X’ is methylene substituted with a C 1 -C 6 alkyl group or a halogen. In some embodiments, X 1 is -CH(Br)- In some embodiments, X 1 is -CHiCHs)-.

In some embodiments of compounds of the present Invention, R 3 is absent.

In some embodiments of compounds of fhe present Invention, R 4 is hydrogen.

In some embodiments of compounds of fhe present Invention, R 6 is hydrogen. In some embodiments, R 5 is G1-C 4 alkyl optionally substituted with halogen. In some embodiments, R 5 is methyl.

In some embodiments of compounds of fhe present Invention, Y 4 is C. in some embodiments, Y 5 is CH. in some embodiments, Y 6 is CH. in some embodiments, Y 1 is C. In some embodiments, Y 2 is G in some embodiments, Y 3 is N. In some embodiments, Y 7 is C.

In some embodiments, a compound of the present Invention has the structure of Formula ie, or a pharmaceutically acceptable salt thereof:

Formula le wherein A is -N(H or CH 3 )C(O)-(CI- )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is hydrogen, optionally substituted amino, optionally substituted C 1 -C 4 alkoxy, optionally substituted C 1 -C 4 hydroxyalkyl, optionally substituted C 1 -C 4 aminoalkyl, optionally substituted C 1 -C 4 haloalkyl, optionally substituted C 1 -C 4 alkyl, optionally substituted C 1 -C 4 guanidinoalkyl, C0-C 4 alkyl optionally substituted 3 to 11-membered heterocycloalkyl, optionally substituted 3 to 8-membered cycloalkyl, or optionally substituted 3 to 8-membered heteroaryl; R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl; R 2 is hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent, or R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl; R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R B is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-mombered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C 1 -C 3 alkyl), C=O, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; and R 10 is hydrogen, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl.

In some embodiments of compounds of the present Invention, R 6 is hydrogen.

In some embodiments of compounds of the present Invention, R 2 is hydrogen, cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 6- membered heterocycloalkyl. in some embodiments, R 2 is optionally substituted C 1 -C 6 alkyl, such as ethyl. In some embodiments, R 2 is tluoro Gi~C 6 alkyl, such as -CH 2 CH 2 F, -CH 2 CHF2, or -CH 2 CF3. in some embodiments of compounds of the present Invention, R 7 is optionally substituted C 1 -C 3 alkyl. In some embodiments, R 7 is C 1 -C 3 alkyl. in some embodiments of compounds of the present Invention, R 8 is optionally substituted C 1 -C 3 alkyl. In some embodiments, R 8 is C 1 -C 3 alkyl, such as methyl. in some embodiments, a compound of the present invention has the structure of Formula If, or a pharmaceutically acceptable salt thereof:

Formula lf wherein A optionally substituted 3 to 6-membered cycloalkylene. optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene; B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is hydrogen, optionally substituted amino, optionally substituted C 1 -C 4 alkoxy, optionally substituted C1-C 4 hydroxyalkyl, optionally substituted C 1 -C 4 aminoalkyl, optionally substituted C 1 -C 4 haloalkyl, optionally substituted C 1 -C 4 alkyl, optionally substituted C 1 -C 4 guanidinoalkyl, C0-C 4 alkyl optionally substituted 3 to 11-membered heterocycloalkyl, optionally substituted 3 to 8-membered cycloalkyl, or optionally substituted 3 to 8-membered heteroaryl; R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl;

R 2 is C 1 -C 6 alkyl or 3 to 6-membered cycloalkyl;

R 7 is C 1 -C 3 alkyl; R 8 is C 1 -C 3 alkyl; and

R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl.

In some embodiments of compounds of the present Invention, R 1 is 5 to 10-membered heteroaryl. in some embodiments, R 1 is optionally substituted 6-membered aryl or optionally substituted 6-membered heteroaryl. stereoisomer thereof. In some embodiments, stereoisomer thereof. In some embodiments,

In some embodiments, a compound of the present invention has the structure of Formula Ig, or a pharmaceutically acceptable salt thereof:

Formula ig wherein A is optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is hydrogen, optionally substituted amino, optionally substituted C 1 -C 4 alkoxy, optionally substituted C 1 -C 4 hydroxyalkyl, optionally substituted C 1 -C 4 aminoalkyl, optionally substituted C 1 -C 4 haloalkyl, optionally substituted C 1 -C 4 alkyl, optionally substituted C 1 -C 4 guanidinoalkyl, C0-C 4 alkyl optionally substituted 3 to 11-membered heterocycloalkyl, optionally substituted 3 to 8-membered cycloalkyl, or optionally substituted 3 to 8-membered heteroaryl;

R 2 is C 1 -C 6 alkyl or 3 to 6-membered cycloalkyl; R 7 is C -C alkyl; R 8 is C 1 -C 3 alkyl; R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; R 12 is optionally substituted C 1 -C 6 alkyl or optionally substituted C 1 -C 6 heteroalkyl; and R 17 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl.

In some embodiments of compounds of the present Invention, X e is N and X f is CH. In some embodiments, X e is CH and X f is N. In some embodiments, X e is CR 17 and X f is N.

In some embodiments of compounds of the present Invention, R i2 is optionally substituted C 1 -C 6

CH 3

Y x heteroalkyl. in some embodiments, R' 2 is V OMe OIVie \ / \ /0Me

In some embodiments, a compound of the present invention has the structure of Formula Ih, or a pharmaceutically acceptable salt thereof:

Formula Ih wherein A is optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is hydrogen, optionally substituted amino, optionally substituted C 1 -C 4 alkoxy, optionally substituted C 1 -C 4 hydroxyalkyl, optionally substituted C 1 -C 4 aminoalkyl, optionally substituted C 1 -C 4 haloalkyl, optionally substituted C 1 -C 4 alkyl, optionally substituted C 1 -C 4 guanldlnoalkyl, C0-C 4 alkyl optionally substituted 3 to 11-membered heterocycloalkyl, optionally substituted 3 to 8-membered cycloalkyl, or optionally substituted 3 to 8-membered heteroaryl; R 2 is C 1 -C 6 alkyl or 3 to 6-membered cycloalkyl; R 7 is C 1 -C 3 alkyl;

R 8 is C 1 -C 3 alkyl; R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

X e is CH, or CR 17 ; and R 17 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl.

In some embodiments, a compound of the present invention has the structure of Formula li, or a pharmaceutically acceptable salt thereof:

Formula li wherein A is optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is hydrogen, optionally substituted amino, optionally substituted C 1 -C 4 alkoxy. optionally substituted C 1 -C 4 hydroxyalkyl, optionally substituted C 1 -C 4 amlnoalkyl, optionally substituted C 1 -C 4 haloalkyl, optionally substituted C 1 -C 4 alkyl, optionally substituted C 1 -C 4 guanidinoalkyl. C0-C 4 alkyl optionally substituted 3 to 11-membered heterocycloalkyl, optionally substituted 3 to 8-membered cycloalkyl, or optionally substituted 3 to 8-membered heteroaryl;

R 2 is C 1 -C 6 alkyl or 3 to 6-membered cycloalkyl;

R 7 is C 1 -C 3 alkyl;

R B is C 1 -C 3 alkyl; and R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl.

In some embodiments of compounds of the present invention, A is optionally substituted 6- membered arylene. In some embodiments, A has the structure: wherein R 13 is hydrogen, hydroxy, amino, cyano, optionally substituted C 1 -C 6 alkyl, or optionally substituted C 1 -C 6 heteroalkyl. In some embodiments, R 13 is hydrogen. In some embodiments, R 13 is hydroxy. In some embodiments, A is an optionally substituted 5 to 10-membered heteroarylene. In some embodiments, A is: . In some embodiments, A is optionally substituted 5 to 6-membered heteroarylene. In some embodiments, A is: , , or

N-S . In some embodiments, A is S In some embodiments of compounds of the present Invention, B is -CHR 9 -. in some embodiments, R 9 is optionally substituted C 1 -C 6 alkyl or optionally substituted 3 to 6-membered cycloalkyl. In some embodiments, R 9 is ,: . , in some embodiments, R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6- membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl.

In some embodiments, B is optionally substituted 6-membered arylene. in some embodiments, B is 6-membered arylene. In some embodiments, B is: . in some embodiments B is absent.

In some embodiments of compounds of the present invention, R 7 is methyl.

In some embodiments of compounds of the present invention, R 8 is methyl.

In some embodiments of compounds of the present invention, R 16 is hydrogen.

In some embodiments of compounds of the present invention, the linker is the structure of

Formula ii: A 1 -(B’ )f-(C ' )g-(B 2 )h-{ D’ )-(B 3 )i-(C 2 )j-(B 4 )k—A 2

Formula II where A' is a bond between the linker and B; A 2 is a bond between W and the linker; B 1 , B 2 , B 3 , and B 4 each, independentiy, is selected from optionally substituted C 1 -C 2 alkylene, optionally substituted C 1 -C 3 heteroalkylene, O, S, and NR N ; R N is hydrogen, optionally substituted C 1 -C 4 alkyl, optionally substituted C 1 -C 3 cycloalkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 C 4 alkynyl , optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted C 1 -C 7 heteroalkyl; C 1 and G 2 are each, independentiy, selected from carbonyl, thiocarbonyl, sulphonyl, or phosphoryl; f, g, h, i, I, and k are each, independently, 0 or 1 ; and D 1 is optionally substituted C 1 -C 10 alkylene, optionally substituted C 2 -C 10 alkenylene, optionally substituted C 2 - C 10 alkynylene, optionally substituted 3 to 14-membered heterocycloalkylene, optionally substituted 5 to 10-membered heteroarylene, optionally substituted 3 to 8-membered cycloalkylene, optionally substituted 6 to 10-membered arylene, optionally substituted C 2 -C 10 polyethylene glycolene, or optionally substituted C 1 -C 10 heteroalkylene, or a chemical bond linking A ! -(B 1 )f-(C i ) g -(B 2 )h- to -(B 3 )i-(C 2 )j-(B 4 )k— A 2 . In some embodiments, the linker is acyclic. In some embodiments, the linker has the structure of Formula lla:

Formula lla wherein X 3 is absent or N; R 14 is absent, hydrogen or optionally substituted C 1 -C 6 alkyl or optionally substituted C 1 -C 3 cycloalkyl; and

L 2 is absent, -C(O)-, -SO 2 -, optionally substituted C 1 -C 4 alkylene or optionally substituted C 1 -C 4 heteroalkylene, wherein at least one of X a , R 14 , or L 2 is present. In some embodiments, the linker has the structure: embodiments, L is 0 | n some embodiments, linker is or comprises a cyclic group. In some embodiments, linker has the structure of Formula ilb:

Formula lIb wherein o is 0 or 1 ;

X b is C(O) or SO 2 ;

R 15 is hydrogen or optionally substituted C 1 -C 6 alkyl;

Gy is optionally substituted 3 to 8-membered cycloalkylene, optionally substituted 3 to 8- membered heterocycloalkylene, optionally substituted 6-10 membered arylene, or optionally substituted 5 to 10-membered heteroarylene; and

L 3 is absent, -0(O)-, -SO 2 -, optionally substituted C 1 -C 4 alkylene or optionally substituted C 1 -C 4 heteroalkylene. in some embodiments, linker has the structure:

In some embodiments of compounds of the present invention, W is hydrogen, optionally substituted amino, optionally substituted C 1 -C 4 alkoxy, optionally substituted C 1 -C 4 hydroxyalkyl, optionally substituted C 1 -C 4 amlnoalkyl, optionally substituted C 1 -C 4 haloalkyl, optionally substituted C 1 -C 4 alkyl, optionally substituted C 1 -C 4 guanidinoalkyl, C0-C 4 alkyl optionally substituted 3 to 8-membered heterocycloalkyl, optionally substituted 3 to 8-membered cycloalkyl, or 3 to 8-membered heteroaryl.

In some embodiments of compounds of the present Invention, W is hydrogen. In some embodiments, W is optionally substituted amino. In some embodiments, W is -NHCH 3 or -N(CH 3 ) 2 . In some embodiments, W is optionally substituted C 1 -C 4 alkoxy. in some embodiments, W is methoxy or iso-propoxy. in some embodiments, W is optionally substituted C 1 -C 4 alkyl. In some embodiments, W is methyl, ethyl, iso-propyl, tert-butyl, or benzyl. In some embodiments, W is optionally substituted amido. in some embodiments, W is . in some embodiments, W is optionally substituted amido. In some O embodiments, W is v%" . In some embodiments, W is optionally substituted C 1 -C 4 hydroxyalkyl. n some embodiments, . In some embodiments, W is optionally substituted C 1 -C 4 amlnoalkyl. In some embodiments, W is W CF 3 CHF 2 is optionally substituted C 1 -C 4 haloalkyl. In some embodiments, W is ¾ , , OG I n some embodiments, W is optionally substituted C 1 -C 4 guanidlnoalkyl. In some embodiments, some embodiments, W is C0-C 4 alkyl optionally substituted 3 to 11-membered heterocycloalkyl. in some

substituted 3 to 8-membered cycloalkyl. In some embodiments,

embodiments, W is optionally substituted 3 to 8-membered heteroaryl. In some embodiments, W is optionally substituted 6- to 10-membered aryl (e g. s phenyl, 4-hydroxy-phenyl, or 2,4-methoxy-phenyl).

In some embodiments, a compound of the present invention is selected from Table 1 , or a pharmaceutically acceptable salt or stereoisomer thereof. In some embodiments, a compound of the present Invention is selected from Table 1 , or a pharmaceutically acceptable sait or atropisomer thereof.

Table 1 : Certain Compounds of the Present invention

Note that some compounds are shown with bonds as flat or wedged. In some instances, the relative stereochemistry of stereoisomers has been determined; in some instances, the absolute stereochemistry has been determined. In some Instances, a single Example number corresponds to a mixture of stereoisomers. All stereoisomers of the compounds of the foregoing table are contemplated by the present invention. In particular embodiments, an atropisomer of a compound of the foregoing table is contemplated. Any compound shown In brackets indicates that the compound is a disastereomer, and the absolute stereochemistry of such diastereomer may not be known.

In some embodiments, a compound of Table 2 is provided, or a pharmaceutically acceptable salt thereof. In some embodiments, a compound of the present invention is selected from Table 2, or a pharmaceutically acceptable salt or atropisomer thereof.

Table 2; Certain Compounds of the Present invention

Note that some compounds are shown with bonds as fiat or wedged. In some instances, the relative stereochemistry of stereoisomers has been determined; in some instances, the absolute stereochemistry has been determined. All stereoisomers of the compounds of the foregoing table are contemplated by the present invention. In particular embodiments, an atropisomer of a compound of the foregoing table is contemplated . in some embodiments, a compound of the present invention is or acts as a prodrug, such as with respect to administration to a cell or to a subject in need thereof. Also provided are pharmaceutical compositions comprising a compound of the present invention, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.

Further provided is a method of treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof. The cancer may, for example, be pancreatic cancer, colorectal cancer, non-small cell lung cancer, acute myeloid leukemia, multiple myeloma, thyroid gland adenocarcinoma, a myelodyspiastic syndrome, or squamous cell lung carcinoma. In some embodiments, the cancer comprises a Ras mutation, such as K-Ras G12C, K-Ras G12D, K-Ras G12V, K-Ras G12S, K-Ras G13G, K-Ras G13D, or K-Ras Q61 L Other Ras mutations are described herein.

Further provided is a method of treating a Ras protein-related disorder in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof.

Further provided is a method of inhibiting a Ras protein in a cell, the method comprising contacting the cell with an effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof. For example, the Ras protein is K-Ras G12G, K-Ras G12D, K-Ras G12V, K-Ras G12S, K-Ras G13C, K-Ras G13D, or K-Ras Q61 L. Other Ras proteins are described herein. The cell may be a cancer cell, such as a pancreatic cancer cell, a colorectal cancer cell, a non-small cell lung cancer cell, an acute myeloid leukemia cell, a multiple myeloma cell, a thyroid gland adenocarcinoma cell, a myelodyspiastic syndrome cell, or a squamous cell lung carcinoma cell. Other cancer types are described herein. The cell may be in vivo or in vitro.

With respect to compounds of the present invention, one stereoisomer may exhibit better inhibition than another stereoisomer. For example, one atropisomer may exhibit inhibition, whereas the other atropisomer may exhibit little or no inhibition.

Methods of Synthesis

The compounds described herein may be made from commercially available starting materials or synthesized using known organic, inorganic, or enzymatic processes.

The compounds of the present invention can be prepared in a number of ways well known to those skilled in the art of organic synthesis. By way of example, compounds of the present invention can be synthesized using the methods described in the Schemes below, together with synthetic methods known in the art of synthetic organic chemistry, or variations thereon as appreciated by those skilled in the art. These methods include but are not limited to those methods described In the Schemes below.

Compounds of Table 1 herein were prepared using methods disclosed herein or were prepared using methods disclosed herein combined with the knowledge of one of skill in the art. Compounds of Table 2 may be prepared using methods disclosed herein or may be prepared using methods disclosed herein combined with the knowledge of one of skill in the art. Scheme 1. General synthesis of macrocyclic esters

A general synthesis of macrocyclic esters is outlined in Scheme 1 . An appropriately substituted Aryl Indole intermediate (1) can be prepared In three steps starting from protected 3-(5-broma-2-iodo-1H- indol-3-yl)-2,2-dimethylpropan-1-oi and appropriately substituted boronic acid, Including Palladium mediated coupling, alkylation, and de-protection reactions.

Methyl-amino-hexahydropyridazine-3-carboxylate-boronic ester (2) can be prepared in three steps, including protection, Iridium catalyst mediated borylation, and coupling with methyl (5V hexahydropyridazine-3-carboxylate. An appropriately substituted acetylpyrrolidine-3-carbonyl-N-methyl-L-valine (4) can be made by coupling of methyl-L-vaiinate and protected (S)-pyrrolidine-3-carboxylic acid, followed by deprotection, coupling with an appropriately substituted carboxylic acid, and a hydrolysis step.

The final macrocyclic esters can be made by coupling of methyl-amino-hexahydropyridazine-3- carboxylate-boronic ester (2) and intermediate (1) in the presence of Pd catalyst followed by hydrolysis and macrolactonization steps to result in an appropriately protected macrocyclic intermediate (5).

Deprotection and coupling with an appropriately substituted acetylpyrrolidine-3-carbonyl-N-methyl-L- valine (4) results in a macrocyclic product. Additional deprotection or functionalization steps are be required to produce a final compound. For example, a person of skill in the art would be able to install into a macrocyclic ester a desired -B-L-W group of a compound of Formula (I), where B, L and W are defined herein, including by using methods exemplified in the Example section herein. Scheme 2, Alternative general synthesis of macrocyclic esters

Alternatively, macrocyclic esters can be prepared as described in Scheme 2. An appropriately protected bromo-indolyl (6) can be coupled in the presence of Pd catalyst with boronlc ester (3), followed by iodination, deprotection, and ester hydrolysis. Subsequent coupling with methyl (S)- hexahydropyridazine-3-carboxylate, followed by hydrolysis and macrolactonization can result in iodo intermediate (7). Coupling in the presence of Pd catalyst with an appropriately substituted boronic ester and alkylation can yield fully a protected macrocycle (5). Additional deprotection or functionalization steps are required to produce a final compound. For example, a person of skill in the art would be able to install into a macrocyclic ester a desired -B-L-W group of a compound of Formula (I), where B, L and W are defined herein, including by using methods exemplified in the Example section herein.

Scheme 3. General synthesis of macrocyclic esters

Alternatively, fully a protected macrocycle (5) can be deprotected and coupled with an appropriately substituted coupling partners, and deprotected to results in a macrocyclic product. Additional deprotection or functionalization steps are be required to produce a final compound. For example, a person of skill in the art would be able to install into a macrocyclic ester a desired -B-L-W group of a compound of Formula (I), where B, L and W are defined herein, including by using methods exemplified in the Example section herein.

Scheme 4. General synthesis of macrocyclic esters

An alternative general synthesis of macrocyclic esters is outlined in Scheme 4. An appropriately substituted indolyl boronic ester (8) can be prepared in four stops starting from protected 3-(5-bromo-2- iodo-1H-indol-3-yl)-2,2-dimethylpropan-1-oi and appropriately substituted boronic acid, Including Palladium mediated coupling, alkylation, de-protoction, and Palladium mediated borylation reactions.

Methyl-amino-3-(4-bromothiazol-2-yl)propanoyl)hexahydropy ridazine-3-carboxylate (10) can be prepared via coupling of (S)-2-amino-3-(4-bromothiazol-2-yl)propanoic acid (9) with methyl (S)- hexahydropyridazine-3-carboxylate.

The final macrocyclic esters can be made by coupling of Methyl-amino-3-(4-bromothiazol-2- yl)propanoyl)hexahydropyridazine-3-carboxylate (10) and an appropriately substituted indoiyl boronic ester (8) in the presence of Pd catalyst followed by hydrolysis and macrolactonization steps to result in an appropriately protected macrocyclic Intermediate (11). Deprotection and coupling with an appropriately substituted carboxylic acid (or other coupling partner) or intermediate 4 can result in a macrocyclic product. Additional deprotection or functionalization steps could be required to produce a final compound 13 or 14.

In addition, compounds of the disclosure can be synthesized using the methods described in the Examples below, together with synthetic methods known in the art of synthetic organic chemistry, or variations thereon as appreciated by those skilled in the art. These methods include but are not limited to those methods described in the Examples below. For example, a person of skill in the art would be able to install into a macrocyclic ester a desired -B-L-W group of a compound of Formula (I) where B, L and W are defined herein, including by using methods exemplified in the Example section herein.

Pharmaceutical Compositions and Methods of Use Pharmaceutical Compositions and Methods of Administration

The compounds with which the invention is concerned are Ras inhibitors, and are useful in the treatment of cancer. Accordingly, one embodiment of the present invention provides pharmaceutical compositions containing a compound of the invention or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, as well as methods of using the compounds of the invention to prepare such compositions.

As used herein, the term “pharmaceutical composition” refers to a compound, such as a compound of the present invention, or a pharmaceutically acceptable salt thereof, formulated together with a pharmaceutically acceptable excipient.

In some embodiments, a compound is present in a pharmaceutical composition In unit dose amount appropriate for administration in a therapeutic regimen that shows a statistically significant probability of achieving a predetermined therapeutic effect when administered to a relevant population in some embodiments, pharmaceutical compositions may be specially formulated for administration in solid or liquid form, including those adapted for the following: oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), tablets, e.g., those targeted for buccal, sublingual, and systemic absorption, boluses, powders, granules, pastes for application to the tongue; parenteral administration, for example, by subcutaneous, intramuscular, Intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained-release formulation; topical application, for example, as a cream, ointment, or a controlled-reiease patch or spray applied to the skin, lungs, or oral cavity; intravaginally or intrarectally, for example, as a pessary, cream, or foam; sublingually; ocularly; transdermally; or nasally, pulmonary, and to other mucosal surfaces

A “pharmaceutically acceptable excipient,” as used herein, refers any inactive ingredient (for example, a vehicle capable of suspending or dissolving the active compound) having the properties of being nontoxic and non-inflammatory in a subject. Typical excipients include, for example: antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, giidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspensing or dispersing agents, sweeteners, or waters of hydration. Excipients include, but are not limited to: butylated optionally substituted hydroxyltoiuene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmeilose, crosslinked polyvinyl pyrroiidone, citric acid, crospovidone, cysteine, ethylcelluiose, gelatin, optionally substituted hydroxylpropyl cellulose, optionally substituted hydroxylpropyl methylcellulose, lactose, magnesium stearate, maltitol, mannitol, methionine, methylcellulose, methyl paraben, microcrystalline cellulose, polyethylene glycol, polyvinyl pyrroiidone, povidone, pregelatinized starch, propyl paraben, retinyl palmitate, shellac, silicon dioxide, sodium carboxymethyl cellulose, sodium citrate, sodium starch glycoiate, sorbitol, starch (corn), stearic acid, stearic acid, sucrose, talc, titanium dioxide, vitamin A, vitamin E, vitamin C, and xylitol. Those of ordinary skill in the art are familiar with a variety of agents and materials useful as excipients. See, e.g., e.g., Ansel, et al., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems. Philadelphia: Lippincott, Williams & Wilkins, 2004; Gennaro, et al., Remington: The Science and Practice of Pharmacy. Philadelphia: Llppincott, Williams & Wilkins, 2000; and Rowe, Handbook of Pharmaceutical Excipients. Chicago, Pharmaceutical Press, 2005. In some embodiments, a composition includes at least two different pharmaceutically acceptable excipients.

Compounds described herein, whether expressly stated or not, may be provided or utilized in salt form, e.g., a pharmaceutically acceptable salt form, unless expressly stated to the contrary. The term “pharmaceutically acceptable salt," as use herein, refers to those salts of the compounds described herein that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and other animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, pharmaceutically acceptable salts are described in: Berge et al., J. Pharmaceutical Sciences 66:1-19, 1977 and in Pharmaceutical Saits: Properties, Selection, and Use, (Eds. P.H. Stahl and C.G. Wermuth), Wiley-VCH, 2008. The salts can be prepared in situ during the final isolation and purification of the compounds described herein or separately by reacting the free base group with a suitable organic acid.

The compounds of the invention may have ionizable groups so as to be capable of preparation as pharmaceutically acceptable salts. These salts may be acid addition salts involving inorganic or organic acids or the salts may, in the case of acidic forms of the compounds of the invention, be prepared from inorganic or organic bases. In some embodiments, the compounds are prepared or used as pharmaceutically acceptable salts prepared as addition products of pharmaceutically acceptable acids or bases. Suitable pharmaceutically acceptable acids and bases are well-known in the art, such as hydrochloric, sulfuric, hydrobromic, acetic, lactic, citric, or tartaric acids for forming acid addition salts, and potassium hydroxide, sodium hydroxide, ammonium hydroxide, caffeine, various amines, and the like for forming basic salts. Methods for preparation of the appropriate salts are well-established in the art.

Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate. camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-optlonally substituted hydroxyl-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate. maleale, maionate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate. phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, toluenesulfonate, undecanoate, valerate salts and the like. Representative alkali or alkaline earth metal salts Include sodium, lithium, potassium, calcium, magnesium and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine. trimethylamine, triethylamlne, ethylamine and the like.

As used herein, the term “subject” refers to any member of the animal kingdom. In some embodiments, “subject" refers to humans, at any stage of development. In some embodiments, “subject” refers to a human patient. In some embodiments, “subject” refers to non-human animais. In some embodiments, the non-human animal is a mammal (e.g., a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a sheep, cattle, a primate, or a pig). In some embodiments, subjects Include, but are not limited to, mammals, birds, reptiles, amphibians, fish, or worms. In some embodiments, a subject may be a transgenic animal, genetically-engineered animal, or a clone. As used herein, the term “dosage form” refers to a physically discrete unit of a compound (e g., a compound of the present invention) for administration to a subject. Each unit contains a predetermined quantity of compound. In some embodiments, such quantity is a unit dosage amount (or a whole fraction thereof) appropriate for administration in accordance with a dosing regimen that has been determined to correlate with a desired or beneficial outcome when administered to a relevant population (i.e. , with a therapeutic dosing regimen). Those of ordinary skill in the art appreciate that the totai amount of a therapeutic composition or compound administered to a particular subject is determined by one or more attending physicians and may involve administration of multiple dosage forms.

As used herein, the term “dosing regimen” refers to a set of unit doses (typically more than one) that are administered individually to a subject, typically separated by periods of time. In some embodiments, a given therapeutic compound (e.g., a compound of the present invention) has a recommended dosing regimen, which may involve one or more doses. In some embodiments, a dosing regimen comprises a plurality of doses each of which are separated from one another by a time period of the same length; in some embodiments, a dosing regimen comprises a plurality of doses and at least two different time periods separating Individual doses. In some embodiments, all doses within a dosing regimen are of the same unit dose amount in some embodiments, different doses within a dosing regimen are of different amounts in some embodiments, a dosing regimen comprises a first dose In a first dose amount, followed by one or more additional doses in a second dose amount different from the first dose amount. In some embodiments, a dosing regimen comprises a first dose in a first dose amount, followed by one or more additional doses in a second dose amount same as the first dose amount. In some embodiments, a dosing regimen is correlated with a desired or beneficial outcome when administered across a relevant population (i.e., is a therapeutic dosing regimen).

A “therapeutic regimen” refers to a dosing regimen whose administration across a relevant population is correlated with a desired or beneficial therapeutic outcome.

The term "treatment” (also “treat” or "treating”), in its broadest sense, refers to any administration of a substance (e.g , a compound of the present invention) that partially or completely alleviates, ameliorates, relieves, inhibits, delays onset of, reduces severity of, or reduces incidence of one or more symptoms, features, or causes of a particular disease, disorder, or condition. In some embodiments, such treatment may be administered to a subject who does not exhibit signs of the relevant disease, disorder or condition or of a subject who exhibits only early signs of the disease, disorder, or condition. Alternatively, or additionally, in some embodiments, treatment may be administered to a subject who exhibits one or more esfabiished signs of the relevant disease, disorder or condition. In some embodiments, treatment may be of a subject who has been diagnosed as suffering from the relevant disease, disorder, or condition. In some embodiments, treatment may be of a subject known to have one or more susceptibility factors that are statistically correlated with increased risk of development of the relevant disease, disorder, or condition.

The term “therapeutically effective amount” means an amount that is sufficient, when administered to a population suffering from or susceptible to a disease, disorder, or condition in accordance with a therapeutic dosing regimen, to treat the disease, disorder, or condition. In some embodiments, a therapeutically effective amount is one that reduces the incidence or severit of, or delays onset of, one or more symptoms of the disease, disorder, or condition. Those of ordinary skill in the art will appreciate that the term “therapeutically effective amount” does not in fact require successful treatment be achieved in a particular individual. Rather, a therapeutically effective amount may be that amount that provides a particular desired pharmacological response in a significant number of subjects when administered to patients in need of such treatment. It is specifically understood that particular subjects may, In fact, be “refractory” to a “therapeutically effective amount." In some embodiments, reference to a therapeutically effective amount may be a reference to an amount as measured in one or more specific tissues (e.g., a tissue affected by the disease, disorder or condition) or fluids (e.g., blood, saliva, serum, sweat, tears, urine). Those of ordinary skill In the art will appreciate that, in some embodiments, a therapeutically effective amount may be formulated or administered in a single dose. In some embodiments, a therapeutically effective amount may be formulated or administered In a plurality of doses, for example, as part of a dosing regimen.

For use as treatment of subjects, the compounds of the invention, or a pharmaceutically acceptable salt thereof, can be formulated as pharmaceutical or veterinary compositions. Depending on the subject to be treated, the mode of administration, and the type of treatment desired, e.g., prevention, prophylaxis, or therapy, the compounds, or a pharmaceutically acceptable salt thereof, are formulated in ways consonant with these parameters. A summary of such techniques may be found in Remington: The Science and Practice of Pharmacy, 21 st Edition , Lippincott Williams & Wilkins, (2005); and Encyclopedia of Pharmaceutical Technoiogy , eds. J. Swarbrick and J. C. Boylan, 1988-1999, Marcel Dekker, New York, each of which is incorporated herein by reference.

Compositions can be prepared according to conventional mixing, granulating or coating methods, respectively, and the present pharmaceutical compositions can contain from about 0.1% to about 99%, from about 5% to about 90%, or from about 1% to about 20% of a compound of the present invention, or pharmaceutically acceptable salt thereof, by weight or volume. In some embodiments, compounds, or a pharmaceutically acceptable salt thereof, described herein may be present in amounts totaling 1-95% by weight of the total weight of a composition, such as a pharmaceutical composition.

The composition may be provided in a dosage form that is suitable for intraarticular. oral, parenteral (e.g., intravenous, intramuscular), rectal, cutaneous, subcutaneous, topical, transdermal, sublingual, nasal, vaginal, intravesicular, iniraurethrai, intrathecal, epidural, aural, or ocular administration, or by injection, inhalation, or direct contact with the nasal, genitourinary, reproductive or oral mucosa. Thus, the pharmaceutical composition may be in the form of, e.g., tablets, capsules, pills, powders, granulates, suspensions, emulsions, solutions, gels including hydrogels, pastes, ointments, creams, plasters, drenches, osmotic delivery devices, suppositories, enemas, injectabies, implants, sprays, preparations suitable for iontophoretic delivery, or aerosols. The compositions may be formulated according to conventional pharmaceutical practice.

As used herein, the term “administration" refers to the administration of a composition (e.g., a compound, or a preparation that includes a compound as described herein) to a subject or system. Administration to an animal subject (e.g., to a human) may be b any appropriate route. For example, in some embodiments, administration may be bronchial (including by bronchial instillation), buccal, enteral, interdermal, intra-arterial, intradermal, intragastrle, intramedullary, intramuscular, intranasal, intraperitoneal, intrathecal, intravenous, intraventricular, mucosal, nasal, oral, rectal, subcutaneous, sublingual, topical, tracheal (including by intratracheal instillation), transdermal, vaginal or vitreai.

Formulations may be prepared in a manner suitable for systemic administration or topical or local administration. Systemic formulations include those designed for injection (e.g., Intramuscular, intravenous or subcutaneous injection) or may be prepared for transdermal, transmucosai, or oral administration. A formulation will generally include a diluent as well as, in some cases, adjuvants, buffers, preservatives and the like. Compounds, or a pharmaceutically acceptable salt thereof, can be administered also In liposomal compositions or as microemulsions.

For injection, formulations can be prepared in conventional forms as liquid solutions or suspensions or as solid forms suitable for solution or suspension in liquid prior to injection or as emulsions. Suitable excipients include, for example, wafer, saline, dextrose, glycerol and the like. Such compositions may also contain amounts of nontoxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents and the like, such as, for example, sodium acetate, sorbitan monolaurate, and so forth.

Various sustained release systems for drugs have also been devised. See, for example, U.S. Patent No. 5,624,677.

Systemic administration may also include relatively noninvasive methods such as the use of suppositories, transdermal patches, transmucosai delivery and intranasal administration. Oral administration is also suitable for compounds of the Invention, or a pharmaceutically acceptable salt thereof. Suitable forms include syrups, capsules, and tablets, as is understood in the art.

Each compound, or a pharmaceutically acceptable salt thereof, as described herein, may be formulated in a variety of ways that are known in the art. For example, the first and second agents of the combination therapy may be formulated together or separately. Other modalities of combination therapy are described herein.

The individually or separately formulated agents can be packaged together as a kit. Non-limiting examples include, but are not limited to, kits that contain, e.g., two pills, a pill and a powder, a suppository and a liquid in a vial, two topical creams, etc. The kit can include optional components that aid in the administration of the unit dose to subjects, such as vials for reconstituting powder forms, syringes for injection, customized IV delivery systems, inhalers, etc. Additionally, the unit dose kit can contain instructions for preparation and administration of the compositions. The kit may be manufactured as a single use unit dose for one subject, multiple uses for a particular subject (at a constant dose or in which the individual compounds, or a pharmaceutically acceptable salt thereof, may vary in potency as therapy progresses); or the kit may contain multiple doses suitable for administration to multiple subjects (“bulk packaging”). The kit components may be assembled in cartons, blister packs, bottles, tubes, and the like.

Formulations for oral use Include tablets containing the active ingredient(s) in a mixture with non-toxic pharmaceutically acceptable excipients. These excipients may be, for example, Inert diluents or fillers (e.g., sucrose, sorbitol, sugar, mannitol, microcrystalline cellulose, starches including potato starch, calcium carbonate, sodium chloride, lactose, calcium phosphate, calcium sulfate, or sodium phosphate); granulating and disintegrating agents (e.g., cellulose derivatives including microcrystailine cellulose, starches including potato starch, croscarmellose sodium, alginates, or alginic acid); binding agents (e.g., sucrose, glucose, sorbitol, acacia, alginic acid, sodium alginate, gelatin, starch, pregeiatinized starch, microcrystalline cellulose, magnesium aluminum silicate, carboxymethylcellulose sodium, methyleellulose, optionally substituted hydrcxylpropyl methylcellulose, ethylcelluiose, polyvinylpyrrolidone, or polyethylene glycol); and lubricating agents, glidants, and antiadhesives (e.g., magnesium stearate, zinc stearate, stearic acid, silicas, hydrogenated vegetable oils, or talc). Other pharmaceutically acceptable excipients can be colorants, flavoring agents, plasticizers, humectants, buffering agents, and the like.

Two or more compounds may be mixed together in a tablet, capsule, or other vehicle, or may be partitioned. In one example, the first compound is contained on the inside of the tablet, and the second compound is on the outside, such that a substantial portion of the second compound is released prior to the release of the first compound.

Formulations for oral use may aiso be provided as chewable tablets, or as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent (e.g , potato starch, lactose, mlcrocrystalline cellulose, calcium carbonate, calcium phosphate or kaolin), or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin, or olive oil. Powders, granulates, and pellets may be prepared using the ingredients mentioned above under tablets and capsules in a conventional manner using, e.g., a mixer, a fluid bed apparatus or a spray drying equipment.

Dissolution or diffusion-controlled release can be achieved by appropriate coating of a tablet, capsule, pellet, or granulate formulation of compounds, or by incorporating the compound, or a pharmaceutically acceptable salt thereof, Into an appropriate matrix. A controlled release coating may include one or more of the coating substances mentioned above or, e.g., shellac, beeswax, glycowax, castor wax, carnauba wax, stearyl alcohol, glyceryl monostearate, glyceryl distearate, glycerol palmltostearate, ethylceliuiose, acrylic resins, dl-poiylaciic acid, cellulose acetate butyrate, polyvinyl chloride, polyvinyl acetate, vinyl pyrrolidone, polyethylene, polymethacrylate, methylmethacrylate, 2-optionally substituted hydroxylmethacrylate, methacrylate hydrogels, 1 ,3 butylene glycol, ethylene glycol methacrylate, or polyethylene glycois. in a controlled release matrix formulation, the matrix material may also include, e.g., hydrated methylcellulose, carnauba wax and stearyl alcohol, carbopoi 934, silicone, glyceryl tristearate, methyl acrylate-methyl methacrylate, polyvinyl chloride, polyethylene, or halogenated fluorocarbon.

The liquid forms in which the compounds, or a pharmaceutically acceptable salt thereof, and compositions of the present invention can be incorporated for administration orally include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.

Generally, when administered to a human, the oral dosage of any of the compounds of the invention, or a pharmaceutically acceptable salt thereof, will depend on the nature of the compound, and can readlly be determined by one skilled in the art. A dosage may bo, for example, about 0.001 mg to about 2000 mg per day, about 1 mg to about 1000 mg per day, about 5 mg to about 500 mg per day, about 100 mg to about 1500 mg per day, about 500 g to about 1500 g per day, about 500 g to about 2000 mg per day, or any range derivable therein.

In some embodiments, the pharmaceutical composition may further comprise an additional compound having antiproliferative activity. Depending on the mode of administration, compounds, or a pharmaceutically acceptable salt thereof, will be formulated into suitable compositions to permit facile delivery. Each compound, or a pharmaceutically acceptable salt thereof, of a combination therapy may be formulated In a variety of ways that are known in the art. For example, the first and second agents of the combination therapy may be formulated together or separately. Desirably, the first and second agents are formulated together for the simultaneous or near simultaneous administration of the agents.

It will be appreciated that the compounds and pharmaceutical compositions of the present invention can be formulated and employed in combination therapies, that is, the compounds and pharmaceutical compositions can be formulated with or administered concurrently with, prior to, or subsequent to, one or more other desired therapeutics or medical procedures. The particular combination of therapies (therapeutics or procedures) to employ in a combination regimen will take into account compatibility of the desired therapeutics or procedures and the desired therapeutic effect to be achieved. It will also be appreciated that the therapies employed may achieve a desired effect for the same disorder, or they may achieve different effects (e.g., control of any adverse effects).

Administration of each drug in a combination therapy, as described herein, can, Independently, be one to four times daily for one day to one year, and may even be for the life of the subject. Chronic, long-term administration may be indicated.

Numbered Embodiments

1] A compound, or pharmaceutically acceptable salt thereof, having the structure of Formula I:

Formula I wherein the doted lines represent 2ero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R i0 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene;

B is -CH(R 9 )- or >C=CR 9 R 9 where the carbon is bound to the carbonyl carbon of -N(R I 1 )C(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

G is optionally substituted C 1 -C 4 alkylene, optionally substituted C 1 -C 4 alkenylene, optionally substituted C 1 -C 4 heteroalkylene, -C(O)O-CH(R 6 )- where C is bound to -C(R 7 R 8 )-, -C(O)NH-€H(R 6 )-

1176

5UB5TITUTE SHEET (RULE 26) where C is bound to -C(R 7 R 8 )-, optionally substituted C 1 -C 4 heteroalkylene. or 3 to 8-membered heteroarylene;

L is absent or a linker;

W is hydrogen, cyano, optionally substituted amino, optionally substituted C 1 -C 4 alkoxy, optionally substituted C 1 -C 4 hydroxyalkyl, optionally substituted C 1 -C 4 aminoalkyl, optionally substituted C 1 -C 4 haloalkyl, optionally substituted C 1 -C 4 alkyl, optionally substituted C 1 -C 4 guanidinoalkyl, C0-C 4 alkyl optionally substituted 3 to 11-membered heterocycloalkyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 3 to 8-membered heteroaryl;

X 1 is optionally substituted C 1 -C 2 alkylene, NR, O, or S(O) n ;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C -C alkynyl, C(O)R’, C(O)OR’, C(O)N(R’) 2 , S(O)R’, S(O) 2 R’, or S(O)sN(R’) s ; each R' is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, Independently, C or N;

Y 5 is CH, CH 2 . or N;

Y 6 is C(O), CH, CH 2 , or N;

R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl, or

R 1 and R 2 combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 2 is absent, hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent or R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8-membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C 1 -C 3 alkyl), C= O, C= S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7a and R 8a are, Independently, hydrogen, halo, optionally substituted C 1 -C 3 alkyl, or combine with the carbon to which they are attached to form a carbonyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is hydrogen, F, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl ;

R 9 and L combine with the atoms to which they are attached to form an optionally substituted 3 to 14-membered heterocycloalkyl;

R 9 is hydrogen or optionally substituted C 1 -C 6 alkyl;

R 10 is hydrogen, haio, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl;

R 10a is hydrogen or haio;

R 1 ' is hydrogen or C 1 -C 3 alkyl; and

R 16 is hydrogen or C 1 -C 3 alkyl.

[2] The compound, or pharmaceutically acceptable salt thereof, of paragraph [1], wherein G is optionally substituted C 1 -C 4 heteroalkylene.

[3] The compound, or pharmaceutically acceptable salt thereof, of paragraph [1] or [2], wherein the compound has the structure of Formula ic: wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CHsiCiGHCH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -N(R 1 , )C(O)- optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is hydrogen, optionally substituted amino, optionally substituted C 1 -C 4 alkoxy, optionally substituted C 1 -C 4 hydroxyalkyl, optionally substituted C 1 -C 4 aminoalkyl, optionally substituted C 1 -C 4 haloalkyl, optionally substituted C 1 -C 4 alkyl, optionally substituted C 1 -C 4 guanidinoalkyl, C0-C 4 alkyl optionally substituted 3 to 11-membered heterocycloalkyl, optionally substituted 3 to 8-membered cycloalkyl, or optionally susbtituted 3 to 8-membered heteroaryl;

X 2 is O or NH;

X 3 is N or CH; n is 0, 1 , or 2;

R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C -C alkynyl, C(O)R’, C(O)OR’, C(O)N(R’) 2 , S(O)R’, S(O) 2 R’, or S(O)sN(R’) s ; each R’ is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, Independently, C or N;

Y 5 and Y 6 are, independently, CH or N;

R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl;

R 2 is hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent or R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8- membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C;-C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C8 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-memhered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C 1 -C 3 alkyl), C=O, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8 ' ' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl; R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; R 10 is hydrogen, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl; and R 11 is hydrogen or C 1 -C 3 alkyl.

[4] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1 ] to

[3], wherein X 2 is NH.

[5] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] fo

[4], wherein X 3 is CH.

[6] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] fo

[5], wherein R 11 is hydrogen.

[7] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] fo [5], wherein R 11 is C 1 -C 3 alkyl.

[8] The compound, or pharmaceutically acceptable salt thereof, of paragraph [7], wherein R” is methyl.

[9] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[6], wherein the compound has the structure of Formula Id:

Formula id wherein the dotted lines represent zero, one, two, three, or four non-adjacent double bonds;

A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6- membered heteroarylene; B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heteroeycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is hydrogen, optionally substituted amino, optionally substituted C 1 -C 4 alkoxy, optionally substituted C 1 -C 4 hydroxyalkyl, optionally substituted C 1 -C 4 aminoalkyl, optionally substituted C 1 -C 4 haloalkyl, optionally substituted C 1 -C 4 alkyl, optionally substituted C 1 -C 4 guanidinoalkyl, C0-C 4 alkyl optionally substituted 3 to 11-membered heterocycloalkyl, optionally substituted 3 to 8-membered cycloalkyl, or optionally susbtituted 3 to 8-membered heteroaryl; n is 0, 1 , or 2; R is hydrogen, cyano, optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, C(O)R', C(O)OR’, C(O)N(R’) 2 , S(O)R’, S(O) 2 R·, or S(O) 2 N(R’) 2 ; each R' is, independently, H or optionally substituted C 1 -C 4 alkyl;

Y 1 is C, CH, or N;

Y 2 , Y 3 , Y 4 , and Y 7 are, Independently, C or N;

Y 5 and Y 6 are, independently, CH or N;

R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl;

R 2 is hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heterocycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent or R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8- membered cycloalkyl or optionally substituted 3 to 14-membered heterocycloalkyl;

R 4 is absent, hydrogen, halogen, cyano, or methyl optionally substituted with 1 to 3 halogens;

R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl, or cyclobutyl;

R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or

R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C 1 -C 3 alkyl), C=O, C=S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl;

R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; and R 10 is hydrogen, hydroxy, G.-Cs alkoxy, or C 1 -C 3 alkyl. 10] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [9] wherein X 1 is optionally substituted C 1 -C 2 alkylene. 11] The compound, or pharmaceutically acceptable salt thereof, of paragraph [10], wherein X 1 is methylene. 12] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [11], wherein R 5 is hydrogen.

[13] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [11], wherein R 5 is C 1 -C 4 alkyl optionally substituted with halogen.

[14] The compound, or pharmaceutically acceptable salt thereof, of paragraph [13], wherein R 5 is methyl.

[15] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[14], wherein Y 4 is G.

[16] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[15], wherein R 4 is hydrogen

[17] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[16], wherein Y 5 is CH.

[18] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[17], wherein Y 6 is CH.

[19] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[18], wherein Y 1 is G.

[20] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[19], wherein Y 2 is C.

[21] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[20], wherein Y 3 is N.

[22] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[21], wherein R 3 is absent

[23] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[22], wherein Y 7 is C.

[24] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [6] or [9] to [23], wherein the compound has the structure of Formula le:

Formula le wherein A is -N(H or CHsJCiOKCH?)- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is hydrogen, optionally substituted amino, optionally substituted C 1 -C 4 alkoxy, optionally substituted C 1 -C 4 hydroxyalkyl, optionally substituted C 1 -C 4 aminoalkyl, optionally substituted C 1 -C 4 haloalkyl, optionally substituted C 1 -C 4 alkyl, optionally substituted C 1 -C 4 guanidinoalkyl, C0-C 4 alkyl optionally substituted 3 to 11-membered heterocycloalkyl, optionally substituted 3 to 8-membered cycloalkyl, or optionally susbtituted 3 to 8-membered heteroaryl;

R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heteroeycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl;

R 2 is hydrogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 7-membered heteroeycloalkyl, optionally substituted 6-membered aryl, optionally substituted 5 or 6-membered heteroaryl; R 3 is absent or R 2 and R 3 combine with the atom to which they are attached to form an optionally substituted 3 to 8- membered cycloalkyl or optionally substituted 3 to 14-membered heteroeycloalkyl; R 5 is hydrogen, C 1 -C 4 alkyl optionally substituted with halogen, cyano, hydroxy, or C 1 -C 4 alkoxy, cyclopropyl. or cyclobulyl; R 6 is hydrogen or methyl; R 7 is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl, or R 6 and R 7 combine with the carbon atoms to which they are attached to form an optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heteroeycloalkyl; R 8 is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted G2-C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heteroeycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or R 7 and R 8 combine with the carbon atom to which they are attached to form C=CR 7' R 8' ; C=N(OH), C=N(O-C 1 -C 3 alkyl), C= O, C= S, C=NH, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; R 7' is hydrogen, halogen, or optionally substituted C 1 -C 3 alkyl; R 8 · ' is hydrogen, halogen, hydroxy, cyano, optionally substituted C 1 -C 3 alkoxy, optionally substituted C 1 -C 3 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted 3 to 8-membered cycloalkyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 5 to 10-membered heteroaryl, or optionally substituted 6 to 10-membered aryl, or

R 7' and R 8' combine with the carbon atom to which they are attached to form optionally substituted 3 to 6-membered cycloalkyl or optionally substituted 3 to 7-membered heterocycloalkyl; R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl; and R 10 is hydrogen, hydroxy, C 1 -C 3 alkoxy, or C 1 -C 3 alkyl.

[25] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [3] to

[24], wherein R 6 is hydrogen.

[26] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[25], wherein R 2 is hydrogen, cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted 3 to 6- membered cycloalkyl, or optionally substituted 3 to 6-membered heterocycloalkyl.

[27'] The compound, or pharmaceutically acceptable salt thereof, of paragraph [26], wherein R 2 is optionally substituted C 1 -C 6 alkyl.

[28] The compound, or pharmaceutically acceptable salt thereof, of paragraph [27], wherein R 2 is ethyl.

[29] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [28], wherein R 7 is optionally substituted C -C alkyl.

[30] The compound, or pharmaceutically acceptable salt thereof, of paragraph [29], wherein R 7 is C 1 -C 3 alkyl.

[31] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [30], wherein R 8 is optionally substituted C 1 -C 3 alkyl.

[32] The compound, or pharmaceutically acceptable salt thereof, of paragraph [31], wherein R 8 is C 1 -C 3 alkyl.

[33] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [32], wherein the compound has the structure of Formula If: Formula If wherein A is -N(H or CH 3 )C(O)-(CH 2 )- where the amino nitrogen is bound to the carbon atom of -CH(R 10 )-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is hydrogen, optionally substituted am.no, optionally substituted G1-C 4 alkoxy, optionally substituted C1-C 4 hydroxyalkyl, optionally substituted C 1 -C 4 aminoalkyl, optionally substituted C 1 -C 4 haloalkyl, optionally substituted C 1 -C 4 alkyl, optionally substituted C 1 -O. guanidinoalkyl, C0-C 4 alkyl optionally substituted 3 to 11-membered heterocycloalkyl, optionally substituted 3 to 8-membered cycloalkyl, or optionally susbtltuted 3 to 8-membered heteroaryl;

R 1 is cyano, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl;

R 2 is C 1 -C 6 alkyl or 3 to 6-membered cycloalkyl;

R 7 is C 1 -C 3 alkyl;

R B is C 1 -C 3 alkyl; and

R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl.

[34] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [33], wherein R 1 is 5 to 10-membered heteroaryl.

[35] The compound, or pharmaceutically acceptable salt thereof, of paragraph [34], wherein R 1 is optionally substituted 6-membered aryl or optionally substituted 6-membered heteroaryl.

[36] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [35], w ereln the compound has the structure of Formula Ig: wherein A is, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is hydrogen, optionally substituted amino, optionally substituted C 1 -C 4 alkoxy, optionally substituted C 1 -C 4 hydroxyalkyl, optionally substituted C 1 -C 4 aminoalkyl, optionally substituted C 1 -C 4 haloalkyl, optionally substituted C 1 -C 4 alkyl, optionally substituted C 1 -O. guanidinoalkyl, C0-C 4 alkyl optionally substituted 3 to 11-membered heterocycloalkyl, optionally substituted 3 to 8-membered cycloalkyl, or optionally susbtituted 3 to 8-membered heteroaryl; R 2 is C 1 -C 6 alkyl or 3 to 6-membered cycloalkyl;

R 7 is C 1 -Cs alkyl; R 8 is C 1 -C 3 alkyl;

R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl;

X e is N, CH, or CR' 7 ;

X f is N or CH;

R 12 is optionally substituted C 1 -C 6 alkyl or optionally substituted C 1 -C 6 heteroalkyl; and

R 17 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl.

[37] The compound, or pharmaceutically acceptable salt thereof, of paragraph [36], wherein X e is N and X f is CH.

[38] The compound, or pharmaceutically acceptable salt thereof, of paragraph [36], wherein X e is CH and X f is N.

[39] The compound, or pharmaceutically acceptable salt thereof, of paragraph [36], wherein X e is CR 17 and X f is N.

[40] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [36] to [39], wherein R 12 is optionally substituted C 1 -C 6 heteroalkyl.

[41] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [36] [42] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [41], wherein the compound has the structure of Formula Ih:

Formula in wherein A is optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

B is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membered arylene, or 5 to 6-membered heteroarylene;

L is absent or a linker;

W is hydrogen, optionaliy substituted amino, optionally substituted C 1 -C 4 alkoxy, optionally substituted C 1 -C 4 hydroxyalkyl, optionally substituted C 1 -C 4 aminoalkyl, optionally substituted C 1 -C 4 haloalkyl, optionally substituted C 1 -C 4 alkyl, optionally substituted C 1 -C 4 guanidinoalkyl, C0-C 4 alkyl optionally substituted 3 to 11-membered heterocycloalkyl, optionally substituted 3 to 8-membered cycloalkyl, or optionally substituted 3 to 8-membered heteroaryl;

R 2 is C 1 -C 6 alkyl or 3 to 6-membered cycloalkyl;

R 7 is C 1 -C 3 alkyl;

R 8 is C 1 -C 3 alkyl;

R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyL or optionally substituted 3 to 7-membered heterocycloalkyl;

X e is CH, or CR' 7 ; and

R 17 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered eycloalkyl, optionally substituted 3 to 6-membered cycloalkenyl, optionally substituted 3 to 6-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted 5 to 10-membered heteroaryl. [43] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [42], wherein the compound has the structure of Formula li:

Formula li wherein A is optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6- membered heterocycloalkylene, optionally substituted 6-membered arylene, or optionally substituted 5 to 6-membered heteroarylene;

R is -CH(R 9 )- where the carbon is bound to the carbonyl carbon of -NHC(O)-, optionally substituted 3 to 6-membered cycloalkylene, optionally substituted 3 to 6-membered heterocycloalkylene, optionally substituted 6-membored aryleno, or 5 to 6-mombered heteroarylene;

L is absent or a linker;

W is hydrogen, optionally substituted amino, optionally substituted C 1 -C 4 alkoxy, optionally substituted C 1 -C 4 hydroxyalkyl, optionally substituted C 1 -C 4 aminoalkyl, optionally substituted C 1 -C 4 haloalkyl, optionally substituted C 1 -C 4 alkyl, optionally substituted C 1 -C 4 guanidinoalkyl, C0-C 4 alkyl optionally substituted 3 to 11-membered heterocycloalkyl, optionally substituted 3 to 8-membered cycloalkyl, or optionally substituted 3 to 8-membered heteroaryl; R 2 is C 1 -C 6 alkyl or 3 to 6-membered cycloalkyl; R 7 is C 1 -C 3 alkyl; R 8 is C 1 -C 3 alkyl; and R 9 is optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 heteroalkyl, optionally substituted 3 to 6-membered cycloalkyl, or optionally substituted 3 to 7-membered heterocycloalkyl. 44] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [43], wherein A is optionally substituted 6-membered arylene.

[45] The compound, or pharmaceutically acceptable salt thereof, of paragraph [44], wherein A has the structure: wherein R 13 is hydrogen, hydroxy, amino, cyano, optionally substituted CT-C 6 alkyl, or optionally substituted Ch-C 6 heteroalkyl. [46] The compound, or pharmaceutically acceptable salt thereof, of paragraph [45], wherein R' 3 is hydrogen. 47] The compound, or pharmaceutically acceptable salt thereof, of paragraph [45], wherein R' 3 is hydroxy.

[48] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [43], wherein A is optionally substituted 5 to 6-membered heteroarylene.

[49] The compound, or pharmaceutically acceptable salt thereof, of paragraph [48], wherein A is:

[50] The compound, or pharmaceutically acceptable salt thereof, of paragraph [49], wherein A is

[51] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [50], wherein B is -CHR 9 -.

[52] The compound, or pharmaceutically acceptable salt thereof, of paragraph [51], wherein R 9 is optionally substituted C 1 -C 6 alkyl or optionally substituted 3 to 6-membered cycloalkyl.

[53] The compound, or pharmaceutically acceptable salt thereof, of paragraph [52], wherein R 9

[54] The compound, or pharmaceutically acceptable salt thereof, of paragraph [53], wherein R 9

CH 3 js: U T 1 H 3

[55] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [50], wherein B is optionally substituted 6-membered arylene.

[56] The compound, or pharmaceutically acceptable salt thereof, of paragraph [55], wherein B is 6-membered arylene. The compound, or pharmaceutically acceptable salt thereof, of paragraph [56], wherein B is:

[58] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [50], wherein B is absent.

[59] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[58], wherein R 7 is methyl.

[60] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[59], wherein R 8 is methyl.

[61] The compound, or pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to

[60], wherein the linker is the structure of Formula II: A 1 -(B’ )f-(C ' )g-(B 2 )h-{ D’ )-(B 3 )i-(C 2 )j-(B 4 )k—A 2

Formula II where A' is a bond between the linker and B; A 2 is a bond between W and the linker; B 1 , B 2 , B 3 , and B 4 each, independentiy, is selected from optionally substituted C 1 -C 2 alkylene, optionally substituted C 1 -C 3 heteroalkylene, O, S, and NR N ; R N is hydrogen, optionally substituted C 1 -C 4 alkyl, optionally substituted C 1 -C 3 cycloalkyl, optionally substituted C 2 -C 4 alkenyl, optionally substituted C 2 -C 4 alkynyl, optionally substituted 3 to 14-membered heterocycloalkyl, optionally substituted 6 to 10-membered aryl, or optionally substituted C 1 -C 7 heteroalkyl; C 1 and G 2 are each, independentiy, selected from carbonyl, thiocarbonyl, sulphonyl, or phosphoryl; f, g, h, i, I, and k are each, independently, 0 or 1 ; and D 1 is optionally substituted C 1 -C 10 alkylene, optionally substituted C 2 -C 10 alkenylene, optionally substituted C 2 - C 10 alkynylene, optionally substituted 3 to 14-membered heterocycloalkylene, optionally substituted 5 to 10-membered heteroarylene, optionally substituted 3 to 8-membered cycloalkylene, optionally substituted 6 to 10-membered arylene, optionally substituted C 2 -C 10 polyethylene glycolene, or optionally substituted C 1 -C 10 heteroalkylene, or a chemical bond linking A ! -(B 1 )f-(C i ) g -(B 2 )h- to -(B 3 )i-(C 2 )j-(B 4 )k— A 2 .

[82] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [61], wherein the linker is acyclic.

[63] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [62], wherein the linker has the structure of Formula lia:

Formula ila wherein X 3 is absent or N;

R 14 is absent, hydrogen, optionally substituted C 1 -C 6 alkyl, or optionally substituted C 1 -C 3 cycloalkyl; and

L 2 is absent, -C(O)-, -SO 2 -, optionally substituted C 1 -C 4 alkylene or optionally substituted C 1 -C 4 heteroalkylene, wherein at least one of X a , R 14 , or L 2 is present.

[64] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [63], wherein the linker has the structure: [65] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [64], wherein

CH 3 the linker has the structure ^ .

[66] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [61], wherein the linker is or comprises a cyclic group. [67] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [61] or [66], wherein the linker has the structure of Formula Mb:

Formula lIb wherein o is 0 or 1 ;

X b is C(O) or S0 2 ;

R 1S is hydrogen or optionally substituted C 1 -C 6 alkyl;

Gy is optionally substituted 3 to 8-membered cycloalkylene, optionally substituted 3 to 8- membered heterocycloalkylene, optionally substituted 6-10 memhered arylene, or optionally substituted 5 to 10-membered heteroarylene; and

L 3 is absent, -C(O)-, -SO 2 -, optionally substituted C 1 -C 4 alkylene or optionally substituted C 1 -C 4 heteroalkylene

[68] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [67], wherein the linker has the structure:

[69] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] herein W is hydrogen.

[70] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] herein W is optionally substituted amino.

[71] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [70], wherein W s or -N(CH 3 ) 2 . [72] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1 ] herein W is optionally substituted arrtido. [73] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [72], wherein W 74] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1], wherein W is optionally substituted C 1 -C 4 alkoxy.

[75] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [74], wherein Whoxy or iso-propoxy.

[76] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1], wherein W is optionally substituted C 1 -C 4 alkyl.

[77] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [76], wherein Whyl, ethyl, iso-propyl, tert-butyl, or benzyl.

[78] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1], wherein W is optionally substituted C 1 -C 4 hydroxyalkyl.

[79] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [78], wherein W

[80] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1], wherein W is optionally substituted C 1 -C 4 aminoalkyl.

[81] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [80], wherein W

[82] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1], wherein W is optionally substituted C 1 -C 4 haloalkyl.

[83] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [82], wherein W

CF 37 V X CHF 2 V " \ V'"CF 3 s, V"GHF 2

[84] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1], wherein W is optionally substituted C 1 -C 4 guanidinoalkyl.

[85] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [84], wherein W

[86] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1], wherein W is C0-C 4 alkyl optionally substituted 3 to 11-membered heterocycloalkyl. [87] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [88], wherein W

10

[88] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] herein W is optionally substituted 3 to 8-membered cycloalkyl.

[89] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [88], wherein W

[90] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] herein W is optionally substituted 3 to 8-membered heteroaryl. [91] The compound, or a pharmaceutically acceptable salt thereof, of paragraph [90], wherein W 92] The compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to 68], wherein W is optionally substituted 6- to 10-membered aryl. 93] The compound, or a pharmaceutically acceptable salt thereof, or paragraph [92], wherein W is phenyl, 4-hydroxy-phenyl, or 2,4-methoxy-phenyl.

[94] A compound, or a pharmaceutically acceptable salt thereof, of Table 1 or 2.

[95] A pharmaceutical composition comprising a compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [94] and a pharmaceutically acceptable excipient.

[96] A method of treating cancer In a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [94] or a pharmaceutical composition of paragraph [95].

[97'] The method of paragraph [96], wherein the cancer is pancreatic cancer, colorectal cancer, non-small cell lung cancer, gastric cancer, esophageal cancer, ovarian cancer or uterine cancer.

[98] The method of paragraph [97], wherein the cancer comprises a Ras mutation.

[99] The method of paragraph [98] wherein the Ras mutation is at position 12, 13 or 61

[100] The method of paragraph [98] wherein the Ras mutation is K-Ras G12C, K-Ras G12D, K- Ras G12V, K-Ras G12S, K-Ras G13C, K-Ras G13D, or K-Ras G61 L.

[101] A method of treating a Ras protein-related disorder in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [94] or a pharmaceutical composition of paragraph [95]

[102] A method of Inhibiting a Ras protein in a cell, the method comprising contacting the cell with an effective amount of a compound, or a pharmaceutically acceptable salt thereof, of any one of paragraphs [1] to [94] or a pharmaceutical composition of paragraph [95]

[103] The method of paragraph [101] or [102], wherein the Ras protein is K-Ras G12G, K-Ras G12D, K-Ras G12V, K-Ras G12S, K-Ras G13C, K-Ras G13D, or K-Ras Q61 L

[104] The method of paragraph [102] or [103], wherein the cell is a cancer cell.

[105] The method of paragraph [104], wherein the cancer cell is a pancreatic cancer cell, a colorectal cancer cell, a non-small cell lung cancer cell, a gastric cancer cell, an esophageal cancer cell, an ovarian cancer cell, or a uterine cancer cell.

Examples The disclosure is further illustrated by the following examples and synthesis examples, which are not to be construed as limiting this disclosure in scope or spirit to the specific procedures herein described. It is to be understood that the examples are provided to Illustrate certain embodiments and that no limitation to the scope of the disclosure is intended thereby. It is to be further understood that resort may be had to various other embodiments, modifications, and equivalents thereof which may suggest themselves to those skilled in the art without departing from the spirit of the present disclosure or scope of the appended claims.

Chemical Syntheses

Definitions used in the foilowing examples and elsewhere herein are:

CH 2 CI2, DCM Methylene chloride, Dichloromethane

CHaCN, MeCN Acetonitrile

Cui Copper (l) iodide

DIPEA Diisopropylethyl amine

DMF N,N-Dimethylformamide

EtOAc Ethyl acetate h hour H 2 O Water

HCl Hydrochloric acid

K3PO4 Potassium phosphate (tribasic)

MeOH Methanol

NasSC Sodium sulfate

NMP N-methyl pyrroiidone

Pd(dppf)Cl2 [1 ,1'-Bis(diphenylphosphino)ferrocene]dichloropaliadium(ll)

Instrumentation

Mass spectrometry data collection took place with a Shimadzu LCMS-2020, an Agilent 1260LC- 8120/8125MSD, a Shimadzu LCMS-2010EV, or a Waters Acquity UPLC, with either a QDa detector or SQ Detector 2. Samples were injected in their liquid phase onto a C-18 reverse phase. The compounds were eluted from the column using an acetonitrile gradient and fed into the mass analyzer. Initial data analysis took place with either Agilent ChemStation, Shimadzu [..absolutions, or Waters Masslynx NMR data was collected with either a Bruker AVANCE III HD 400MHz, a Bruker Ascend 500MHz instrument, or a Varian 400MHz, and the raw' data was analyzed with either TopSpin or Mestreiab Mnova. Synthesis of Intermediates

Intermediate 1. Synthesis of 3-{5-bromo-1-ethyl-2-[2-[(1S) 1-met oxyethyl]pyr!din-3- yl]irsdol-3-yl)-2,2-dimethyipropars-1-ol

Step 1. To a mixture of 3-((fe/†-butyldiphenylsllyi)oxy)-2,2-dimethylpropanoyl chloride (85 g, 137' mmol, crude) in DCM (120 ml_) at 0 °C under an atmosphere of Nz was added 1 M SnCL in DCM (137' mL, 137 mmol) slowly. The mixture was stirred at 0 °C for 30 min, then a solution of 5-bromo-1H-lndole (28.8 g, 137 mmol) in DCM (40 mL) was added dropwise. The mixture was stirred at 0 °C for 45 min, then dilufed with EtOAc (300 L), washed with brine (100 mL x 4), dried over NazSC and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 1-(5-bromo-1H-indol-3-yl)-3-((tert-butyldiphenylsllyl)oxy)-2 ,2-dimethylpropan-1- one (55 g, 75% yield). LCMS (ESI): m/z [M+Na] calc’d for C 2 sHszBrNO 2 SiNa 556 1 ; found 556.3.

Step 2. To a mixture of 1-(5-bromo-1H-indol-3-yl)-3-((tert-butyldiphenylsllyl)oxy)-2 ,2- dimethylpropan-1-one (50 g, 93 6 mmol) In THF (100 L) at 0 ,3 C under an atmosphere of Nz was added UBH4 (6.1 g, 281 mmol). The mixture was heated to 60 °C and stirred for 20 h, then MeOH (10 mL) and EtOAc (100 mL) were added and the mixture washed with brine (50 mL), dried over NazSCU, filtered and the filtrate concentrated under reduced pressure. The residue was diluted with DCM (50 mL), cooled to 10 c C and diludine (9 5 g, 37.4 mmol) and TsOH.HzO (890 mg, 4 7 mmol) added. The mixture was stirred at 10 °C for 2 h, filtered, the filtrate concentrated under reduced pressure and the residue was purified by silica gei column chromatography to give 1-(5-bromo-1 W-indol-3-yl)-3-((tert-butyldiphenylsllyi)oxy)-2,2- dimethylpropan-1-one (41 g, 84% yield). LCMS (ESI): m/z [M+H] calc’d for C 2 9H 3 4Br OSi 519.2; found 520 1 ; 1 H NMR (400 MHz, CDCI3) 6 7.96 (s, 1H), 775 - 7.68 (m, 5H), 7.46 - 7.35 (m, 6H), 7.23 - 7.19 (m, 2H), 6.87 (d, J == 2.1 Hz, 1H), 3.40 (s, 2H), 2.72 (s, 2H), 1.14 (s, 9H), 0.89 (s, 6H).

Step 3. To a mixture of 1-(5-bromo-1H-indol-3-yl)-3-((tert-butyldiphenylsllyl)axy)-2 ,2- dimethylpropan-1-one (1 5 g, 2.9 mmol) and iz (731 g, 2 9 mmol) in THF (15 ml) at rt was added AgOTf (888 mg, 3.5 mmol). The mixture was stirred at rt for 2 h, then diluted with EtOAc (200 mL) and washed with saturated NazSzO 3 (100 mL), dried over anhydrous NazSCh and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 5-bromo-3-(3-((tert-butyldiphenylsllyl)axy)-2,2-dimethylprop yl)-2-iodo-1H-indole (900 mg, 72% yield) as a solid. 1 H NMR (400 MHz, DMSO-cfe) 5 11 .70 (s, 1H), 7.68 (d, J = 1 .3 Hz, 1H), 7.64 - 7.62 (m, 4H), 7.46 - 7.43 (m, 6H), 7.24 - 7.22 (d, 1H), 7.14 - 7 12 (dd, J= 8.6, 1.6 Hz, 1H), 3.48 (s, 2H), 2 63 (s, 2H), 1.08 (s, 9H), 0.88 (s, 6H)

Step 4. To a stirred mixture of HCOOH (66.3 g, 1 .44 mol) in TEA (728 g, 7.2 mol) at 0 °C under an atmosphere of Ar was added (4S,5S)-2-chloro-2-methyl-1-(4-methylbenzenesulfonyl)-4,5-di phenyl-1 ,3- diaza-2-ruthenacyclopontane cymone (3.9 g, 6.0 mmol) portion-wise. The mixture was heated to 40 °C and stirred for 15 min, then cooled to rt and 1-(3-bromopyridin-2-yl)ethanone (120 g, 600 mmol) added in portions. The mixture was heated to 40 °C and stirred for an additional 2 h, then the solvent was concentrated under reduced pressure. Brine (2 L) was added to the residue, the mixture was extracted with EtOAc (4 x 700 mL), dried over anhydrous NA 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give (1 S)-1-(3- bromopyridin-2-yl)ethanol (100 g, 74% yield) a an oil. LCMS (ESI): m/z [M+H] calc’d for OHeBrNG 201 .1 ; found 201.9.

Step 5. To a stirred mixture of (1 S)-1-(3-bromopyridin-2-yl)e†hanoi (100 g, 495 mmol) in DMF (1 L) at 0 °C was added NaH, 60% dispersion in oil (14.25 g, 594 mmol) in portions. The mixture was stirred at 0 °C for 1 h. Mel (140.5 g, 990 mmol) was added dropwise at 0 °C and the mixture was allowed to warm to rt and stirred for 2 h. The mixture was cooled to 0 “C and saturated NH+CI (5 L) was added. The mixture was extracted with EtOAc (3 x 1 .5 L), dried over anhydrous NA 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 3-bromo-2-[(1 S)-1-methoxyethyl]pyridine (90 g, 75% yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for GsHioBrNO 215.0; found 215.9

Step 6. To a stirred mixture of 3-bromo-2-[(18)-1-methoxyethyl]pyridine (90 g, 417 mmol) and Pd(dppf)Cl2 (30.5 g, 41 .7 mmol) in toluene (900 mL) at rt under an atmosphere of Ar was added bis(pinacoiato)dlboron (127 g, 500 mmol) and KOAc (81 .8 g, 833 mmol) In portions. The mixture was heated to 100 ,3 G and stirred for 3 h. The filtrate was concentrated under reduced pressure and the residue was purified by AiaO 3 column chromatography to give 2-[(1 S)-1-methoxyethyl]-3-(4,4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridine (100 g, 63% yield) as a semi-solid. LCMS (ESI): m/z [M+H] calc’d for C 1 4H 2 2BNO 3 263.2; found 264.1 .

Step 7, To a stirred mixture of 5-bromo-3-[3-[(tert-butyldiphenylsllyl)axy]-2,2-dimethylprop yl]-2- ioda-1H-indaie (140 g, 217 mmol) and 2-[(1 S)-1-methoxyethyl]-3-(4.4,5,5-tetramethyl-1 ,3,2-dioxaborolan- 2-yl)pyridine (100 g, 380 mmol) in 1 ,4-dioxane (1 .4 L) at rt under an atmosphere of Ar was added K2CO 3 (74.8 g, 541 mmol), Pd(dppf)Cl2 (15.9 g, 21 .7 mmol) and H 2 O (280 mL) in portions. The mixture was heated to 85 °G and stirred for 4 h, then cooled, H 2 O (5 L) added and the mixture extracted with EtOAc (3 x 2 L). The combined organic layers wore washed with brine (2 x 1 L), dried over anhydrous Na2SO+ and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 5-bromo-3-[3-[(iefi-butyldiphenylsllyi)oxy]-2,2-dimethylprop yl]-2-[2-[(1 S)- 1-methoxyethyl]pyridin-3-yl]-1H-indole (71 g, 45% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for CsrH+sBrbbOaSi 654.2: found 655.1 .

Step 8. To a stirred mixture of 5-bromo-3-[3-[(iert-butyldiphenylsllyi)oxy]-2,2-dimethylprop yl]-2-[2- [(1 S)-1-methoxyethyl]pyridin-3-yl]-1H-indole (71 g, 108 mmol) in DMF (0.8 L) at 0 °C under an atmosphere of Nz was added CS2CO 3 (70.6 g, 217 mmol) and Etl (33.8 g, 217 mmol) in portions. The mixture was warmed to rt and stirred for 16 h then H 2 O (4 L) added and the mixture extracted with EtOAc (3 x 1 .5 L). The combined organic layers were washed with brine (2 x 1 L), dried over anhydrous NazSCk and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 5-bromo-3-[3-[(tert-butyldiphenylsilyl)oxy]-2,2-dimethylprop yl]-1-ethyl- 2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]indole (66 g, 80% yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for Css^yBrNzO 2 Si 682.3: found 683.3.

Step 9. To a stirred mixture of TBAF (172.6 g, 660 mmol) in THF (660 mL) at rt under an atmosphere of Nz was added 5-bromo-3-[3-[(tert-butyldiphenylsllyl)oxy]-2,2-dimethylprop yl]-1-ethyl-2-[2- [(1 S)-1-methoxyethyl]pyridin-3-yl]indole (66 g, 97 mmol) in portions. The mixture was heated to 50 °C and stirred for 16 h, cooled, diluted with HzO (5 L) and extracted with EtOAc (3 x 1 .5 L). The combined organic layers were washed with brine (2 x 1 L), dried over anhydrous NazSO^ and filtered. After filtration, the filtrate was concentrated under reduced pressure. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 3-(5-bromo-1-ethyl-2-[2-[(1 S)-1- methoxyethyl]pyridin-3-yl]indol-3-yl)-2,2-dimethylpropan-1-o l (30 g, 62% yield) as a solid. LCMS (ESI): /z [M+H] calc’d for C 2 aHzgBrNzO 2 444.1 ; found 445.1 .

Intermediate 1. Alternative Synthesis through Fisher indoie Route.

Step 1. To a mixture of /-PrMgCl (2M in in THF, 0.5 L) at -10 c 'C under an atmosphere of Nz was added n-BuLi, 2.5 M in hexane (333 mL, 833 mmol) dropwise over 15 min. The mixture was stirred for 30 min at -10 c 'C then 3-bromo-2-[(1 S)-1-methoxyethyl]pyridine (180 g, 833 mmol) in THF (0.5 L) added dropwise over 30 min at -10 c C. The resulting mixture was warmed to -5 °C and stirred for 1 h, then 3,3- dimethyloxane-2,6-dione (118 g, 833 mmol) in THF (1 .2 L) was added dropwise over 30 min at -5 °G. The mixture was warmed to 0 °C and stirred for 1 .5 h, then quenched with the addition of pre-cooied 4M HCi in 1 ,4-dioxane (0.6 L) at 0 °C to adjust pH ~5 The mixture was diluted with ice-water (3 L) and extracted with EtOAc (3 x 2 5 L). The combined organic layers were dried over anhydrous NazSC , filtered, the filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 5-[2-[{1 S)-1-methoxyethyl]pyridin-3-yl]-2,2-dimethyl-5-oxopentanoic acid (87 g, 34% yield) as a solid LCMS (ESI): m/z [M+H] calc’d for C 1 5H 2 1NQ4279.2; found 280.1 .

Step 2. To a mixture of 5-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]-2,2-dimethyl-5-oxopentanoic acid (78 g, 279 mmol) in EtOH (0.78 L) at rt under an atmosphere of N 2 was added (4-bromophenyl)hydrazine HCI salt (88.7 g, 307 mmol) in portions. The mixture was heated to 85 °C and stirred for 2 h, cooied to rt, then 4M MCI in 1 ,4-dioxane (69.8 mL, 279 mmol) added dropwise. The mixture was heated to 85 °G and stirred for an additional 3 h, then concentrated under reduced pressure and the residue was dissolved in TEA (0.78 L). The mixture was heated to 60 °G and stirred for 1 .5, concentrated under reduced pressure and the residue adjusted to pH -5 with saturated NaHCO 3 , then extracted with EtGAc (3 x 1 .5 L) The combined organic layers were dried over anhydrous NaaSC , filtered, the filtrate concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 3-(5-bromo-2- [2-[(1 S)-1-methoxyethyl]pyridin-3-yl]-1H-indol-3-yl)-2,2-dimethylp ropanoic acid and ethyl (S)-3-(5-bromo- 2-(2-(1-methaxyethyl)pyridin-3-yl)-1H-indol-3-yl)-2,2-dimeth ylpropanoate (78 g, crude). LCMS (ESI): m/z [M+H] calc’d for C 2 T½BrN 2 O;3430.1 and CsaHizBrNiCb 458.1 ; found 431 .1 and 459.1 .

Step 3, To a mixture of 3-(5-bromo-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]-1H-indol-3-yl)-2,2- dimethylpropanoic acid and ethyl (S)-3-(5-bromo-2-(2-(l-methoxyethyl)pyritiin-3-yl)-1H-indol- 3-yl)-2,2- dimethylpropanoate (198 g, 459 mmol) in DMF (1 .8 L) at 0 “C under an atmosphere of N 2 was added CS2GO 3 (449 g, 1 .38 mol) in portions. Etl (215 g, 1 .38 mmol) in DMF (200 mL) was then added dropwise at 0 °C. The mixture was warmed to rt and stirred for 4 h then diluted with brine (5 L) and extracted with EtOAc (3 x 2.5 L). The combined organic layers were washed with brine (2 x 1 .5 L), dried over anhydrous NA 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give ethyl 3-(5-bromo-1-ethyl-2-[2-[(1 S)-1-methoxyethyl]pyridin-3- yi]indol-3-yl)-2,2-dimethylpropanoate (160 g, 57% yield) as a solid. LCMS (ESI): m/z [M+H] cale’d for C 2 5H 3 iBrN 2 >3486.2; found 4872.

Step 4, To a mixture of ethyl 3-(5-bromo-1-ethyl-2-[2-[(18)-1-methoxyethyl]pyridin-3-yl]in dol-3- yi)-2,2-dimethylpropanoaie (160 g, 328 mmol) in THF (1 .6 L) at 0 °C under an atmosphere of N 2 was added L1BH4 (28.6 g, 1 .3 mol). The mixture was heated to 60 °C for 16 h, cooied, and quenched with pre- cooled (0 °C) aqueous NH4CI (5 L). The mixture was extracted with EtOAc (3 x 2 L) and the combined organic layers were washed with brine (2 x 1 L), dried over anhydrous NA 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give to two atropisomers of 3-(5-bromo-1-ethyl-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)- 1H-indol-3-yl)-2,2-dlmethylpropan-1-oi (as single atropisomers) (60 g, 38% yield) and (40 g, 26% yield) both as solids. LCMS (ESI): m/z [M+H] calc'd for C 2 3H 2 9BrN 2 02444.1 ; found 445.2. intermediate 2 and intermediate 4. Synthesis of (S)-1-((S)-2 ({tert-butoxycarbonyl )amino)-3- (3-(4A5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-5-

{(triisopropylsHyi)oxy)pheriyi)propanoyi)hexahyd!Opyrsda2 :ine-3-oarboxylate

Step 1. To a mixture of {S)-methyl 2-(tert-butoxycarbonylamino)-3-(3-hydroxyphenyl)propanoate (10.0 g, 33.9 mmol) in DCM (100 rnL) was added imidazole (4.6 g, 67.8 mmol) and TiPSCl (7.8 g, 40.7 mmol). The mixture was stirred at rt overnight then diluted with DCM (200 mL) and washed with h½0 (150 mL x 3). The organic layer was dried over anhydrous NA 2 SO 4 , filtered, concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give (S)-methyl 2-(tert- butoxycarbonylamino)-3-(3-(triisopropylsllyloxy)phenyl)-prop anoate (15.0 g, 98% yield) as an oil. LCMS (ESI): m/z [M+Na] calc’d for Gi-T-UiNGsSiNa 474.3; found 474.2.

Step 2. A mixture of (S)-methyl 2-(tert-butoxycarbonylamino)-3-(3-(triisopropylsllyloxy)phen yl)- propanoate (7.5 g, 16.8 mmol), PinB2 (6.3 g, 24.9 mmol), [lr(OMe)(COD)]2 (1 .1 g, 1.7 mmol) and 4 -tert- butyl-2-(4-tert-butyl-2-pyridyi)pyrid!ne (1 .3 g, 5.0 mmol) was purged with Ar ( x3), then THF (75 L) was added and the mixture placed under an atmosphere of Ar and sealed. The mixture was heated to 80 °C and stirred for 16 h, concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give (8)-methyl 2-(fe/t-butoxycarbonylamino)-3-(3-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)-5-(triisopropylsllyloxy)phenyl)-propanoat e (7.5 g, 78% yield) as a solid. LCMS (ESI): m/z [M+Na] calc’d for CacTteBNOySINa 600.4; found 600.4; 1 H NMR (300 MHz, CD 3 OD) d 7.18 (s, 1H),

7.11 (s, 1H), 6.85 (s, 1H), 4.34 (m, 1H), 3.68 (s, 3H), 3.08 (m, 1H), 2.86 (m, 1H), 1 .41 - 1 .20 (m, 26H),

1 .20 - 1 .01 (m, 22H), 0.98 - 0.79 (m, 4H).

Step 3. To a mixture of triisopropylsllyl ( S)-2-((ierf-butoxycarbonyl)amino)-3-(3-(4, 4.5,5- tetramethyl-1 ,3,2-dioxaboroian-2-yl)-5-((triisopropylsllyl)oxy)phenyl)pro panoate (4.95 g, 6.9 mmol) in MeOH (53 mL) at 0 °C was added LIOH (840 g, 34.4 mmol) in HzO (35 L) The mixture was stirred at 0 °C for 2 h, then acidified to pH ~5 with 1 M HCI and extracted with EtOAc (250 l. x 2). The combined organic layers were washed with brine (100 L x 3), dried over anhydrous NaaSO-t, filtered and the filtrate concentrated under reduced pressure to give (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(4,4,5,5-tetramethyl - 1 ,3,2-dioxaborolan-2-yl)-5-((triisopropylsllyi)oxy)phenyl)pro panoic acid (3.7 g, 95% yield), which was used directly in the next step without further purification. LCMS (ESI): m/z [M+NFL] calc’d for CaaHsoBNOySiNhU 581 4; found 581.4.

Step 4. To a mixture of methyl (S)-hexahydropyridazine-3-carboxylate (6.48 g, 45.0 mmol) in DCM (200 mL) at 0 °C was added NMM (41 .0 g, 405 mmol), (S)-2-((tert-butoxycarbonyl)amino)-3-(3- (4,4,5,5-tetramethyM ,3,2-dioxaborolan-2-yl)-5-((triisopropylsllyl)oxy)phenyl)pro panoic acid (24 g,

42.6 mmol) in DCM (50 ml.) then HOBt (1 .21 g, 9.0 mmol) and EDCI MCI salt (12.9 g, 67 6 mmol). The mixture was warmed to rt and stirred for 16 h, then diluted with DCM (200 mL) and washed with H 2 O (3 x 150 L). The organic layer was dried over anhydrous Na2SO, filtered, the filtrate concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give methyl (S)-1- ((S)-2-((feff-butoxycarbonyl)amino)-3-(3-(4,4,5,5-tetramethy l-1 ,3,2-dioxaborolan-2-yl)-5- ((triisopropylsilyl)oxy)phenyl)propanoyl)hexahydropyridazine -3-carboxylate (22 g, 71% yield) as an oil. LCMS (ESi): m/z [M+H] calc’d for CssHeoBNaOeSi 689.4; found 690.5.

Intermediate 3. Synthesis of (SHeri-butyl S-methyl -S-CCSJ-M-methylpyrrolidine-S- carboxamido)butanoate

Step 1. To a mixture of (S)-1-(feff-butaxycarbonyl)pyrrolidine-3-carboxylic acid (2.2 g, 10.2 mmol) in DMF (10 ml) at rt was added HATU (7.8 g, 20.4 mmol) and DIPEA (5 mL). After stirring at rt for 10 min, /erf-butyl methyl-L-vaiinate (3.8g, 20.4 mmol) in DMF (10 mL) was added. The mixture was stirred at rt for 3 h, then diluted with DCM (40 mL) and H 2 O (30 mL). The aqueous and organic layers were separated, and the organic layer was washed with H 2 O (3 * 30 mL), brine (30 L), dried over anhydrous NA 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give (S)-tert-butyl 3-{((S)-1-(feff-butoxy)-3-methyl-1-oxobutan-2- yl)(methyl)carbamoyl)pyrroiidine-l -carboxylate (3.2 g, 82% yield) as an oil. LCMS (ESI): m/z [M+Na] calc ' d for CacTteNaGsNa 407.3; found 407.2.

Step 2. A mixture of (S)-te/T-butyl 3-(((S)-1-(tert-butoxy)-3-methyl-1-oxobutan-2- yl)(methyl)carbamoyl)pyrroiidine-1-carboxylate (3.2 g, 8.4 mmol) in DCM (13 mL) and TFA (1 .05 g, 9.2 mmol) was stirred at rt for 5 h. The mixture was concentrated under reduced pressure to give ( S)-tert butyl 3-methyl-2-((S)-N- methylpyrrolidine-3-carboxamido)butanoate (2.0 g, 84% yield) as an oil. LCMS (ESI): m/z [M+H] calc ' d for C 1 5H 2 8N 2 O 3 284.2; found 285.2. intermediate 5. Synthesis of !ert-butyl ((6 3 S,4S)-11-ethyl-12-(2-(CS)-1-mefhoxyethyl )pyndin- 3-yl)-10 10-dimefhyl-5 7-d!Oxo-25-((tr!!SOpropyl sllyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ! 6 6 -hexahydro-1 1 H-8-oxa

1 (5,3)-irsdoia-6(1 ,3)-pyridazirsa-2(1 ,3)-berszenacycioif ridecaphane-4-yl)earbamate.

Step 1, To a stirred mixture of 3-(5-bromo-1-ethyl-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]indol-3- yi)-2,2-dimethylpropan-1-ol (30 g, 67 mmol) and methyl (3S)-1-[(2S)-2-[(ieri-butoxycarbonyl)amino]-3-[3- (4,4,5,5-teiramethyl-1 ,3 s 2-dioxaborolan-2-yl)-5-[(triisopropylsiiyi)oxy]phenyl] propanoyl]-1 ,2-diazinane-3- carboxylate (55.8 g, 80.8 mmol) in 1 ,4-dioxane (750 ml.) at rt under an atmosphere of Ar was added NaaCO 3 (17 9 g, 168.4 mmol), Pd(DtBPF)Cl2 (4.39 g, 6.7 mmol) and H 2 O (150.00 mL) in portions. The mixture was heated to 85 °C and stirred for 3 h, cooled, diluted with H 2 O (2 L) and extracted with EtOAc (3 x 1 L). The combined organic layers were washed with brine (2 x 500 ml.), dried over anhydrous NA 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give methyl (3S)-1-[(2S)-2-[(ie / t-butoxycarbonyl)amino]-3-[3-[1- ethyl-3-(3-hydroxy-2 2-dimethylpropyl)-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]indol-5-yl]-5- [(triisopropylsl iyi)oxy]phenyl3propanoyi]-1 ,2-diazinane-3-carboxylate (50 g, 72% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for Csh rNsOeSi 927.6; found 928.8.

Step 2. To a stirred mixture of methyl (3S)-1-[(2S)-2-[(/e 7-butoxycarbonyl)amino]-3-[3-[1-ethyl-3- (3-hydroxy-2,2-dimethylpropyl)-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]indol-5-yl]-5- [{triisopropylsi iyl)oxy]phenyl]propanoyl] 1 ,2-diazinane-3-carboxylate (50 g, 54 mmol) in DCE (500 mL) at rt was added trimethyltin hydroxide (48.7 g, 269 mmol) in portion. The mixture was heated to 65 °C and stirred for 16 h, then filtered and the filter cake washed with DCM (3 x 150 mL). The filtrate was concentrated under reduced pressure to give (3S)-1-[(2S)-2-[(tert-butoxycarbonyl)amino]-3-[3-[1-ethyl-3- (3-hydroxy-2,2-dimethylpropyl)-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]indol-5-yl]-5- [(triisapropylsllyi)oxy]phenyl3propanoyi]-1 2-diazinane-3-carboxylic acid (70 g, crude), which was used directly in the next step without further purification. LCMS (ESI): m/z [M+H] calc’d for C 5 iHysNsOsSi 913 5; found 914.8.

Step 3. To a stirred mixture of (3S)-1-[(2S)-2-[(tert-butcxycarbonyl)amino]-3-[3-[1-ethyl-3- (3- hydroxy-2,2-dimethylpropyl)-2-[2-[(1 S)-1-methoxyethyl]pyridin-3-yl]lndoi-5-yl]-5-

[(triisopropylsllyi)oxy]phenyl3propanoyl]-1 ,2-diazinane-3-carboxylic acid (70 g) in DCM (5 L) at 0 °C under an atmosphere of N 2 was added DIPEA (297 g, 2.3 mol), HOST (51 .7 g, 383 mmol) and EDCI (411 g, 2 1 mol) in portions. The mixture was warmed to rt and stirred for 16 h, then diluted with DCM (1 L), washed with brine (3 x 1 L), dried over anhydrous Na2SOi and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give ierf-butyl ((6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5 ,7-dioxo-2 5 - ((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-4-yl)carbamate (36 g, 42% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C51H73N5O7S1 895.5; found 896.5.

Intermediate 8. Synthesis of tert-butyf M-[(83,145)-21-ίq€ΐo-18,18·eΐ!hΊbίH¾ίI-9,15·eΐ!q co-4- [(trnsopropylsHyl)oxy]-16-oxa-10,22,28- triazapentacyclo[18,5.2.1 A [2,6].1 A [10,14].0 A [23,27]]iionacosa-1(28),2,4,6(29),20,23(27),24-heptaen -

8-yl]carbarriate.

Step 1. This reaction was undertaken on 5-batches in parallel on the scale illustrated below.

Into a 2L round-bottom flasks each were added 5-brcmo-3-[3-[(tert-butyldiphenylsllyl)oxy]-2,2- dimethylpropyl]-1H-indole (100 g, 192 mrrsoi) and TBAF (301 .4 g, 1.15 mol) In THF (1 .15 1) at rt. The resulting mixture was heated to 50 °C and stirred for 18 h, then the mixture was concentrated under reduced pressure. The combined residues were diluted with H 2 O (5 L) and extracted with EtOAc (3 x 2 L). The combined organic layers were washed with brine (2 x 1 .5 L), dried over anhydrous NB2S04 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give 3-(5-bromo-1H-indol-3-yl)-2,2-ciimethylpropan-1-oi (310 g, crude) as a solid LCMS (ESI): m/z [M+H] calc’d for C 1 aHisBrNO 281 .0 and 283.0; found 282.1 and 284 1 .

Step 2. This reaction was undertaken on 2-batches in parallel on the scale illustrated below.

To a stirred mixture of 3-(5-bromo-1H-indol-3-yl)-2,2-dimethylpropan-1-ol (135 g, 478 mmol) and TEA (145.2 g, 1 .44 mol) in DCM (1 .3 L) at 0 °C under an atmosphere of N 2 was added AC 2 O (73.3 g, 718 mmol) and DMAP (4.68 g, 38.3 mmol) in portions. The resulting mixture was stirred for 10 min at 0 °C, then washed with H 2 O (3 x 2 L). The organic layers from each experiment were combined and washed with brine (2 x 1 L), dried over anhydrous NA 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by column chromatography to give 3-(5-bromo-1 H- indol-3- yi)-2,2-dimethylpropyl acetate (304 g, 88% yield) as a solid. 1 H NMR (400 MHz, DMSO-dfe) d 11.16 - 11.11 (rn, 1H), 7.69 (d, J= 2.0 Hz, 1H), 7.32 (d, J= 8.6 Hz, 1H), 7.19 - 7.12 (m, 2H), 3.69 (s, 2H), 2.64 (s, 2H), 2.09 (s, 3H), 0.90 is, 6H).

Step 3, This reaction was undertaken on 4-batches in parallel on the scale illustrated below.

Into a 2L round-bottom flasks were added methyl (2S)-2-[(fe/†-butoxycarbonyl)amino]-3-[3- (4,4,5,5-teiramethyl-1 ,3,2-dioxaborolan-2-yl)-5-[(triisopropylsllyl)oxy]phenyl]pro panoate (125 g, 216 mmol), 1 ,4-dioxane (1 L), H 2 O (200 mL), 3-(5-bromo-1H-indol-3-yl)-2,2-dimethylpropyl acetate (73.7 g, 227 mmol), K2CO 3 (59.8 g, 433 mmol) and PdiDtBPFJCIs (7.05 g, 10.8 mmol) at rt under an atmosphere of Ar. The resulting mixture was heated to 65 °C and stirred for 2 h, then diluted with f-fcO (10 L) and extracted with EtOAc (3 x 3 L). The combined organic layers were washed with brine (2 x 2 L), dried over anhydrous NasSCh and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by column chromatography to give methyl (2S)-3-(3-[3-[3-(acetylcxy)-2.2-dimethylpropyl]-1 H- indol-5-yl]-5-[(triiscpropylsllyi)oxy]phenyl)-2-[(terf-butox ycarbonyl)aminc]prcpanoate (500 g, 74% yield) as an oil. LCMS (ESI): m/z [M+Na] calc’d for CaaHssNaOSiNa 717.4; found 717.3

Step 4, This reaction was undertaken on 3-batchs' in parallel on the scale Illustrated below.

To a stirred mixture of methyl (2S)-3-(3-[3-[3-(acetyloxy)-2,2-dimethylpropyl]-1H- indol-5-yl]-5- [(triisopropylsllyl)oxy]phenyl)-2-[(ieri-butoxycarbonyl)amin o]propanoate (150 g, 216 mmol) and NaHCO 3 (21 .76 g, 259 mmol) in THE (1 .5 L) was added AgOTf (665 g, 259 mmol) in THE dropwise at 0 °C under an atmosphere of nitrogen la (49.3 g, 194 mmol) in THF was added dropwise over 1 h at 0 °C and the resulting mixture was stirred for an additional 10 min at 0 °C. The combined experiments were diluted with aqueous NaaSaO 3 (5 L) and extracted with EtOAc (3 x 3 L). The combined organic layers were washed with brine (2 x 1 .5 L). dried over anhydrous Na 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by column chromatography to give methyl (2S)-3- (3-[3-[3-(acetyloxy)-2,2-dimethylpropyl]-2-lodo-1H-indol-5-y l]-5-[(triisopropylsllyl)oxy]phenyl)-2-[(terf- butoxycarbonyl)amino]propanoate (420 g, 71% yield) as an oil. LCMS (ESI): m/z [M+Na] calc'd for CasHsyiNaOySiNa, 843.3; found 842.9.

Step 5. This reaction was undertaken on 3-batchos in parallel on the scale illustrated below.

To a 2L round-bottom flask were added methyl (2S)-3-(3-[3-[3-(ace†yloxy)-2,2-dimethylpropyl]-2- iodo-1H-indol-S-yl]-S- triisopropylsllylJoxy]phenyl^- tefi-butoxycarbonylJamino]propanoate (140 g, 171 mmol), MeOH (1 .4 L) and K3PO4 (108.6 g, 512 mmol) at 0 °C. The mixture was warmed to rt and stirred for 1 h, then the combined experiments were diluted with H 2 O (9 L) and extracted with EtOAc (3 x 3 L). The combined organic layers were washed with brine (2 x 2 L), dried over anhydrous NA 2 SO 4 , filtered and the filtrate was concentrated under reduced pressure to give methyl (2S)-2-[(ferf-butoxycarbonyl)amino]- 3-[3-[3-(3-hydroxy-2,2-dimethylpropyl)-2-iodo-1H-indol-5-yl] -5-[(triisopropylsllyl)oxy]phenyl]propanoate (438g, crude) as a solid. LCMS (ESI): m/z [M+Na] calc'd for CayHssINaOsSiNa 801 .3; found 801 .6.

Step 6. This reaction was undertaken on 3-batches in parallel on the scale illustrated below.

To a stirred mixture of methyl (2S)-2-[(tert-butaxycarbonyl)amino]-3-[3-[3-(3-hydroxy-2,2- dimethylpropyl)-2-iodo-1H-indol-5-yl]-5-[(triisopropylsllyl) oxy]phenyl]propanoate (146 g, 188 mmol) in THF (1 46 L) was added LiOH (22.45 g, 937 mmol) in H 2 O (937 mL) dropwise at 0 °C. The resulting mixture was warmed to rt and stirred for 1 .5 h [note: LCMS showed 15% de-TIPS product]. The mixture was acidified to pH 5 with 1 M HCi (1 ) and the combined experiments were extracted with EtOAc (3 x 3 L). The combined organic layers were washed with brine (2 x 2 L), dried over anhydrous NaaSCH, filtered and the filtrate was concentrated under reduced pressure to give (2S)-2-[(tefi-butoxycarbonyl)amino]-3-[3-[3- (3-hydroxy-2,2-dimethylpropyl)-2-iodo-1H-indol-5-yl]-5-[(tri isopropylsllyi)oxy]phenyl]propanoic acid (402 g, crude) as a sol id. LCMS (ESI): m/z [M+Na] calc’d for CssHssl^OeSiNa 787.3; found 787.6.

Step 7, To a stirred mixture of (2S)-2-[(/ert-butoxycarbonyl)amino]-3-[3-[3-(3-hydroxy-2,2- dimethylpropyl)-2-iodo-1 f-/-indol-5-yl]-5-[(iriisopropylsllyl)oxy]phenyl]propanolc acid (340 g, 445 mmol) and methyl (3S)-1 ,2-diazinane-3-carboxylate (96.1 g, 667 m ol) in DCM (3.5 L) was added NMM (225 g, 2.2 mol), EDCI (170 g, 889 mmol), HOST (12.0 g, 88.9 mmol) portionwise at 0 °C. The mixture was warmed to rt and stirred for 16 h, then washed with H 2 O (3 x 2.5 L), brine (2 x 1 L), dried over anhydrous NasSCb and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by column chromatography to give methyl (3S)-1-[(2S)-2-[(ierf-butoxycarbonyl)amino]-3-[3-[3-(3-hydro xy-2,2- dimethylpropyl)-2-iodo-l --/ indol-5-yl]-5-[{triisopropylsllyl)oxy]phenyl]propanoyi]-1 ,2-diazinane-3- carboxylate (310 g, 62% yield) as an oil. LCMS (ESI): m/z [M+H] calc'd for C 4 2H63IN4O7S1 890.4; found 890.8.

Step 8. This reaction was undertaken on 3-batches in parallel on the scale illustrated below.

To a stirred mixture of methyl (3S)-1-[(2S)-2-[(fe/f-buioxycarbonyl)amino]-3-[3-[3-(3-hydro xy-2 > 2- dimethylpropyl)-2-iodo-1H-indol-5-yl]-5-[(triisopropylsllyl) oxy]phenyl]propanoyi]-1 ,2-diazinane-3- carboxylate (85.0 g, 95.4 mmol) in THF (850 mL) each added LiOH (6.85 g, 286 mmol) in H 2 O (410 mL) dropwise at 0 °C under an atmosphere of N 2 . The mixture was stirred at 0 c C for 1 .5 h [note: LCMS showed 15% de-TIPS product], then acidified to pH 5 with 1 M HCI and the combined experiments extracted with EtOAc (3 x 2 L) The combined organic layers were washed with brine (2 x 1 .5 L), dried over anhydrous LteSC , filtered and the filtrate was concentrated under reduced pressure to give (3S)-1- [(2S)-2-[(feri-butoxycarbonyl)amino]-3-[3-[3-(3-hydroxy-2,2- dimethylpropyl)-2-iodo-1H-indol-5-yl]-5- [(triisopropylsllyl)oxy]phenyl]propanoyl]-1 ,2-diazinane-3-carboxylic acid (240 g, crude) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 4 1H61IN4O7S! 876 3; found 877.6.

Step 9, This reaction was undertaken on 2-batches in parallel on the scale illustrated below.

To a stirred mixture of (3S)-1-[(2S)-2-[(iert-butoxycarbonyl)amino]-3-[3-[3-(3-hydro xy-2,2- dimethylpropyl)-2-iodo-1H-indol-5-yl]-5-[(triisopropylsllyl) oxy]phenyl]propanoyi]-1 ,2-diazinane-3-earboxylic acid (120 g, 137 mmol) in DCM (6 L) was added DIPEA (265 g, 2.05 rnol), EDCl (394 g, 2.05 mol), HOBΪ (37 g, 274 mmol) in portions at 0 °C under an atmosphere of N 2 . The mixture was warmed to rt and stirred overnight, then the combined experiments were washed with H 2 O (3 x 6 L), brine (2 x 6 L), dried over anhydrous NaaSCb and filtered. After filtration, the filtrate was concentrated under reduced pressure. The filtrate was concentrated under reduced pressure and the residue was purified b column chromatography to give tert-butyl L ί - b,I b^I-^Io-I B,I d-aίGhqΐG^I-q,I d-aίoco- - Ίΐeorp^IeIIgOocg]- 16-oxa-10,22,28-triazapentacycloil 8.5.2.1 L [2,6].1 L [10,14].0 A [23,27]]nonacosa- 1 (26),2,4,6(29),20,23(27),24-heptaen-8-yl]carbamate (140 g, 50% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 4 1H59IN4GSSI 858.9; found 858.3.

Intermediate 7. Synthesis of (6 3 S ! 4 s )-4-amino-1 1 -ethyl-2 5 -hydroxy-1 2 -(4-

Step 1, To a mixture of 3-bromo-4-(methoxymethyl)pyridine (1 .00 g, 5.0 mmol), 4, 4,5,5- teirameihyl-2-(tetramethyl-1 ,3,2-dioxaboroian-2-yl)-1 ,3,2-dioxaboroiane (1.51 g, 5.9 mmol) and KOAc (1.21 g, 12.3 mmol) in toluene (10 mL) at rt under an atmosphere of Ar was added Pd(dppf)Cl2 (362 g, 0.5 mmol). The mixture was heated to 110 °C and stirred overnight, then concentrated under reduced pressure to give 4-(methoxymethyl)-3-(4,4,5,5-tetramethyM ,3,2-dioxaboroian-2· ylipyridine, which was used directly in the next step directly without further purification. LCM8 (ESI): m/z [M+H] calc’d for C 1 3H 2 0BNO 3 249.2; found 250.3.

Step 2. To a mixture of 4-(methoxymethyl)-3-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yi)pyridine (290 mg, 1.16 mmol), K3PO4 (371 mg, 1.75 mmol) and terf-butyl N -[(8S,14S)-21-iodo-18,18- dimethyl-9,15-dioxo-4-[(triisopropylsilyi)oxy]-16-oxa-10,22, 28- triazapentacyclo[18.52.1 L [2,6].1 L [10,14].0 A [23,27]]nonaeosa-1 (26),2,4,6(29),20,23(27),24-heptaen-8- yi]carbamate (500 mg, 0.58 mmol) in 1 ,4-dioxane (5 mL) and H 2 O (1 L) at rt under an atmosphere of Ar was added Pd(dppf)Ci2 (43 mg, 0.06 mmol). The mixture was heated to 70 °C and stirred for 2 h, then H 2 O added and the mixture extracted with EtOAc (2 x 10 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous NA 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gei column chromatography to give tert-butyl N- [(8S,14S)-21-[4-{methoxymethyl)pyridin-3-yl]-18,18-dimethyl- 9,15-dioxo-4-[(triisopropylsllyl)oxy]-16-oxa- 10,22,28-triazapentacyclo[18.5 2.1 L [2,6].1 L [10,14].0 A [23,27]]nonaeosa-1 (26), 2, 4, 6(29), 20, 23(27), 24- heptaen-8-yl]carbamate (370 mg s 74% yield) as a foam. LCM8 (ESI): m/z [M+H] calc’d for C 4 8Hs7N5C>78i 853.6; found 854.6.

Step 3. A mixture of tert-butyl N -[(8S,14S)-21-[4-(methoxymethyl)pyridin-3-yl]-1 S,18-dimethyl- 9,15-d ioxo-4-[(tri isopropylsi iyl)oxy]-16-oxa-10,22,28- triazapentacyclo[18.5.2.1 A [2,6].1 A [10,14].0 A [23,27]]nonacosa-1 (26),2,4,6{29),20,23(27),24-heptaen-8- yl]carbamate (350 g, 0.41 mmol), Cs2C03 (267 mg, 0.82 mmol) and Eti (128 mg, 0.82 mmol) in DMF (4 ml.) was stirred at 35 °C overnight. H 2 O was added and the mixture was extracted with EtOAc (2 x 15 l.). The combined organic layers were washed with brine (15 ml..), dried over anhydrous NteSC and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give /erf-butyl N- [(8S,14S)-22-ethyl-21-[4-(methoxymethyl)pyridin-3-yl]-18,18- dimethyl-9,15-dioxo-4-[(triisopropylsilyl)oxy]-10-oxa-10,22, 28- triazapentacyclo[18.5.2.1 A [2,6].1 A [10,14].0 A [23,27]]nonacosa-1 (26),2,4,6(29),20,23(27),24-heptaen-8-yl] carbamate (350 mg, 97% yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for C 5 oHnNsOSi 881 .5; found 882.6.

Step 4, A mixture of tert- butyl N- [(8S l 14S)-22-ethyl-21-[4-(methoxymethyl)pyridin-3-yl]-18 ! 18- dimethyl-9,15-dioxo-4-[(triisopropylsllyl)oxy] -16-oxa-10,22,28- triazapentacyclo[18.5.2.1 A [2,6].T A [10,14].0 A [23,27]]nonacosa-1 (26),2,4,6(29),20,23(27),24-heptaen-8-yl] carbamate (350 mg, 0.4 mmol) and 1 M TBAF in THF (0.48 mL, 0.480 mmol) in THF (3 mL) at 0 “C under an atmosphere of Ar was stirred for 1 h. The mixture was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give tert- butyl N -[(8S,14S)-22-ethyl-4- hydroxy-21-[4- (methoxymeihyl)pyridin-3-yl]-18,18-dimethyl-9,15-dioxo-16-ox a-10,22,28- triazapentacyclo[18.52.1 A [2,6].1 L [10,14].0 A [23,27]]nonacosa-1 (26), 2,4, 6(29),20, 23(27), 24-heptaen-8- yl]earbamate (230 mg, 80% yield) as an oil. LCMS (ESI): m/z [M+H] ealc’d for C 4 1H51 N5O77254; found 726.6.

Step 5, To a mixture of terf-butyl N -[(8S,14S)-22-ethyl-4-hydroxy-21-[4- (methoxymethyl)pyridin- 3-yl]-18,18-dimethyl-9, 15-dioxo-16-oxa-10,22,28- triazapentacyclo[18.52.1 A [2,6].1 L [10,14].0 A [23,27]]nonacosa-1 (26), 2,4, 6(29),20, 23(27), 24-heptaen-8- yi]earbamate (200 mg, 0.28 mmol) in 1 ,4-dioxane (2 mL) at 0 c, C under an atmosphere of Ar was added 4M MCI In 1 ,4-dioxane (2 mL, 8 mmol). The mixture was allowed to warm to rt and was stirred overnight, then concentrated under reduced pressure to give (6 3 S,4 s )-4-amino-1 1 -ethyl-2 5 -hydroxy-1 2' -(4- (methoxymethyl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H-8-oxa-1 (5,3)-indola-6(1 ,3)- pyridazina-2(1 ,3)-benzenacycloundecaphane-5,7-dione (200 mg). LCMS (ESI): m/z [M+H] calc’d for C 3 6H43N5O5 625.3; found 626 5. Intermediate 8. Synthesis of tert- butyl ((6 3 S,4S,2)-1 1 -ethyl -1 2 -(2-((S)-1- rrsethoxyethyl )pyrsd!ri-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 f -8-oxa-2(42)- fhiazoia-1(5,3)- doia-6(1,3)-pyridazirsacycioundecapharie-4-yl)carbamate

Step 1, To a solution of methyl (2S)-3-(4-bromo-1 ,3-thiazol-2-yl)-2- (terf- butoxycarbonyl)a ino]propanoate (110 g, 301 .2 mmol) in THF (500 mL) and H 2 O (200 ml.) at room temperature was added LiOH (21 .64 g, 903.6 mmol). The solution was stirred for 1 h and was then concentrated under reduced pressure. The residue was adjusted to pH 6 with 1 M MCI and then extracted with DCM (3 x 500 mL) The combined organic layers were, dried over NaaSO^ filtered, and concentrated under reduced pressure to give (S)-3-(4-bromothiazol-2-yl)-2-((tert-butoxycarbonyl)amino)pr opanoic acid (108 g, crude). LCMS (ESI): m/z [M+H] calc’d for C H Br^C S 351 .0; found 351 .0.

Step 2. To a solution of (S)-3-(4-bromothiazol-2-yl)-2-((feff-butoxycarbonyl)amino)pr opanoic acid (70 g, 199.3 mmol) in DCM (500 mL) at 0 °C was added methyl (3S)-1 ,2-diazinane-3-carboxylate bis(trifluoroacetic acid) salt (111 .28 g, 298.96 mmol), NMM (219.12 ml. 1993.0 mmol), EDCI (76.41 g, 398.6 mmol) and HOBt (5.39 g, 39.89 mmol). The solution was warmed to room temperature and stirred for 1 h. The reaction was then quenched with H 2 O (500 mL) and was extracted with EtOAc (3 x 500 mL). The combined organic layers were dried over Na2SO„ filtered, and concentrated under reduced pressured. The residue was purified by silica gel column chromatography to give methyl (S)-1-((S)-3-(4- bromothiazol-2-yl)-2-((terf-butoxycarbonyl)amino)propanoyi)h exahydropyridazine-3-carboxylate (88.1 g, 93% yield). LCMS (ESI): m/z [M+H] calc’d for Cr/HKsBrtTiOsS 477.1 ; found 477.1 .

Step 3. To a solution of 3-(5-bromo-1-eihyl-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1H -indol-3-yl)- 2,2-dlmeihylpropan-1-ol (60 g, 134.7 mmol) in toluene (500 mL) at room temperature was added bis(pinacoiato)diboron (51.31 g, 202.1 mmol), Pd(dppf)Cl2 (9.86 g, 13.4 mmol), and KOAc (26.44 g, 269 mmol). The reaction mixture was then heated to 90 °C and stirred for 2 h. The reaction solution was then cooled to room temperature and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give (S)-3-(1-ethyl-2-(2-(1-methoxyethyl)pyridin-3-yl)-5-(4, 4,5,5- tetramethyl-1 ,3,2-dioxaboroian-2-yl)-1H-indol-3-yl)-2.2-dimethylpropan-1- ol (80.6 g, 94% yield). LCMS (ESI): m/z. [M+H] calc'd for C 2 9H42BN 2 O4493.32; found 493.3.

Step 4. To a solution of (S)-3-(1-ethyl-2-(2-(1-mothoxyethyl)pyridin-3-yl)-5-(4,4,5 J 5-tetramethyl- 1 ,3,2-dioxaboroian-2-yl)-1H-indol-3-yl)-2 2-dimethylpropan-1-ol (30 g, 60.9 mmol) in toluene (600 ml.), dioxane (200 mL), and H 2 O (200 mL) at room temperature was added methyl (S)-1-((S)-3-(4- bromolhiazoi-2-yl)-2-((fert-butoxycarbonyl)amino)propanoyi)h exahydropyhdazine-3-earboxylale (43.82 g, 91.4mmol) s K3PO4 (32.23 g, 152.3 mmol) and Pd(dppf)Cl2 (8.91 g, 12.18 mmol). The resulting solution was heated to 70 °C and stirred overnight. The reaction mixture was then cooled to room temperature and was quenched with H 2 O (200 ml.). The mixture was extracted with EtOAc and the combined organic layers were dried over NA 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give methyl (S)-1-((S)-2-((iert-butoxycarbonyl)amino)-3- (4-(1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-met hoxyethyl)pyridin-3-yl)-1H-indol-5-yl)thiazol-2- yi)propanoyl)hexahydropyridazine-3-carboxylate (39.7 g, 85% yield). LCMS (ESI): m/z [M+H] calc’d for CwHssNeOS 763.4; found 763.3.

Step 5, To a solution of methyl (S)-l -((S)-2-((iert-butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3- hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin -3-yl)-l H-indol-5-yl)thiazol-2- yl)propanoyl)hexahydropyridazine-3-carboxylate (39.7 g, 52.0 mmol) in THE (400 mL) and HsO (100 L) at room temperature was added LiOHeH 2 O (3.74 g, 156.2 m ol). The mixture was stirred for 1 .5 h and was then concentrated under reduced pressure. The residue was acidified to pH 6 with 1 M HCl and extracted with DGM (3 x 1000 mL). The combined organic layers were dried over Na?S04, filtered, and concentrated under reduced pressure to give (S)-1-((S)-2-((tert-butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3 - hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin -3-yl)-1 f7-indol-5-yl)thiazoi-2- yi)propanoyl)hexahydropyridazine-3-carboxylic acid (37.9 g, crude). LCMS (ESI): m/z [M+H] calc'd for C 3 9H53N6O7S 749.4; found 749.4.

Step 6, To a solution of (S)-1-((S)-2-((tert-butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3 -hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1H-in dol-5-yl)thiazol-2- yi)propanoyl)hexahydropyridazine-3-carboxylic acid (37.9 g, 50.6 mmol ), HOBt (34.19 g, 253.0 mmol) and DIPEA (284.4 mL, 1518 mmol) in DCM (4 L) at 0 c 'C was added EDCi (271 .63 g, 1418.9 mmol). The resulting mixture was warmed to room temperature and stirred overnight. The reaction mixture was then quenched with H 2 O and washed with 1 M HCI (4 x 1 L). The organic layer was separated and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give tort-butyl ((6 3 S,4S,Z)-1 ’-ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5 ,7-dioxo- 8 1 ,6 2 ,6 3 ,6 4 ,8 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-thiazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4- yi)carbamate (30 g, 81% yield). LCMS (ESI): m/z [M+H] calc'd for CsgHsiNsOsS 731 .4; found 731 .3.

Step 7, To a solution of tort-butyl ((8 3 S,4S,Z)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)- 10,1 O-dimethyl-SJ-dioxo-e 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 /7-8-oxa-2(4,2)-thiazola-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-4-yl)carbamate (6 g, 8.21 mmol) In DCM (60 mL) at 0 °C was added TFA (30 mL). The mixture was stirred for 1 h and was then concentrated under reduced pressure to give (6 3 S,4S,Z)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 - hexahydro-1 1 W-8-oxa-2(4,2)-thiazoia-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-5,7-dione (7.0 g, crude). LCMS (ESI): m/z [M+H] calc'd for C 3 H«N 6 04S 631 .3; found: 630.3. intermediate 9. Synthesis of CS)-3-bromo-5-iode-2- (1-methoxyethyl) pyridine.

Intermediate 9,

Step 1, To a stirred solution of 3-bromo-2-[(1 S)-1-methoxyethyl]pyridine (80.00 g, 370.24 mmol, 1 .00 equiv) and bis(pinacolato)diboron (141 .03 g, 555.3 mmol, 1 .50 equiv) in THF (320 mL) was added dtbpy (14.91 g, 55.5 mmol) and chloro(1 ,5-cyclooctadiene)iridium(l) dimer (7.46 g, 11 .1 mmol) under argon atmosphere. The resulting mixture was stirred for 16 h at 75 °C under argon atmosphere. The mixture was concentrated under reduced pressure. The resulting mixture was dissolved in EtOAc (200 mL) and the mixture was adjusted to pH 10 with NaaCO 3 (40 g) and NaOH (10 g) (mass 4:1) In water (600 mL). The aqueous layer was extracted with EtOAc (SOOmL). The aqueous phase was acidified to pH = 6 with HCi (6 N) to precipitate the desired solid to afford 5-bromo-6-[(1 S)-l-me†hoxyethyl]pyridin-3-ylboronic acid (50g, 52.0% yield) as a light-yellow solid. LCMS (ESI): m/z [M+H] calc’d for CsHnBBrNO 3 259.0: found 260.0.

Step 2. To a stirred solution of 5-bromo-6-[(1 S)-1-methoxyethyl]pyridin-3-ylboronic acid (23.00 g, 88.5 mmol) in ACN (230 mL) were added NIS (49.78 g, 221 .2 mmol) at room temperature under argon atmosphere . The resulting mixture was stirred for overnight at 80 °C under argon atmosphere. The resulting mixture was concentrated under reduced pressure. The resulting mixture was dissolved in DCM (2.1 L) and washed with NaaSaOa (3 x 500 L). The organic layer was dried over anhydrous NaaSOc After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford ( S)-3-bromo-5-iodo-2-( 1-methoxyethyl)pyridine (20 g, 66.0% yield). LCMS (ESi): m/z [M+H] calc ' d for CsHgBrlNO 340.9; found 341 .7.

Intermediate 10, Synthesis of tert-butyl ((6 3 S,4S,2)-41-ethyM 2 -(2-({S)-1”!iiethoxyethyf)-5-C 4 - ^b^I Irq¾2ϊh-1-nI^p^hAgI)-1O ! 1ϋ^™b^K5 ! 7-$!qco-6 1 ,6 2 ,d 3 ! 6 4 ,6 5 ,6 6 -ΐΊ6C0ΐΊg<I?Ό-1 1 ί·#-8-o 3- 2{4,2)-thsaaoia-1(5,3)-indola-6(1 ! 3)-pyrsdaasnacycloi!rsdecapharse-4-yl)carbamate Step 1. Into a 3L 3-necked round-bottom flask purged and maintained with an inert atmosphere of argon, was placed 3-bromo-5-iodo-2-[(1 S)-1-methoxyethyl]pyridine (147' g, 429.8 mmol) benzyl piperazine-1-carboxylate (94.89 g, 429.8 mmol), PdtOAc)? (4.83 g, 21 .4 m ol), BlNAP (5.35 g, 8.6 mmol), CsiiCO 3 (350.14 g, 1074.6 mmol), toluene (1 L). The resulting solution was stirred for overnight at 100 c C in an oil bath. The reaction mixture was cooled to 25 “C after reaction completed. The resulting mixture was concentrated under reduced pressure. The residue was applied onto a silica gel column with ethyl acetate/hexane (1 :1 ). Removal of solvent under reduced pressure gave benzyl (S)-4-(5-bromo-6-(1- methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (135 g, 65.1% yield) as a dark yellow solid. LCMS (ESI): m/z [M+H] calc ' d for Ciol-biBrNsC 433.1 ; found 434.1 .

Step 2. Into a 3-L 3-necked round-bottom flask purged and maintained wiih an inert atmosphere of argon, was placed benzyl 4-[5-bromc-6-[(1 S)-1-methoxyethyl]pyridin-3-yl]piperazine-1-carboxylate (135 g, 310.8 mmol), bis(pinacolato)diboron (86 82 g, 341 .9 mmol), Pd(dppf)Ci2 (22 74 g, 31 .0 mmol), KOAc (76.26 g, 777.5 m ol), Toluene (1 L) The resulting solution was stirred for 2 days at 90 °C in an oil bath. The reaction mixture was cooled to 25 °C. The resulting mixture was concentrated under vacuum. The residue was applied onto a neutral alumina column with ethyl acetate/hexane (1 :3) Removal of solvent under reduced pressure gave benzyl (S)-4-(6-(1-methoxyethyl)-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxabGrolan- 2-yl)pyridin-3-yl)piperazine-1-carboxylate (167 g, crude) as a dark yellow solid. LCMS (ESI): mi?.. [M+H] calc'd for C 2 sHaeBNaOs 481 .3; found 482.1 .

Step 3. Into a 3-L 3-necked round-bottom flask purged and maintained with an Inert atmosphere of argon, was placed (S)-4-(6-(1-methoxyethyl)-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaboroian-2-yl)pyridin-3- yl)piperazine-1-carboxylate (167 g, 346.9 mmol), 5-bromo-3-[3-[(tert-butyldiphenylsilyl)oxy]-2,2- dimethylpropyl]-2-lodo-1H-lndoie (224.27 g, 346.9 mmol), Pd(dppf)Cl2 (25.38 g, 34.6 mmol), dloxane (600 ml.), H 2 O (200 mL), K3PO4 (184.09 g, 867.2 mmol), Toluene (200 mL) The resulting solution was stirred for overnight at 70 °C in an oil bath. The reaction mixture was cooled to 25 °C after reaction completed. The resulting mixture was concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/hexane (1 :1 ). Removal of solvent under reduced pressure gave benzyl (S)-4-(5-(5- bromo-3-(3-((fer/-butyldiphenylsllyl)oxy)-2,2-dimethylpropyl )-1 7-indol-2-yl)-6-(1-methoxyethyl)pyridin-3- yl)piperazine-1-carboxylate (146 g, 48.1% yield) as a yellow solid. LCMS (ESI): m/z [M+H] calc ' d for C 4 9H57BrN40 4 Si 872.3; found 873.3.

Step 4, To a stirred mixture of benzyl (S)-4-(5-(5-bromo-3-(3-((tert-butyldiphenylsllyl)oxy)-2,2- dimethylpropyl)-1 H- indol-2-yl)-6-(1-methoxyethyl)pyridin-3-yl)piperazine-1-carb oxylate (146 g, 167.0 mmol) and CS2CO 3 (163.28 g, 501 .1 mmol) in DMF (1200 L) was added C 2 H5! (52.11 g, 334.0 mmol) in portions at 0 °C under Nz atmosphere. The final reaction mixture was stirred at 25 °C for 12 h. Desired product could be detected by LCMS. The resulting mixture was diluted with EA (1 L) and washed with brine (3 x 1 5L). The organic layers were dried over anhydrous NazSC After filtration, the filtrate was concentrated under reduced pressure to give benzyl (S)-4-(5-(5-bromo-3-(3-((itert-butyldiphenylsilyl)oxy)-

2.2-dimethylpropyl)-1-ethyl-1H-indol-2-yl)-6-(1-methoxyet hyl)pyridin-3-yl)piperazine-1-carboxylate (143 g, crude) as a yellow solid that was used directly for next step without further purification. LCMS (ESI): m/z [M+H] calc’d for C 5 iHeiBr^CbSi 900 4; found 901 .4.

Step 5, To a stirred mixture of benzyl benzyl (S)-4-(5-(5-bromo-3-(3-((tert-butyldiphenylsllyl)oxy)-

2.2-dlmethylpropyl)-1-ethyl-1H-indol-2-yl)-6-(1-methoxyet hyl)pyridin-3-yl)piperazine-1-carboxylate (143 g, 158.5 mmol) in DMF (1250 mL) was added CsF (72.24 g, 475.5 mmol). Then the reaction mixture was stirred at 60 °C for 2 days under Nz atmosphere. Desired product could be detected by LCMS. The resulting mixture was diluted with EA (1 L) and washed with brine (3 x 1 L). Then the organic phase was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/3) to afford two atropisomers of benzyl (S)-4-(5-(5-bromo-1-ethyl-3-(3-hydroxy-2,2- dlmethylpropyl)-1H-indol-2-yl)-6-(1-me†hoxyethyl)pyridin-3 -yl)piperazine-1-carboxylate A (38 g, 36% yield, RT = 1 .677 min in 3 min LCMS(0.1% FA)) and B (34 g, 34% yield, RT = 1 .578 min In 3 min LCMS(0.1 % FA)) both as yellow solid. LCMS (ESI): m/z [M+H] calc'd for C 3 sH43BrN 4 0 4 663.2; found 662 2.

Step 6. Into a 500-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed benzyl (S)-4-(5-(5-bromo-1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)- 1H-indol-2-yl)-6-(1-methoxyethyl)pyridin-3-yl)piperazine-1-c arboxylate A (14 g, 21 .1 mmol), bis(pinacolato)diboron (5.89 g, 23.21 mmol), PdidppfjClz (1 .54 g, 2.1 mmol), KOAc (5.18 g, 52.7 mmol), Toluene (150 mL). The resulting solution was stirred for 5 h at 90 °C in an oil bath. The reaction mixture was cooled to 25 °C. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/3) to give benzyl (S)-4-(5-(1-ethyl-3-(3-hydroxy- 2,2-dimethylpropyl)-5-(4,4 > 5,5-teiramethyl-1 ,3,2-dioxaborolan-2-yl)-1H-indol-2-yl)-6-(1- methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (12 g, 76.0% yield) as a yellow solid. LCMS (ESI): m/z [M+H] calc’d for C 4 1H55BN4O6710.4; found 711 .3

Step 7. Into a 250-rrsL round-bottom flask purged and maintained with an inert atmosphere of argon, was placed benzyl (S)-4-(5-(1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-5-(4,4,5, 5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)-1H-indol-2-yl)-6-(1-methoxyethyl)pyridin- 3-yl)piperazine-1-carboxylate (10.8 g, 15.2 mmol), methyl (3S)-1-[(2S)-3-(4-bromo-1 ,3-thiazol-2-yl)-2-[(feri-butoxycarbonyl)amino]propanoyl]-1 ,2- diazinane-3-carboxylate (7.98 g, 16.7 mmol), Pd(dtbpf)Ci2 (0.99 g, 1 .52 mmol), K3PO4 (8.06 g, 37.9 mmol), Toluene (60 rrsL), dloxane (20 ml..), H 2 O (20 ml..). The resulting solution was stirred for 3 h at 70 °C in an oil bath. The reaction mixture was cooled to 25 °C. The resulting solution was extracted with EtOAc (2 x 50 ml) and concentrated under reduced pressure. The residue was applied onto a silica gel column with ethyl acetate/hexane (10:1 ). Removal of solvent to give methyl (S)-1-((S)-3-(4-(2-(5-(4- ((benzyloxy)carbonyl)piperazin-1-yl)-2-((S)-1-methoxyethyl)p yridin-3-yl)-1-ethyl-3-(3-hydroxy-2,2- dimethylpropyl) -1H-indol-S-y thiazol^-yl^-^iert-butoxycarbonyljaminoJpropanoy hexahydropyridazinG- 3-carboxylate (8 g, 50.9% yield) as a yellow solid. LCMS (ESI): m/z [M+H] calc ' d for C 5 aHssNeOsS 980.5: found 980.9.

Step 8, To a stirred mixture of methyl (S)-l -((S)-3-(4-(2-(5-(4-((benzyloxy)carbonyl)piperazin-1- yi)-2-((S)-l -methoxyethyl)pyridin-3-yl)-l -ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-1H-indol-5-yl)thiazo i-2-yl)- 2-((tert-butoxycarbonyl)amino)propanoyi)hexahydropyridazine- 3-carboxylate (12 g, 12.23 m ol) in THF (100 mL)/H 2 O (100 mL) was added LiOH (2.45 g, 61 .1 m ol) under N 2 atmosphere and the resulting mixture was stirred for 2 h at 25 °C. Desired product could be detected by LCMS. THF was concentrated under reduced pressure. The pH of aqueous phase was acidified to 5 with HCL (1 N) at 0 ,3 G. The aqueous layer was extracted with DCM (3 x 100ml). The organic phase was concentrated under reduced pressure to give (S)-1-((S)-3-(4-(2-(5-(4-((benzyloxy)carbonyl)piperazin-1-yl )-2-((S)-1- methoxyethyl)pyridin-3-yl)-1-ethyl-3-(3-hydroxy-2,2-dimethyl propyl)-1H-indol-5-yl)thiazoi-2-yl)-2-((tert- butoxycarbonyl)amino)propanoyi)hexahydropyridazine-3-carboxy lic acid (10 g, 84.5% yield) as a light yellow solid. LCMS (ESI): m/z [M+H] calc d for C 5 iHeeNsCbS 966.5; found 967.0.

Step 9. Into a 3-L round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed (S)-1-({S)-3-(4-(2-(5-(4-((benzyloxy)carbonyl)piperazin-1-yl )-2-((S)-1- methoxyethyl)pyridin-3-yl)-1-ethyl-3-(3-hydroxy-2,2-dimethyl propyl)-1H-indol-5-yl)thlazol-2-yl)-2-((tert- butoxycarbonyl)amino)propanoyi)hexahydropyridazine-3-carboxy lic acid (18 g, 18.61 mmol), ACN (1.8 L), DlEA (96.21 g, 744.4 mmol), EDCI (107.03 g, 558.3 mmol), HOST (25.15 g, 186.1 mmol). The resulting solution was stirred for overnight at 25 °C. The resulting mixture was concentrated under reduced pressure after reaction completed. The resulting solution was diluted with DCM (1 L). The resulting mixture was washed with HCI (3 x 1 L, 1 N aqueous). The resulting mixture was washed with water (3 x 1 L). Then the organic layer was concentrated, the residue was applied onto a silica gel column with ethyl acetate/hexane (1 :1 ). Removal of solvent under reduced pressure gave benzyl 4-(5-((6 3 S,4S,Z)-4-((terf- butoxycarbonyl)amino)-1 1 -ethyl-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 , 6 s , 6 6 -hexahydro-1 1 H-8-oxa-2(4, 2)- thiazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-1 2 -yl)-6-((S)-1-methoxyethyl)pyridin-3- yl)piperazine-1-carboxylate (10.4 g, 54.8% yield) as a light yellow solid. LCMS (ESI): rn/z [M+H] calc’d tor C5IH 64 N 8 08S 948.5; found 949.3. Step 10. into a 250-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed benzyl 4-(5-((6 3 S > 4S,Z)-4-((ierf-butoxycarbonyl)amino)-1 , -ethyl-10,10-dimethyl-5,7- dioxo-6 1 ,6 2 ,6 3 ,6' ! ,6 5 ,6 6 -hexahydrG-1 , /7-8-oxa-2(4,2)-thiaz.oia-1 (5,3)-indola-6{1 ,3)- pyridazinacycloundecaphane-1 2 -yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-c arboxylate (10.40 g, 10.9 mmol), Pd(OH) 2 /C (5 g, 46.9 mmol), MeOH (100 mb). The resulting solution was stirred for 3 h at 25 °C under 2 atm hb atmosphere. The solids were filtered out and the filter cake was washed with MeOH (3 x 100 mL). Then combined organic phase was concentrated under reduced pressure to give tert-butyl ((6 3 S,4S,Z)-1 /-ethyl-1 2 -(2-((S)-1-methoxyethyl)-5-(piperazin-1-yl)pyridin-3-y l)-10,10-dimethyl-5,7-dioxo- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-thiazoia-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4- yl)carbamate (8.5 g, 90.4% yield) as a light yellow solid. LGMS (ESI): m/z [M+H] calc’d for C 4 3Hs8Ns06S 814.4; found 815.3.

Step 11. Into a 1000-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed tert-butyl ((6 3 S,4S,2)-1 7-ethyl-1 2 -(2-((S)-1-methoxyethyl)-5-(piperazin-1-yl)pyridin-3- yi)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,8 4 ,8 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-thiazola-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-4-yl)carbamate (8.5 g, 10.4 mmol), MeOH (100 mL), AcOH (1 .88 g, 31 .2 mmol) and stirred for 15 mins. Then HCHO (1 .88 g, 23.15 mmol, 37% aqueous solution) and NaBHgCN (788 mg, 12.5 mmol) was added at 25 °C. The resulting solution was stirred for 3 h at 25 c C. The resulting mixture was quenched with 100 mL water and concentrated under reduced pressure to remove MeOH. The resulting solution was diluted with 300 mL of DCM. The resulting mixture was washed with water (3 x 100 mL). Removal of solvent gave tert- butyl ((6 3 S,4S,2)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo- 6' > 6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 W-8-oxa-2(4,2)- thiazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)carbamate (8.2 g, 90.1 % yield) as a yellow solid. LCMS (ESI): m/z [M+H] calc d for C 4 4HsoNsOsS 828.4; found 829.3.

Example A11. Synthesis of methyl (3S)-3-{[(1 S)-1-{[(8S,14S)-22-ethyl-4-hydroxy-21-[4- {methoxymethyl )pyridirs-3-yl]-18,18-dimethyl-9,15-d!Oxo-16 Oxa-10,22,28-

Step 1. To a mixture of tert-butyl N -methyl-N- ((S)-pyrrolidine-3-carbonyl)-L-valinate (500 mg, 1 .8 mmol) and TEA (356 mg, 3.5 mmol) in DCM (10 rnL) at 0 °C was added methyl carbonochloridate (199 mg, 2.1 mmol) dropwise. The mixture was aliowed to warm to rt and was stirred for 12 then concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give methyl (S)-3-(((S)-1-(tert-butoxy)-3-methyl-1-oxobutan-2-yl)(methyl )earbamoyi)pyrrolidine-1-carboxylate (550 mg, 82%) as an oil. LCMS (ESI): m/z [M+H] calc’d for C 1 7H 3 0N 2 O5342.2; found 343.2.

Step 2. A mixture of methyl (S)-3-(((S)-1-(/e/7-butoxy)-3-methyl-1-oxobutan-2- yl)(methyl)carbamoyl)pyrroiidine-1-carboxylate (500 mg, 1 .46 mmol), DCM (8 mL) and TFA (2 mL) was stirred at rt for 3 h. The mixture was concentrated under reduced pressure with azeotropic removal of HjO using toluene (5 mL) to give N- ((S)-1-(methoxycarbonyl)pyrroiidine-3-carbonyl)-N -methyl-L-valine (400 mg) as an oil. LCMS (ESI): m/z [M+H] calc’d for C 1 3H 2 2N 2 O5286.2; found 287.2.

Step 3. To a mixture of (6 3 S,4S)-4-amino-1 '-ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)- 10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione (80 g, 0.13 mmol), N -((S)-1-(methoxycarbanyl)pyrrolidine-3- carbonyl)-N- methyl-L-valine (55 mg, 0.19 mmol) and DIPEA (165 mg, 1 .3 m ol) in DMF (2 mL) at 0 °C was added COMU (77 mg, 0.18 mmol). The mixture was stirred at 0 °G for 2 h, then concentrated under reduced pressure and the residue was purified by prep-HPLC to give methyl (38)-3-{[(1 S)-1-{[(8S,14S)- 22-ethyl-4-hydroxy-21-[4-(methoxymethyl)pyridin-3-yl]-18,18- dlmethyl-9,15-dloxo-16-oxa-1 G,22,28- triazapentacyclo[18 5.2.1 2 , e 1 1Q , 1 4Q 23 , 27 ]nonacosa-1 (28), 2,4, 8(29),20, 23(27), 24-heptaen-8-yl]carbamoyl)- 2-methylpropyl3(methyl)carbamoyi)pyrrolidine-1-carboxylate (51 g, 45% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for C 4 9H63N7O9893.5; found 894.7; 1 H NMR (400 MHz, DMSO-flfe) d 9.33 (s, 1H), 8.88 - 8.66 (m, 2H), 8.62 (s, 1H), 8.17 - 8.06 (m, 1H), 7.92 (d, 8.7 Hz, 1H), 7.79 - 7.68 (m, 1H), 7.65 - 7.49

(m, 2H), 7.21 - 7.11 (m, 1H), 7.01 (d, J = 11 .8 Hz, 1H), 6.71 - 6.40 (m, 1H), 5.54 - 5.30 (m, 1H), 5 28 - 4.99 (m, 1H), 4 87 - 4.56 (m, 1H), 4.46 - 4.21 (m, 3H), 4.11 - 3 89 (m, 3H), 3.70 (s, 1H), 3.65 - 3.59 (m, 4H), 3.35 (s, 2H), 3.24 (s, 2H), 3.18 - 3.07 (s, 1H), 3.00 - 2.58 (m, 8H), 2.22 - 2.01 (m, 4H), 1 .81 (d, J - 11.4 Hz, 2H), 1.72 - 1.42 (m, 2H), 1.15 - 0.64 (m, 13H), 0.43 (d, J = 16.4 Hz, 3H).

Example A17. Synthesis of (2S)-M-[(85 ! 14S)-22-ethyl -4-hydroxy-21-[4- (methoxymethyl)pyrldirs-3-yl]-18,18-dimethyl-9,15-d!Oxo-16 Oxa-10,22,28- friazapentacy o[18.5.2.1 2 , e .1 10 s 14 .0 23 , 27 ]nonacosa-1 (26), 2,· 4,6(29), 20,23(27), 24-heptaen-8-yl]-2-{1-

Step 1, A mixture of ie/t-butyl (2S)-3-methyl-2-[W-methyl-1-(3S) -pyrroiidin-3- yiformamido]butanoate (290 mg, 1 .0 mmol) and ethyl formate (755 g, 10.2 mmol) was heated to 60 °C and stirred for 12 h. The mixture was concentrated under reduced pressure to give tert- butyl (2S)-2-[1 [(3S)-1-formylpyrrolidin-3-yl]-N -methylformamido]-3-methylbutanoate (300 g, 85% yield) as a solid. LCMS (ESI): m/z [M+H-tBu] calc ' d for C 1 2H 2 0N 2 O4256.1 ; found 257.2.

Step 2, To a mixture of terf-butyl (2S)-2-[1-[(3S)-1-formylpyrrolidin-3-yl]-W-methylformamido]- 3- methylbutanoate (290 mg, 0.93 mmol) in DCM (3 mL) at rt was added TFA (1 mL). The mixture was stirred at rt for 2 h, then concentrated under reduced pressure to give (2S)-2-[1-[(3S)-1-formylpyrroildln-3- yi]-W-methylformamido]-3-methylbutanoic acid (260 mg, 98%) as an oil. LCMS (ESI): m/z [M+H] calc d for C 1 2H 2 0N 2 O4256.1 ; found 257.2.

Step 3, To a mixture of (6 3 S,4S)-4-amino-1 1 -eLhyl4 ¾ -hyd!Oxy-1 2 -(4-(rnethoxymethyl)pyridin-3-yl)- 10,1 G-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione (60 mg, 0 1 mmol), 2,6-dimethylpyridine (15.4 mg, 0.14 mmol) and (2S)-2-[1-[(3S)-1-formylpyrrolidin-3-yl]-W-methylformamido]- 3-methylbutanoic acid (37 mg, 0.14 mmol) in MeCN (2 mL) at 0 °C under an atmosphere of N 2 was added COMU (62 mg, 0.14 mmol). The mixture was stirred at 0 °C for 12 h, then concentrated under reduced pressure and the residue was purified by prep-HPLC to give (2S)-N -[(88,14S)-22-ethyl-4-hydroxy-21-[4-(methoxymethyl)pyridin-3 -yl]-18,18- dimethyl-9,15-dioxo-16-oxa-10,22,28-triazapentacyclo[18.5.2. 1 2 , 6 .1 10 , 14 .0 23 , 27 ]nonacosa- 1 (26),2,4,6(29),20,23(27),24-heptaen-8-yl3-2-{1-[(3¾-1-formy lpyrrolidin-3-yl]-N-methylformamido)-3- methylbutanamide (35 mg, 42%) as a solid. LCMS (ESI): m/z [M+H] calc'd for C 4 SHSIN70S 863.5; found 864.5; 1 H NMR (400 MHz, DMSO- /6) d 8.79 - 8.61 (m, 2H), 8.51 (d, J- 7.8 Hz, 3H), 8.31 - 8.09 (m, 1H), 7.93 (s, 1H), 7.68 - 748 (m, 3H), 7.25 - 6.97 (m, 2H), 6.71 - 6.43 (m, 1H), 5.40 (d, J = 24.8 Hz, 1H), 5.22 (s, 1H), 4.86 - 4.34 (m, 1H), 4.23 (t, J= 13.8 Hz, 3H), 4.12 - 3.84 (m, 3H), 3.83 - 3 54 (m, 4H), 3.22 (d, J = 1.7 Hz, 2H), 3.09 (d, J == 143 Hz, 1H), 3.01 - 2.92 (m, 1H), 2.99 - 2.93 (m, 2H), 2.92 - 2.65 (m, 5H), 2.07 (d, J = 12.2 Hz, 4H), 1.80 (s, 1H), 1.74 - 1.48 (m, 2H), 1.08 (t, J = 7.1 Hz, 2H), 1.03 - 0.54 (m, 12H), 043 (d, J = 16.2 Hz, 3H).

Example A6. Synthesis of (2S)-N-[(8S,14S)-22-ethyl-4-hy roxy-21-[4- {mefhoxymethyl)pyridsrs-3-yl]-18,18-difnethyl-9,15-d!Oxo-16 Oxa-10,22,28- triazapentacy o[18 , 5.2.1 2 , 6, 1 ia , 14 .0 23 , 27 ]noriacosa-1 (26), 2,4, 6(29), 20, 23(27), 24 heptaers-8 yl] 2-{1

Step 1. A mixture of ierf-butyl (2S)-3-methyl-2-[W-methyl-1-(3S)-pyrrolidin-3- ylformamido]butanoate (300 mg, 1 .1 mmol) and DIPEA (409 mg, 3.2 mmol) in MeCN (4 mL) at 0 °C was added bromoacetyl bromide (256 mg, 1 .3 mol) dropwise. The mixture was stirred at 0 °C for 30 min, then concentrated under reduced pressure and the residue was purified by C 1 8-siiica gel column chromatography to give tert-butyl (2S)-2-[1-[(3S)-1-(2-bromoacetyl)pyrroiidin-3-yl]-N -methylformamido]-3- methylbutanoate (350mg, 73% yield) as an oil. LCMS (ESI): m/z [M+H] calc ' d for Cr/HjsBrNjCb 404.1 ; found 405.2 and 407.2.

Step 2, To a mixture of tert-butyl (2S)-2-[l -[(3S)-1-(2-bromoacetyl)pyrroiidin-3-yl]-W- methylformamido]-3-methylbutanoate (110 g, 0.27 mmol) and K?003 (75 mg, 0.54 mmol) in DMF (2 mL) at 0 °C was added (3S)-pyrrolidin-3-ol (36 mg, 0.41 mmol) dropwise. The mixture was stirred at 0 c C for 1 h, then concentrated under reduced pressure and the residue was purified by prep-HPLC to give tert- butyl (2S)-2-[1-[(3S)-1-[2-[{3S)-3-hydroxypyrrolidin-1-yl]acetyl]p yrrolidin-3-yl]-W-methylformamido]-3- methylbutanoate (60 mg, 48% yield) as an oil. LCMS (ESI): m/z [M+H] ealc’d for C 2 1H 3 7N3O5411 .3; found 412.5.

Step 3. To a mixture of terf-butyl (2S)-2-[1-[(3S)-1-[2-[(3S)-3-hydroxypyrroiidin-1- yi]acetyl]pyrroiidin-3-yl]-N- methylformamido]-3-methylbutanoate (60 mg, 0.15 mmol) in DCM (0.50 mL) at 0 ,3 C was added TFA (0.50 mL, 6.7 mmol) dropwise. The mixture was warmed to rt and stirred for 2 h, then concentrated under reduced pressure with toluene (x 3) to give (2S)-2-[1-[(3S)-1-[2-[(3S)-3- hydroxypyrroiidin-1-yl]acetyl]pyrroiidin-3-yl]-W-methylforma mido]-3-methylbutanoic acid (70 mg, crude) as an oil. LCMS (ESI): m/z [M+H] calc'd for C 1 7H 2 9N3O5355.2; found 356 4.

Step 4. To a mixture of (6 3 S,4S)-4-amino-1 1 -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)- 10,10-dimethyl-6',6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-Sndola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione (60 mg, 0.1 mmol) and DIPEA (124 g, 1.0 mmol) in DMF (1 ml..) at -10 °C was added (2S)-2-[1-[(3S)-1-[2-[(3S)-3-hydroxypyrrolidin-1-yl]acetyl]p yrrolidin-3-yl]-N- methylformamido]-3-methylbutanoic acid (51 g, 0.14 mmol) and GIF (40 mg, 0.14 mmol) in portions.

The mixture was stirred at -10 °C for 1 h, then diluted with H 2 O (30 ml) and extracted with EtOAc (3 x 10 ml.). The combined organic layers were washed with brine (1 x 10 ml), dried over anhydrous NA 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by prep- HPLC to give (2S)-N-[(8S,14S)-22-ethyl-4-hydroxy-21-[4-(methoxymethyl)pyr idin-3-yl]-18,18-dimethyl- 9,15-dioxo-16-oxa-10,22,28-triazapentacyclo[18.5.2.1 a , 6 .1 "VTCF^lnonacosa-

1 (26),2,4,6(29),20,23(27),24-heptaen-8-yl]-2-{1-i(3S)-1-{2-[( 3 ?)-3-hydroxypyrrolidin-1-yl]acetyl)pyrrolidin- 3-yl]-N -methylformamido)-3-methylbutanamide (8.6 mg, 8% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for GsaHzoNeOs 962.5; found 963.5; 1 H NMR (400 MHz, CD3OD) 6 8.70 (id, J = 5.1 , 1 .6 Hz, 1H), 8.66 - 8.48 (m, 1H), 8.07 - 7.90 (m, 1H), 7.76 (dd, J = 9.9, 5.2 Hz, 1H), 7.61 (tt, J= 9.9, 2.0 Hz, 1H), 7.52 (dt, J = 8.7, 3.5 Hz, 1H), 7.11 - 6.97 (m, 1H), 6.62 6.47 (m, 1H), 5.68 - 5.48 (m, 1H), 4.79 (dt, J = 11.2, 9.1 Hz, 1H), 4.53 - 4.18 (m, 4H), 4.16 - 3.86 (m, 3H), 3.85 3.56 (m, 7H), 3.55 - 3.46 (m, 1H), 3.42 (d, J = 4.6 Hz, 4H), 3.26 · 3.01 (m, 3H), 3.01 - 2.60 (m, 9H), 2.42 2.01 (m, 6H), 1 .92 (s, 1H), 1 .75 (s, 2H), 1 .62 (q, J= 12.7 Hz, 1H), 1.26 · 0.80 (m, 13H), 0.61 - 0.40 {m, 3H).

Example A24. Synthesis of (2S)-W-[(8S,14S)-22-ethyl-4-hydroxy-21-[4- Cmethoxyniethyl)pynd -3-yl]-18, 18-dirriethyl-9,15-di!OXo-16-0X0-10,22,28- trlazapenfacy o[18.5.2.1 2 , ® .1 1 °, 1 ' ! .O 23 , 27 ]n0nac0sa-1(26),2,4,6(29),2O,23(27),24-heptaen-8-yl]-2 -{1- [(3S5-1-methariesyifO!iylpyrrolidin-3-yl]-N -r!iethyl fomianild0)-3-methyl bi!tariamide

Step 1. To a mixture of tert-butyl N -methyl-N- ({S)-pyrrolidine-3-carbonyl)-L-valinate (500 mg, 1 .8 mmol) in DCM (8 mL)at 0 °C under an atmosphere of Na was added TEA (356 mg, 3.5 mmol), followed by MsCI (242 g, 2.1 mmol). The mixture was warmed to rt and was stirred for 3 h, then washed with brine (2 x 10 mL). The combined organic layers were dried over anhydrous NaaSOs, filtered, and the filtrate concentrated under reduced pressure and the residue was by purified by silica gel column chromatography to give tert-butyl ( V-methyl-N -((S)-1-(methylsulfonyl)pyrrolidine-3-carbonyl)-L-valinate (540 mg, 85%) as an oil. LCMS (ESI): rrt/z [M+H] calc’d for C 1 6H 3 0N 2 O5S 362.2; found 363.1 .

Step 2. A mixture of tert-butyl N -methyl-N- ((S)-1-(methylsulfonyl)pyrrolidine-3-carbonyl)-L- valinato (570 mg, 1 .6 mmol), DGM (8 mL) and TFA (2 mL) at rf under an atmosphere of N 2 was stirred for 1 h. The mixture was concentrated under reduced pressure with toluene (5 ml) to give N- methyl-N-((S)- 1-(methylsulfonyl)pyrroiidine-3-carbonyl)-L-valine (500 g) as an oil. LCMS (ESI): m/z [M+H] calc’d for C 1 2H 2 2N 2 O5S 305.1 ; found 306.2.

Step 3. To a mixture of (6 3 S,4S)-4-amino-1 1 -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)- 10,10-dimethyl-6 1 ,6 2 ,6 3 ,8 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-8(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione (80 mg, 0.13 mmol) in DMF (2 mL) at 0 °C under an atmosphere of N 2 was added DIPEA (165 mg, 1.3 mmol), N -methyl-N -((S)-1-(methylsul!onyl)pyrrGlidine-3-earbonyl)-L- valine (59 mg, 0.19 mmol) and COMU (71 mg, 0.17 mmol). The mixture was stirred at 0 °C for 1 h, then concentrated under reduced pressure and the residue was purified by prep-HPLC to give (2 S)-N- [(88,148)-22-e†hyl-4-hydroxy-21-[4-(me†hoxymethyl)pyridi n-3-yl]-18,18-dimethyl-9,15-dioxo-l 6-oxa- 10,22,28-triazapentacyclo[18.5.2.1 2 , 6 .1 10 , 14 .0 23 , 27 ]nonacosa-1 (26),2,4,6(29),20,23(27),24-heptaen-8-yl]-2-

{1-[(3S)-1-methanesulfonylpyrrolidin-3-yl3-N -methylformamido)-3-methylbutanam!de (42 mg, 36% yield) as a solid. LCMS (ESI): /z [M+H] calc’d for C-sHeaHzOsS 913.4; found 914.6; T-i NMR (400 MHz, DMSO-cfe) d 9.35 - 9.33 (m, 1H), 8.74 - 8.62 (m, 2H), 8.52 (s, 1H), 8.19 - 8.11 (m, 1H), 7.92 (s, 1H), 7.64 - 7.60 (m, 2H), 7.53 (t, J = 9.0 Hz, 1H), 7.22 - 7.10 (m, 1H), 7.02 (s, 1H), 6.58 - 6.48 (m, 1H), 5.37 - 5.24 (m, 1H), 5.19 - 5.04 (m, 1H), 4.30 - 4.18 (m, 3H), 4.07 - 3.91 (m, 3H), 3.75 - 3.49 (m, 6H), 3.22 (d, J= 1.5

Hz, 2H), 2.97 - 2.91 (m, 4H), 2.92 - 2.65 (m, 7H), 2.27 (s, 1H), 2.06 (d, J ~ 14.4 Hz, 3H), 1.85 (d, J - 35.3 Hz, 2H), 1 .70 - 1 50 (m, 2H), 1 .09 - 0.88 (m, 8H), 0.85 - 0.72 (m, 5H), 0 43 (d, = 17.8 Hz, 3H)

Example A37. Synthesis of (255-N-[(85,14ίϊ)-22-b11 I-4-1 <ΐGqcg-21-[4- {methoxymethyl )pyri irs-3-yl]-18,18-difnethyl -9,15-d!Oxo-16 Oxa-10,22,28- fnazapentacy o[18.5.2.1 2 , s .1 l V 4 -0 23 , 2r ]rsoriacosa-1(26),2,4 ! 6(29),20,23(27),24-heptaeri-8-yl]-2-{1- [(S^-1-^S-hydroxyaaetidirs-1-y suiforsyllpyrroydin-S-yll-M-fnethylformamidol-S-fnethylbutan amide

Step 1, To a mixture of tert-butyl W-methyl-N -((i>)-pyrrolidine-3-carbonyl)-L-valinate (500 mg, 1 .8 mmol) in DCM (20 ml.) ar rt was added TEA (356 mg, 3.5 m ol) and 3-{benzyloxy)azetidine-1-sulfonyl chloride (460 mg, 1 .8 mmol). The mixture was stirred at rt overnight, then concentrated under reduced pressure and the residue was purified by prep-HPLC to give ierf-butyl N -((S)-1-((3-(benzyloxy)azetidin-1- yl)sulfonyl)pyrroiidine-3-carbonyl)-W-methyl-L-valinate (390 g, 44% yield) of as an oil. LCMS (ESI): m/z [M+H] calc’d for C 2 5H 3 9N3O6S 509.3; found 510.5.

Step 2, A mixture of tert-butyl W-((S)-1-((3-(benzyloxy)azetidin-1-yl)sulfonyl)pyrrolidine-3 - carbonyl)-N -methyl-L-vaiinate (390 mg, 0.77 mmol), DCM (4 rrsL) and TEA (1 ml) at rt under an atmosphere of M2 was stirred at rt for 2 h. The mixture was concentrated under reduced pressure with toluene (10 mL x 2) to give N- ({S)-1-({3-(benzyioxy)azetidin-1-yl)sulfonyl)pyrroiidine-3-c arbonyl)-N- methyl-L-valine (370 mg, crude) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 2 1H 3 1 N3Q6S 453.2; found 454.5.

Step 3. To a mixture of (6 3 S,4S)-4-amino-1 l -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)- 10,10-dimethyl-6 1 I 6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione (60 mg, 0.1 mmol) in DMF (8 mL) at 0 °C under an atmosphere of NK was added DIPEA (124 mg, 0.96 mmol), N- ((S)-1-((3-(benzyloxy)azetidin-1-yl)sulfonyl)pyrrolidine-3- carbonyl)-N- methyl-L-valine (65 mg, 0.14 mmol) and COMU (58 mg, 0.13 mmol). The mixture was stirred at 0 °C for 1 h, then concentrated under reduced pressure and the residue was purified by prep-HPLC to give (3S)-1-((3-(benzyloxy)azetidin-1-yl)sulfonyl)-W-((2S)-1-(((6 3 S,4S)-1 1 -ethyl-2 5 -hydroxy-l 2 -(4- (methoxymethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 T-/-8-oxa-1 (5,3)- indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan- 2-yl)-N- methylpyrroiidine-3-carboxamide (52 mg, 51% yield) as an oil. LCMS (ESI): m/z [M+H] calc'd for C57H72N8O10S 1060.5; found 1061 .3

Step 4. A mixture of (3S)-1-((3-(benzyioxy)azetidin-1-yl)sulfonyl)-N-((2S)-1-(((6 3 S,4S)-1 1 -ethyl- 2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyhdin-3-yl)-10,10-diiTiethyl-5,7-d ioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1- oxobufan-2-yl)-N-methylpyrrolidine-3-carboxamide (55 g, 0.05 mmol), MeOH (3 ml..) and Pd(OH) 2 / ' C (11 g, 20% by weight) was stirred under a H 2 atmosphere for 12 h. The mixture was filtered, the filtrate was concentrated under reduced pressure and the residue was purified by prep-HPLC to give {2S)-N- [(8S,14S)-22-ethyl-4-hydroxy-21-[4-(meihoxymethyl)pyridin-3- yl]-18,18-dimethyl-9,15-dioxo-16-oxa- 10,22,28-triazapentacyclo[18.5.2.1 2 , e .1 1 °, 14 .0 a3 , 2 nnonacosa-1 (26),2,4,6(29),20,23(27),24-heptaen-8-yl]-2- {1-[(3S)-1-[(3-hydroxyazetidin-1-yl)sulfonyl]pyrroiidin-3-yl ]-N- methylformamido)-3-methylbutanamide (6.5 mg, 13% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 5 oHeeNsOioS 970.5; found 971 .2; 1 H NMR (400 MHz, DMSO-dfe) d 9.33 - 9.29 (m, 1H), 8.75 - 8.65 (m, 2H), 8.52 (s, 0.5H), 8.15 - 8.06 (m, 0.5H), 7.92 (s, 1H), 7.65 - 7.50 (m, 3H), 7.22 - 7.14 (m, 1H), 7.02 (s, 1H), 6.58 6.46 (m, 1H), 5.84 5.80 (m, 1H), 5.28 · 5.22 (m, 0.6H), 4.75 · 4.69 (m, 0.4H), 4.45 · 4.12 (m, 4H), 4.05 3.88 (m, 5H), 3.72 3.50 (m, 7H), 3.22 (s, 2H), 3.12 3.04 (m, 1H), 2.94 2.70 (m, 7H), 2.29 - 2.03 (m, 5H), 1 .90 · 1 .77 (m, 2H), 1 .76 - 1 .45 (m, 2H), 1 .24 (s, 1H), 1 .08 1 .02 (m, 2H), 1 .01 0.72 (m, 12H), 0.5 - 0.43 (m, 3H).

Example A42. Synthesis of (3S)-N3-[(1S)-1-{[(SS,14S)-22-etlhyl-4-hydraxy-21-[4- Cmethoxymethyl)pynd -3-yf]-18,18-diiriethyf-9,15- soxo-16-oxa-10,22,28- trlazapentacy o[18,5.2.1 2 , ® ,1 1 °, 1 ' ! .0 23 , 27 ]nonacosa-1(26),2,4,6(29),2O,23(27),24-heptaen-8-

Step 1. To a mixture of ferf-butyl (2S)-3-methyl-2-[N-methyl-1-(3S)-pyrrolidin-3- ylformamido]butanoate (200 mg, 0.7 mmol) and TEA (142 mg, 1 .4 mmol) In DCM (10 mL) at 0 °C under an atmosphere of N 2 was added dimethylcarbamyl chloride (91 mg, 0.84 mmol) in portions. The mixture was warmed to rt and stirred for 1 h, then H 2 O added and the mixture extracted with DCM (3 x 50 mL). The combined organic layers were washed with brine (1 x 5 mL), dried over anhydrous NA 2 SO 4 , filtered and the filtrate concentrated under reduced pressure to give tert-butyl (2S)-2-[1-[(3S)-1- (dimethylcarbamoyl)pyrroiidin-3-yl]-N- methylformamido]-3-methylbutanoate. which was used in the next step without further purification.

Step 2. A mixture of tert-butyl (2S)-2-[1-[(3S)-1-(dimethylcarbamoyl)pyrraiidin-3-yl]-W- methylformamido]-3-methylbutanoate (335 g, 0.94 mmol) in DCM (10 ml..) and TFA (2 ml., 26.9 mmol) was stirred at rt for 2 h. The mixture was concentrated under reduced pressure to give (2S)-2-[1-[(3S)-1- (dimethylcarbamoyl)pyrroiidin-3-yl]-N- methylformamido]-3-methylbutanoic acid, which was used directly in the next step without further purification. LCMS (ESI): m/z [M+H] calc'd for C 14H 2 5N3O4299.2; found 300 2.

Step 3. To a mixture of (6 3 S,4S)-4-amino-1 1 -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)- 10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione (80 mg, 0.13 mmol) and (2S)-2-[1-[(3S)-1- (dimethylcarbamoyl)pyrroiidin-3-yl]-N- methylformamido]-3-methylbutanoic acid (57 mg, 0.19 mmol) in MeCN (3 mL) at 0 °C under an atmosphere of N 2 was added lutidine (137 mg, 1 .3 mmol) and COMU (77 mg, 0.18 mmol) in portions. The mixture was stirred at 0 °C for 1 h, then concentrated under reduced pressure and the residue was purified by prep-HPLC to give (3S)-N3-[(1 S)-1-{[(8S,14S)-22-ethyl-4- hydroxy-21-[4-(methoxymethyl)pyridin-3-yl]-18,18-dimethyl-9, 15-dioxo-16-oxa-10, 22,28- triazapentacyclo[18.5.2.1 2 , 6 .1 10 , 14 .0 23 , 27 ]nonacosa-1 (26),2,4,6(29),20,23(27),24-heptaen-8-yl]carbamoyl)- 2-methylpropyl]-N 1 ,L/1 ,L/3-trimethylpyrroiidine-l ,3-dicarboxamide (45.6 mg, 39% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 5 oHeeNeOe 906.5; found 907.4; 1 H NMR (400 MHz, DMSO-efe) 6 9.31 - 9.30 (m, 1H), 8.72 - 6.71 (m, 1H), 8.59 (d, J= 50.4 Hz, 1H), 7.92 - 7.90 (m,1H), 7.74 - 7.42 (m, 3H), 7.23 - 7.08 (m, 1H), 7.00 id. ,/ ··· 13.4 Hz, 1H), 6.56 - 6.49 (m, 1H), 5.45 - 5.32 (m, 1H), 5.26 - 5.04 (m, 1H), 4.87 - 4.64 (m, 1H), 4.53 - 4,35 (m, 1H), 4.32 - 4.09 (m, 3H), 4.12 - 3.81 (m, 3H), 3.81 - 3.37 (m, 6H), 3.23 (t, J - 1.6 Hz, 2H), 3.12 - 3.10 (m, 1H), 3.01 - 2.52 (m, 13H), 2.23 - 1.95 (m, 4H), 1.81 (s, 1H), 1.67 (s, 1H), 1.60 - 1.47 (m, 1H), 1.28 - 1.22 (m, 1H), 1.21 - 1.14 (m, 1H), 1 11 - 1.02 (m, 2H), 1.02 - 0.66 (m, 12H), 0 43 (d, J- 16 8 Hz, 3H).

Example A27. Synthesis of (2¾-N -[(8S,14¾-22-ethyl-4-hydroxy-21-[4- (methoxymethyl )pyrldirs-3-yl]-18,18-dimethyl-9,15-d!Oxo-16-oxa-10,22,28-

Step 1. A mixture of tert- butyl (2S)-3-methyl-2-[N- methyl-1-(3S)-pyrrolidin-3- ylformamido]butanoate (80 mg 0.28 mmol), Ti(Oi-Pr)4 (88 mg, 0.31 mmol) and paraformaldehyde (26mg 0.29 mmol) in MeOH (2 mL) was stirred at rt under an atmosphere of air overnight. The mixture was cooled to 0 °C and NaBH(OAc)3 (107 mg, 0.51 mmol) was added. The mixture was warmed to rt and stirred for 2 h, then cooled to 0 °C and H 2 O (0.2 mL) added. The mixture was concentrated under reduced pressure and the residue was purified by C 1 8-silica gel column chromatography to give terf-butyl (2S)-3-methyl-2-[W-methyl-1-[(3S)-1-methylpyrrolidin-3-yl]fo rmamido]butanoate (97 mg, crude) as an oil. LCMS (ESI): m/z [M+H] calc’d for C 1 6H 3 0N 2 O 3 298.2; found 299.3.

Step 2. A mixture of fe/T-butyl (2S)-3-methyl-2-[N -methyl-1-[(3S)-1-methylpyrrolidin-3- yl]formamido]butanoate (97 mg, 0.32 mmol) in DGM (2 mL) and TFA (1 mL, 13.5 mmol) was stirred at rt for 1 h, then the mixture was concentrated under reduced pressure to give (2S)-3-methyl-2-[W-methyl-1 [(3S)-1-methylpyrrolidin-3-yl]formamido]butanoic acid (100 g, crude) as an oil. LCMS (ESI): m/z [M+H] calc ' d for C 1 2H 2 2N 2 G3242.2; found 243.2.

Step 3. To a mixture of (6 3 S,4S)-4-amino-1 '-ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)- 10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione (80 mg, 0.13 mmol) and (2S)-3-methyl-2-[N -methyl-1-[(3S)-1- methylpyrrolldin-3-yl]formamido]butanoic acid (47 g, 0.19 mmol) in MeCN (2 mL) at 0 c C was added 2,6-dimethylpyridine (137 mg, 1 .3 mmol) and GOMU (77 mg, 0.18 mmol). The mixture was warmed to rt and stirred for 1 h, then concentrated under reduced pressure and the residue was purified by prep-HPLC to give (28)-N- [(8S,14S)-22-ethyl-4-hydroxy-21-[4-(methoxymethyl)pyridin-3- yl]-18,18-dimethyl-9,15- dioxo-16-oxa-10,22,28-triazapentacyclo[18.5.2.1 2 , ® 1 1 °, 14 .0 23 , 27 ]nonacosa-1 (28), 2, 4, 6(29), 20,23(27), 24- heptaen-8-yl]-3-methyl-2-{N -methyl-1-[(3S)-1-methylpyrrolidin-3-yl]formamido)butanamide (28 mg, 26% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 4 8H63N7O7849.5; found 850.5; 1 H NMR (400 MHz, DMSO-dfe) 6 9.31 (s, 1H), 8 72 (t, J = 5.1 Hz, 1H), 8.67 - 8.50 (m, 1H), 798 - 7.87 (m, 1H), 7.67 - 7.47 (m, 3H), 7.22 - 7.07 (m, 1H), 7.01 (s, 1H), 6.53 (d, - 40.1 Hz, 1H), 5.44 - 5.00 (m, 2H), 4.46 - 4.12 (m, 3H), 4.08 - 3 79 (m, 3H), 3.79 - 3.45 (m, 3H), 3.22 (d, J == 1 2 Hz, 2H),3.14 - 2.94 (m, 2H), 2.92 - 2.55 (m, 10H), 2.43 - 2 20 (m, 4H), 2.19 - 1.92 (m, 4H), 1.81 (d, J = 11 9 Hz, 2H), 1.67 (s, 1H), 1.53 (s, 1H), 1.09 (t, J = 7.1 Hz, 1H), 1.02 - 0.91 (m, 3H), 0 91 - 0.80 (m, 5H), 0.80 - 0.67 (m, 3H), 0.42 (d, J= 21 .7 Hz, 3H).

Example A23. Synthesis of (2S)- -[(8S,14S)-22-ethyl-4-hydroxy-21-[4- (methoxymethyl )pyridirs-3-yl]-18,18-dimethyl-9,15-d!Oxo-16-oxa-10,22,28- triazapentaoycio[18.5.2.1 2 , s .T l0 14 .0 a3 , 2r ]^o^¾cosa-1(26),2,4,6(29),20 23(27),24-heptaen-8-yl]-2-{1- [(3S)-1-(2-hydroxyethyl )pyrrol! in-3-yl]-N-methyl formamido)-3-methyl butariarr!!de

Step 1. To a mixture of terf-butyl (2S)-3-methyl-2-[N-meihyl-1-(3S)-pyrrolidin-3- ylformamido]butanoate vanadium (200 mg, 0.6 mmol) and 2-bromoethanol (224 g, 1 .8 m ol) in DMF (5 ml.) at rt was added CS2CO 3 (777 mg, 2.4 rnmol) and Kl (50 mg, 0.3 mmol). The mixture was stirred at rt for 16 h then diluted with H 2 O and extracted with EtOAc (3 x 100 mL). The combined organic layers were washed with brine (2 x 100 mb), dried over anhydrous NA 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by C 1 8-silica gel column chromatography to give /erf-butyl (2S)-2-[1-[(3S)-1-(2-hydroxyethyl)pyrroiidin-3-yl]-W-methylf ormamido]-3-methylbutanoate (201 mg, crude) as a solid. LCMS (ESI): m/z [M+H] calc’d for Co^lNLO. 328.2; found 329.4.

Step 2, A mixture of tert-butyl (2S)-2-[1-[(3S)-1-(2-hydroxyethyl)pyrroiidin-3-yl]-N- methylformamido]-3-methylbutanoate (100 mg, 0.3 rnmol) in DCM (1 mL) and TEA (0.50 mL) at rt was stirred for 1 h, then concentrated under reduced pressure to give (2S)-2-[1-[(3S)-1-(2- hydroxyethyl)pyrroiidin-3-yl]-N -methylformamido]-3-methylbutanoic acid (110 g, crude) as an oil. LCMS (ESI): m/z [M+H] calc’d for C 1 3H 2 4N 2 O4272.2; found 273.2.

Step 3, To a mixture of (6 3 S,4S)-4-a ino-1 1 -ethyl-2 5 -hydroxy-1 2 -(4-(methoxymethyl)pyridin-3-yl)·· 10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 ' H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2(1 ,3)- benzenacycloundecaphane-5,7-dione (60 mg, 0.1 mmol) and (2S)-2-[1-[(3S)-1-(2-hydroxyethyl)pyrrolidin- 3-yl]-iV-methylformamido]-3-methylbutanoic acid (31 mg, 0.11 mmol) in MeCN (2 mL) at 0 °C under an atmosphere of N 2 was added 2,6-dimethylpyridine (103 mg, 1 .0 mmol) and COMU (58 g, 0.13 mmol). The mixture was warmed to rt and stirred for 1 h, then concentrated under reduced pressure and the residue was purified by prep-HPLG to give (2S)-N-[(8S,14S)-22-ethyl-4-hydroxy-21-[4- (methoxymethyl)pyridin-3-yl]-18,18-dimethyl-9,15-dioxo-16-ox a-10,22,28- triazapentacyclo[18.5.2.1 s , 6 .1 10 , ,4 .0 S3 , s? ]nonacosa-1 (26),2,4,6(29),20,23(27),24-heptaen-8-yl]-2-{1-[(3S)-1 - (2-hydroxyethyl)pyrrolidin-3-yl]-N-methylformamido)-3-methyl butanamide (13 mg, 16% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for C 4 9H65N7O8879.5; found 880.3; 1 H NMR (400 MHz, DMSO-db) d 8.72 (t, J = 5.3 Hz, 1H), 8 68 - 8.58 (m, 1H), 8.52 is, 1H), 7.93 (d, J = 10.6 Hz, 1H), 7.68 - 7.58 (m, 2H), 7 53 (d, J= 7.1 Hz, 1H), 7.21 - 7.07 (m, 1H), 7.01 (s, 1H), 6.52 (d, J = 42.8 Hz, 1H), 5.35 (d, J= 25.5 Hz, 1H), 5.22 - 4.97 (m, 1H), 4.59 - 4.35 (m, 1H), 4.23 (t, J = 13.8 Hz, 3H), 4.11 - 3.81 (m, 3H), 3.81 - 3.56 (m, 2H), 3.56 - 3.47 (m,3H), 3.22 (d, J= 1.2 Hz, 2H), 3.09 (d, J = 12.6 Hz, 1H), 2.99 - 2.65 (m, 10H), 2.57 - 2.53(m,1H), 2.47 - 2.19 (m, 2H), 2.14 - 2.08(m, 1H), 2.08 (s, 1H), 2.06 - 1.98 (m, 2H), 1.81 is, 2H), 1.59 (d, J= 55.9 Hz, 2H), 1.14 - 0.67 (m, 13H), 0.42 (d, J= 22.1 Hz, 3H).

Example A57. Synthesis of (2S)-N-[(85,14S)-22-ethyl-4-hydroxy-21-[4- (methoxymethyf)pynd -3-yf ]-18,18-dimethyl-9,1 S-dioxo-16-oxa-10,22,28- trlazapentacy o[18.5.2.1 2 , iS .1 1<t , 14 .0 :23 , 27 ]nonacosa-1(28),2,4,6(29),2O,23(27),24-heptaen-8-yl]-3 - methyl-2-(N-methylmethanesulfonamido)butanam de

Step 1. A mixture of tert-butyl N- [(SS,14S)-22-ethyl-21-[2-(2-methoxyethyl)phenyl]-18,18- dimethyl-9,15-dioxo-16-oxa-10,22,28-triazapentacyclo[18.5.2. 1 A [2,6].1 A [10,14].0 A [23,27]]nonacosa- 1 (26), 2, 4, 6(29), 20,23(27), 24-heptaen-8-yl]carbamate (880 mg, 1.2 mmol), DCM (10 mL) and TFA (5 mL) was stirred at 0 °C for 30 min. The mixture was concentrated under reduced pressure to give (8S,14S)-8- amino-22-ethyl-21-[4-(methoxymethyl)pyridin-3-yl]-18,18-dime thyl-4-[(triisopropylsllyi)oxy]-16-oxa- 10,22,28-triazapentacyclo[18.5.2.1 A [2,6].1 A [10,14].0 A [23,27]]nonacosa-1 (26), 2, 4, 6(29), 20, 23(27), 24- heptaene-9,15-dione, that was used directly in the next step without further purification. LCMS (ESI): m/z [M+H] calc’d for C+sHesNsOsSi 781 .5; found 782.7.

Step 2, To a mixture of (8S,14S)-8-amino-22-ethyl-21-[4-(methaxymethyl)pyridin-3-yl] -18,18- dimethyl-4-[(triisopropylsllyi)oxy]-16-oxa-10,22,28- triazapentacyclo[18.5.2.1 A [2,6].1 A [10,14].0 A [23,27]]nonacosa-1 (26), 2,4, 6(29),20, 23(27), 24-heptaene-

1229

5UB5TITUTE SHEET (RULE 26) 9,15-dione (880 mg, 1 13 mmol) and (2S)-2-[(terf-butoxycarbonyl)(methyl)amino]-3-methylbutanoic acid (521 mg, 2.3 mmol) in DMF (8 8 ml.) at 0 °C was added DIPEA (1 .45 g, 11 3 mmol) and COMU (88 mg, 0.21 mol). The mixture was stirred at 0 °C for 30 min, then diluted with H 2 O (100 ml.) and extracted with EtOAc (3 x 100 mL) The combined organic layers were washed with brine (3 x 100 ml..), dried over anhydrous NA 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by prep-TLC to give tert-butyl N-[(1 S)-1-[[(8S,14S)-22-ethyl-21-[4-(methoxymethyl)pyridin-3- yl]-18,18-dimethyl-9,15-dioxo-4-[(triisopropylsllyl)oxy]-16- oxa-10,22,28- triazapentacyclo[18.5.2.1 '[2,6].T''i10,14].0 A [23,27]]nonacosa-1 (26),2,4,6(29),20,23(27),24-heptaen-8- yl]carbamoyl]-2-methylpropyl]-N-methylcarbamate (1 g, 89% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 5 eHo-NeOeSi 994.6; found 995.5.

Step 3, A mixture of tert-butyl N-[{ 1 S)-1-[[(8S,14S)-22-ethyl-21-[4-(methoxymethyl)pyridin-3-yl]- 18,18-dimethyl-9,15-dioxo-4-[{triisopropylsllyl)oxy]-16-oxa- 10,22,28- triazapentacyclo[18.5.2.1 L [2,6].1 L [10,14].0 A [23,27]]nonacosa-1 (26),2,4,6(29),20,23(27),24-heptaen-8- yi]carbamoyl]-2-methylpropyl] N- methylcarbamate (1 .0 g, 1 .0 mmol), DCM (10 mL) and TFA (5 L) was stirred for 30 min. The mixture was concentrated under reduced pressure and the residue was basified to pH -8 with saturated NaHCCb, then extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (3 x 10 L), dried over anhydrous NasSCu, filtered and the filtrate concentrated under reduced pressure to give (2S)-W-[(8S,14S)-22-ethyl-21-[4-(methoxymethyl)pyridin-3-yl] -18,18-dimethyl- 9,15 dloxo-4-[(triisopropylsllyl)oxy] 16-oxa-10,22,28- triazapentacyclo[18.5.2.1 L [2,6].1 L [10,14].0 A [23,27]]nonacosa-1 (26), 2,4, 6(29),20, 23(27), 24-heptaen-8-yl]- 3-methyl-2-(methylamino)butanamide (880 mg, 98% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 5 iHy NeOsSi 894.5; found 895.5.

Step 4. To a mixture of (2S)-N -[(8S,14S)-22-ethyl-21-[4-(methoxymethyl)pyridin-3-yl]-18,18 - dimethyl-9,15-dioxo-4-[(triisopropylsilyl)oxy]-16-oxa-10,22, 28- triazapentacyclo[18.52.1 L [2,6].1 L [10,14].0' v [23,27]]nonacosa-1 (26), 2,4, 8(29),20, 23(27), 24-heptaen-8-yl]- 3-methyl-2-(methylamino)butanamide (90 mg, 0.1 mmol) in DCM (2 mL) at 0 °C was added DIPEA (85 g, 0.5 mmol) and MsCI (14 mg, 0.12 mmol). The mixture was stirred at 0 C for 30 min, then concentrated under reduced pressure and the residue diluted with H 2 O (5mL) and extracted with EtOAc (3 x 5 ml.). The combined organic layers were washed with brine (3 x 5 ml), dried over anhydrous Na 2 S0 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by prep-TLC to give (2S)-N- [(8S,14S)-22-ethyl-21-[4-(methoxymethyl)pyridin-3-yl]-18,1 S-dimethyl-9,15- dioxo-4-[(triisopropylsllyi)oxy]-16-oxa-10,22,28- triazapentacyclo[18.5.2.1 A i2,6].T'f10,143.0 A [23,27]]nonacosa-1 (26), 2,4, 8(29),20, 23(27), 24-heptaen-8-yl]- 3-methyl-2-(N- methylmethanesulfonamido)butanamide (60 g, 61% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 5 ^HjeNsOsSSi 972.5; found 973.7.

Step 5. To a mixture of (2S)-W-[(8S,14S)-22-ethyl-21-[4-(methoxymethyl)pyridin-3-yl] -18,18- dimethyl-9,15-dioxo-4-[(triisopropylsilyl)oxy]-16-oxa-10,22, 28- triazapentacyclofl 8.5.2.1 L [2,6].1 L [10,14].0 A [23,27]]nonacosa-1 (26), 2,4, 6(29),20, 23(27), 24-heptaen-8-yl]- 3-methyl-2-(N- methylmethanesulfonamido)butanamide (60 mg, 0.06 mmol) in THE (2 mL) at 0 °C was added 1 M TBAF In THE (6 L, 0.006 mmol). The mixture was stirred at 0 °C for 30 min, then diluted with H 2 O (5 mL) and extracted with EtOAc (3 x 5 mL). The combined organic layers were washed with brine (3 x 5 mL), dried over anhydrous teSO. and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by prep-TLC to give (2S)-W-[(8S,14S)-22-ethyl-4-hydroxy-21-[4- (methoxymethyl)pyridin-3-yl]-18,18-dimethyl-9,15-dioxo-16-ox a-10,22,28- triazapentacyclo[18.5.2.1 2 ® 1 1t V 4 -0 23 , 2? ]nonacosa-1 (26), 2,4,6(29), 20, 23(27), 24-heptaen-8-yl]-3-methyl-2- (N-methylmethanesulfonamido)butanamide (50 mg, 99% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for CwHseNeOeS 816.4; found 817.5; 1 H NMR (400 MHz, DMSO-db) d 9.34 (d, J = 1 .8 Hz, 1H), 8 72 (t, J = 5.2 Hz, 1H), 8.65 (d, J = 5.8 Hz, 1H), 7.99 - 7.86 (m, 1H), 771 - 7.45 (m, 3H), 7.19 (d, J = 41 5 Hz, 1H), 7.03 (t, J = 1.9 Hz, 1H), 6.66 (d, J= 10.4 Hz, 1H), 5.34 (q, J == 8.1 Hz, 1H), 5.14 (dd, J == 62.7, 12.2 Hz, 1H), 4.55 - 4.15 (m, 3H), 4.14 - 3.80 (m, 4H), 3.80 - 3.46 (m, 3H), 3.23 (s, 1H), 3.02 - 2.72 (m, 8H), 2 68 (s, 2H), 2.15 - 1.89 (m, 3H), 1.82 (d, J = 12.4 Hz, 1H), 1.76 - 1.62 (m, 1H), 1.54 (q, J = 12.7 Hz, 1H), 1.24 (s, 1H), 1.08 (t, J= 7.1 Hz, 2H), 1.03 - 0.86 (m, 9H), 0.81 (s, 2H), 0.46 (s, 3H).

Example A43. Synthesis of (2i¾-/^[(8S,14S)-22-ethyl-4-hydroxy-21-[4- (met hoxymethyl )pyndiri-3-yf ]-18,1 B-dirrjethyl -9,1 S-dioxo-16-oxa-10,22,28- trlazapentacy o[18.5.2.1 2 , ,5 .1 1<t , 14 .0 :23 , 27 ]nonacosa-1(28),2,4,6(29),20,23(27),24-heptaen-8-yl]-2 -(2- hydroxy-N -methylacetarTi!do)-3-8Tsethylbytanar!i!cle

Step 1. To a mixture of (2S)-N -[(8S,14S)-22-ethyl-21-[4-(methoxymethyl)pyridin-3-yl]-18,18 - dimethyl-9,15-dioxo-4-[(triisopropylsllyi)oxy]-16-oxa-10,22. 28- triazapentacyclo[18.5.2.1 L [2,6].1 L [10,14].0 A [23,27]]nonacosa-1 (26), 2,4, 6(29),20, 23(27), 24-heptaen-8-yl]- 3-methyl-2-(methylamino)butanamide (100 mg, 0.11 mmol) in DCM (1 mL) at 0 °C was added DlPEA (72 g, 0.56 mmol) and 2-chloro-2-oxoethyl acetate (11 .53 g, 0.11 mmol). The mixture was warmed to rt and stirred for 30 min, then concentrated under reduced pressure, diluted with water (3 mL) and extracted with EtOAc (3 x 3 mL). The combined organic iayers were washed with brine (3 x 3 mL), dried over anhydrous MasSOl· and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by prep-TLC to give [[(1 S)-1-[[(8S,14S)-22-ethyl-21-[4-(methoxymethyl)pyridin-3-yl]- 18,18- dimethyl-9,15-dioxo-4-[(triisopropylsilyi)oxy]-16-oxa-10,22, 28- triazapentacyclo[18.52.1 L [2,6].1 L |10,14].0 A [23,27]]nonacosa-1 (26),2,4,6(29),20,23(27),24-heptaen-8- yl]carbamoyl]-2-methylpropyl](methyl)carbamoyi]methyl acetate (80 mg, 72% yield) as a solid. LCMS (ESI): m/z [M+H] calc'd for C 5 sHysNeOgSI 994.6; found 995.7.

Step 2. A mixture of [[(1 S)-1-[[(8S,14S)-22-ethyl-21-[4-(methoxymethyl)pyridin-3-yl]- 18,18- dimethyl-9.15-dioxo-4-[(triisopropylsllyi)oxy]-16-oxa-10,22, 28- triazapentacyclo[18.5.2.1 A [2,6].1 A [10,14].0 A [23,27]]nonacosa-1 (26), 2, 4,6(29), 20, 23(27), 24-heptaen-8- yi]carbamoyl3-2-methylpropyl](methyl)carbamoyi]methyl acetate (80 g, 0.080 mmol), DCM (1 ml.) and aqueous NH4OH (0.8 ml) was stirred at rt overnight. H 2 0 (5 mL) was added and the mixture was extracted with EtOAc (3 x 5 mL). The combined organic layers were washed with brine (3 x 5 mL), dried over anhydrous Na 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by prep-TLC to give (2S)-N- [(8S,14S)-22-ethyl-21-[4-(methoxymethyl)pyridin-3-yl]-

18.18-dimethyl-9,15-dioxo-4-[(triisopropylsllyl)oxy]-16-o xa-10,22,28- triazapentacyclo[18.5.2.1 L [2,6].1 L [10,14].0 A [23,27]]nonacosa-1 (26), 2,4, 6(29), 20, 23(27), 24-heptaen-8-yl]- 2-(2-hydroxy-W-methylacetamido)-3-methylbutanamide (60 mg, 78% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 5 sh NeOeSi 952.6; found 953.7.

Step 3. A mixture of (2S)-N -[(8S,14S)-22-ethyl-21-[4-(methoxymethyl)pyridin-3-yl]-18,18 - dimethyl-9,15-dioxo-4-[(triisopropylsllyl)oxy] -16-oxa-10,22,28- triazapentacyclo[18.5.2.1 L [2,6].1 L [10,14].0 A [23,27]]nonacosa-1 (26),2,4,6(29),20,23(27),24-heptaen-8-yl]- 2-(2-hydroxy-N -methylacetamido)-3-methylbutanamide (60 mg, 0.06 mmol), THF (2 L) and 1 M TBAF in THF (6 L, 0.006 mmol) at 0 °C was stirred for 30 min. F½0 (3 L) was added and the mixture was extracted with EtOAc (3 x 3 L). The combined organic layers were washed with brine (3 x 3 mL), dried over anhydrous NazSC The filtrate was concentrated under reduced pressure and the residue was purified by prep-TLC to give (2S)-N- [(8S,14S)-22-ethyl-4-hydroxy-21-[4-(methoxymethyl)pyridin-3- yl]-

18.18-dimethyl-9,15-dioxo-16-oxa-10,22,28-triazapentacycl o[18.5.2.1 2 , 6 1 1i \ 14 0 23 , 2? ]nonacosa-

1 (26) ,2, 4, 6(29), 20,23(27), 24-heptaen-8-yl]-2-(2-hydroxy-N- methylacetamidG)-3-methylbutanamide (20 g, 40% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 4 4H56N6O8 796.4; found 797.6; 1 H NMR (400 MHz, CD3OD) d 8.70 (dd, J = 5.7, 4.4 Hz, 1H), 8.66 - 8.49 (m, 1H), 8.00 (dd, J - 4.6, 1 .7 Hz, 1H), 7.76 (dd, J- 9.9, 5.2 Hz, 1H), 7.60 (dt, J- 8.7, 1.6 Hz, 1H), 7.56 - 7.47 (m, 1H), 7.29 - 7.18 (m, 1H), 7.10 - 6.98 (m, 1H), 6.54 (dt, J - 3.6, 1 .7 Hz, 1H), 5.67 - 5.55 (m, 1H), 4.77 (dd, J 11 .2, 8.4 Hz, 1H), 4.57 - 4.39 (m, 3H), 4.39 - 4.20 (m, 3H), 4.19 - 3.91 (m, 2H), 3.90 - 3.65 (m, 3H), 3.60 (dd, J = 11 .0, 1 8 Hz, 1H), 3.42 (s, 1H), 3.32 (s, 1H), 3 29 - 3.15 (m, 1H), 3.10 - 2.97 (m, 1H), 2.97 - 2.82 (m, 5H), 2.82 - 2.63 (m, 2H), 235 - 2.11 (m, 3H), 1.94 id, J = 13.2 Hz, 1H), 1.82 - 1.49 (m, 3H), 1.31 (s, 1H), 1 19 (t, J = 7.2 Hz, 2H), 1.09 - 0.95 (m, 7H), 0.95 - 0.83 (m, 5H), 0.50 (d, J= 32.4 Hz, 3H). Example A50. Synthesis of oxoIan-3-yl-/^[(1S)-1-{[(8S,14S)-22-ethyl -4-hydroxy-21-[4- (methoxymethyl )pyridirs-3-yl]-18,18-diniethyL9,15-d!Oxo-16-oxa-10,22,28-

Step 1. To a mixture of methyl (2S)-3-methyl-2-(methylamino)butanoate (500 mg, 3.4 mmol) and TEA (1 .44 ml, 14.2 mmol) in DGM (20 mL) at rt was added oxolan-3-yl carbonochloridate (1 .04 g, 6.9 mmol). The mixture was stirred at rt for 1 h, then sat. NEUGI added and the mixture extracted with DGM (3 x 10 mL). The combined organic layers were washed with brine (1 x 10 mL), dried over anhydrous NA 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give methyl (2S)-3-methyl-2 [methyl (oxoian-3- yloxy)carbonyl]amino]butanoate (800 mg, 89% yield) as an oil. 1 H NMR (300 MHz, GDCb) d 4.57 - 4.05 (m, 1H), 3.99 - 3.78 (m, 4H), 3.70 (s, 3H), 3.26 (s, 1H), 2.99 2.68 (m, 3H), 2.26 - 1 .83 (m, 3H), 1 .06 - 0.76 (m, 6H).

Step 2. A mixture of methyl (28)-3-meihyl-2 [methyl (oxoian-3-yloxy)carbonyl]amino]butanaate (1 g, 3.9 mmol) and 2M NaOH (19.3 mL, 38.6 mmol) in MeOH (20 mL) was stirred at rt for 1 h. The mixture was concentrated under reduced pressure and the residue was extracted with MTBE (3 x 10 L). The aqueous layer was acidified to pH -2 with 2 M HCl then extracted with DCM (3 x 20 mL). The combined organic layers were washed with brine (2 x 10 L), dried over anhydrous NasSCL, filtered and the filtrate was concentrated under reduced pressure to give (28)-3-meihyl-2-[methyl[(oxolan-3- yloxy)carbonyl]amino]butanoic acid (630 mg, 67% yield) as an oil. 1 H NMR (300 MHz, CDGIa) d 5.32 (br. s, 1H), 445 - 4.08 (m, 1H), 4.04 - 3.81 (m, 4H), 2.93 (d, J - 6.9 Hz, 3H), 2 38 - 1 .93 (m, 3H), 1 .06 (t, J - 5.6 Hz, 3H), 0.94 (d, J - 6 7 Hz, 3H).

Step 3. To a mixture of (6 3 8,4S)- 4-amino- 1 1 -ethyl-2 5 -hydroxyl-1 2 -(4-(methoxymethyl)pyridin-3- yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H- 8-oxa-1 (5,3)-indola-6(1 ,3)-pyridazina-2( 1 ,3)- benzenacycloundecaphane-5,7-dione (80 mg, 0.13 mmol), (2S)-3-methyl-2-[methyl[(oxolan-3- yloxy)carbonyl]amino]butanoic acid (63 mg, 0.26 mmol) and DIPEA (165 mg, 1 .3 mmol) in DMF (2 mL) at 0 °C was added COMU (38 mg, 0.19 mmol). The mixture was stirred at 0 °C for 30 min, then the mixture was concentrated under reduced pressure and the residue was purified by prep-HPLC to give oxoian-3- yi-N -[(1 S)-1-{[{SS,14S)-22-ethyl-4-hydroxy-21-[4-(methoxymethyl)pyri din-3-yl]-18,18-dimethyl-9,15-diaxo- 16-oxa-10,22,28-iriazapentacyclo[18.5.2.1 2 , 8 1 10 , 14 0 23 T i ]nonacosa-1 (26),2,46(29),20,23(27),24-heptaen- 8-yl]carbamoyi)-2-methylpropyl]-N -methylcarbamate (50 mg, 45% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 4 7HsoNeOg 852.4; found 853.5; 1 H NMR (400 MHz, DMSO-cfe) 6 9.34 - 9.18 (m, 1H), 8.72 (t, J= 5.1 Hz, 1H), 8.58 (d, J = 47.8 Hz, 1H), 8.48 - 8.15 (m, 1H), 7 91 (s, 1H), 7.70 - 7.57 (m, 2H), 7.55 - 746 (m, 1H), 7.13 (d, J = 24.7 Hz, 1H), 7.01 is, 1H), 6.56 (d, J = 9.2 Hz, 1H), 5.34 (s, 1H), 5.28 - 5.00 (m, 2H), 4.40 (d, J = 13.3 Hz, 1H), 4.33 - 4.14 (m, 4H), 4.12 - 3.45 (m, 10H), 3.23 (s, 1H), 3.10 (d, J = 14.5 Hz, 1H), 2.99 - 2.62 (m, 6H), 2.20 - 1.99 (m, 4H), 1.80 (s, 1H), 1.66 (s, 1H), 1.52 (d , J = 12.2 Hz, 1H), 1.09 (t, J = 7.1 Hz, 2H), 0.99 - 0.89 (m, 6H), 0.87 - 0.76 (m, 5H), 0.42 (d, J = 24.2 Hz, 3H).

Example A277. The synthesis of (2¾- -((6 3 S,4S,å)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)-5-(4- methyl piperazin-1-yl)pyr!dlrs-3-yl)-10,10-trimethyl -5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 #f-8-oxa- 2{4,2)-¾ϊ3 z oΐ3-1(5,3)-I^ol0-6(1,3)-rgp 3 z Ih0qgoIouheI©q3 Iΐ3hq-4^I)-3-!Ώ6ΐ:I'ϊnI-2··(1,3,3- trlmethyli!reido)biitanamide

Step 1. A soiution of Intermediate 10 (8.2 g, 9.89 mmol) in dioxano (40 ml) at 0 °C under nitrogen atmosphere, was added HGl (40 ml, 4M in dioxane). The reaction soiution was stirred at 0 °G for 1 h, then concentrated under reduced pressure. The resulting mixture was diluted with DCM (600 ml.) and saturated sodium bicarbonate aqueous soiution (400 mL). The organic phase was separated and washed with brine (500 mL x 2), then concentrated under reduced pressure to afford (6 3 S,4S,Z)-4-amino- 1 '-ethyl-1 2 -(2-((S)-1-methoxyeihyl)-5-(4-methylpiperazin-1-yl)pyr idin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,8 4 ,6 5 ,6 6 - hexahydro-1 1 W-8-oxa-2(4,2)-thiazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-5,7-dione (7.2 g, 94.8% yield) as solid. LCMS (ESI): m/z [M+H] calc’d for CagHjaNeCUS 728.4; found 729.3.

Step 2, A mixture of (6 3 S,4S,Z)-4-amino-1 '-ethyl-1 2 -(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimeth l·6 1 ,6 2 ,6 3 ,6 4 6 5 ,6 6 -hexahydro-1 1 /7-8-oxa-2(4,2)-†hiazola- 1 (5.3)-indola-6(1 ,3)-pyridazinacycloundecaphane-5,7-dione (6 g, 8.23 mmol) and lithium N- (dimethylcarbamoyl)-N-methyl-L-valinate (4.28 g, 20.58 mmol) in DMF (80 mL),was added DlEA (53.19 g, 411 .55 mmol). The reaction mixture was stirred for 5 minutes, then added GIF (3.43 g, 12.35 mmol) in one portion. The resulting solution was stirred at 25 °C for 1 h, then quenched with water (100 mL), extracted with EtOAc (300 mL). The organic layer was separated and washed with saturated ammonium chloride aqueous solution (100 mL x 3) and water (100 mL x 2). The combined organic layers were concentrated under reduced pressure. The residue was purified by reverse phase chromatography to afford (2S)-N- ((6 3 S,4S,Z)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyr idin-3-yl)-10,10-dimethyl-5,7- dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 , /7-8-oxa-2(4,2)-thiaz.oia-1 (5,3)-lndola-6(1 ,3)- pyridazlnacycloundecaphane-4-yl)-3-methyl-2-(1 ,3,3-trimethylureidc)butanamide (2.5 g, 33.2% yield) as a solid 1 H NMR (400 MHz, DMSO-d6) d 8.52 - 8.34 (m, 3H), 7.82 (s, 1H), 7.79 - 7.69 (m, 1H), 7.60 - 7.50 (m, 1H), 7.26 - 7.16 (m, 1H), 5.64 - 5.50 (m, 1H), 5 20 - 5.09 (m, 1H), 4.40 - 4.08 (m, 5H), 3.92 - 3.82 (m, 1H), 3.66 - 3.50 (m, 2H), 3.37 - 3.35 (m. 1H), 330 - 3.28 (m, 1H), 3.28 - 3.20 (m, 4H), 3.19 - 3.15 (m, 3H), 3.12 - 3. 04 (m, 1H), 2.99 - 2 89 (m, 1H), 2.81 (s, 6H), 2.77 (s, 4H), 2 48 - 2 38 (m, 5H), 2.22 (s, 3H), 2.16 - 2.04 (m, 2H), 1 .88 - 1 .78 (m, 2H), 1 .60 - 1 .45 (m, 2H), 1 39 - 1 .29 (m, 3H), 0.97 - 0.80 (m, 12H), 0.34 (s, 3H). LCMS (ESI): m/z. [M+H] calc'd for C 4 8H 68 N!o0 6 S 912 5; found 913.6.

Example A265. The synthesis of N-((6 3 S,4S,Z)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyI)pyridin-3- yi)-10,1Q-dimethyl-5,7-dioxo-6 1 ,8 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 f -S-0xa-2{4,2)-thia;zala-1{5,3)-indoia- 6(1, 3)-pyridaz!nacycloundecapharie-4-yl)-4-rnethylp!peraz!ne 1-carboxamide

Step 1. To a stirred solution of 1-methylpiperazine (100 mg, 1 .148 mmol) and Pyridine (275.78 mg, 3.44 mmol) in DCM (3 mL) were added BTC (112.5 mg, 0.38 mmol) in DGM (1 ml_) dropwise at 0 “C under nitrogen atmosphere. The reaction was stirred for 2 hh 0 c C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure to afford 4-methylpiperazine-1-carbonyl chloride (250 mg, crude) as an oil.

Step 2. To a stirred solution of Intermediate 8 (100 mg, 0.16 mmol) and pyridine (100 mg, 1 272 mmol) in AGN (2 mL) was added 4-methylpiperazine-1-carbonyl chloride (38 67 mg, 0.24 mmol) dropwise at 0 c 'C under nitrogen atmosphere. The reaction mixture was stirred for 2 hh at 0 °C under nitrogen atmosphere. The resulting mixture was diluted with water (100 mL) and extracted with EtOAc (100 mL x 3) The combined organic layers were washed with brine (50 mL x 3), dried over anhydrous Na 2 SO 4 , then filtered and concentrated under reduced pressure. The residue was purified by reverse flash chromatography to give N-((6 3 S,4S,Z)-1 '-ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5 ,7- dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 J 6 6 -hexahydro-1 , /7-8-oxa-2(4,2)-thiazoia-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-4-yl)-4-methylpiperazine-1-carbox amide (20 mg, 16.7% yield) as a solid. 1 H NMR (400 MHz, DMSO-cfe) d 8.76 (dd, J = 4.8, 1.7 Hz, 1H), 8.50 is, 1H), 8.14 (d, J= 2.5 Hz, 1H), 7.79 (d, J == 9.1 Hz, 2H), 7.77 - 7.72 (m, 1H), 7.58 (d, J = 8.6 Hz, 1H), 7.52 (dd, J = 7.7, 4.7 Hz, 1H), 6.82 (d, J == 9.0 Hz, 1H), 5.32 (t, J = 9.0 Hz, 1H), 4.99 (d, J= 12.1 Hz, 1H), 4.43 - 4.02 (m, 5H), 3.57 (d, J = 3 1 Hz,

2H), 3.26 (d, J = 8.4 Hz, 6H), 2.97 (d, J= 14.3 Hz, 1H), 2.80 - 2.66 (m, 1H), 2.55 (s, 1H), 2.40 (d, J = 14.4 Hz, 1H), 2.32 (d, J= 5.9 Hz, 4H), 2.21 (s, 3H), 2.09 (d, J - 12.1 Hz, 1H), 1.77 (d, J - 18 8 Hz, 2H), 1 52 (dd, J = 11 8, 5.4 Hz, 1H), 1 .37 (d, J = 6.0 Hz, 3H), 1 .24 (s, 1H), 0.90 (s, 3H), 0.85 (t, J = 7.0 Hz, 3H), 0.32 (s, 3H). LG MS (ESI): m/z [M+H] calc’d for C^oHszNsOsS 756 38; found 757.3.

Example A598. The synthesis of (2S)- -((6 3 S ! 3S,4S,2)-1 1 -efhyl-3-methoxy-1 2 -(2-{iS -1-

Step 1. A mixture of benzyl (2S)-3-methyl-2-(methylamino)butanoaie (500 mg, 2.26 mmol) and dimethylcarbamyi chloride (1 .215 g, 11 .3 m ol) in THF (5 mL), was added TEA (2.286 g, 22.59 mmol) and DMAP (276.02 mg, 2.26 mmol) in portions under nitrogen atmosphere. The reaction mixture was stirred at 65 °C for 12 hh under nitrogen atmosphere, then quenched with water (100 mL) and was extracted with EtOAc (50 mL x 3). The combined organic phase was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by reverse phase chromatography to afford benzyl N-(dimethylcarbamoyl)-N-methyl-L-valinate (400 g, 583% yield) as an oil. LCMS (ESI): /z [M+H] calc’d for C 1 SH 2 4N 2 O 3 292.2; found 293.1 . Step 2. A mixture of benzyl N-(dimethylcarbamoyl)-N-methyl-L-vaiinate (400 mg, 1 .37 mmol) and palladium hydroxide on carbon (400 mg, 2.85 mmol) in MeOH (10 mL) was stirred for 4 hh under hydrogen atmosphere. The reaction mixture was filtered and the filter cake was washed with MeOH (100 ml. x 3). The filtrate was concentrated under reduced pressure to afford N-(dimethylcarbamoyi)-N-methyl- L-valine (200 g, crude) as an oil. LG MS (ESI): m/z [M+H] calc'd for C9H18N 2 O 3 202.1 ; found 203.1 .

Step 3. A solution of 4-bromo-1 ,3-thiazole-2-carboxylic add (10 g, 48.07 mmol) in DCM (100 mL), was added oxaiyi chloride (16.27 L, 192.28 mmol) and DMF (0.11 mL, 1 .53 mmol) at 0 °C. The reaction was stirred for at room temperature for 2 hh, then concentrated under reduced pressure to afford 4-bromo-1 ,3-thiazole-2-carbonyl chloride (10.8 g, crude).

Step 4, A solution of ethyl 2-[(diphenylmethylidene)amino]acetate (12.75 g, 47.69 mmol) in THF (100 mL) at -78 °C, was added Li HMDS (47.69 mL, 47.69 mmol), and stirred at -40 °G for 30 minutes. Then the reaction mixture was added a solution of 4-bromo-1 ,3-thiazole-2-carbonyl chloride (10.8 g,

47.69 mmol) in THF (100 mL) at -78 °C and stirred at room temperature for 12 hh. The resulting mixture was quenched with water (100 mL), extracted with EtOAc (100 L x 3). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford ethyl 3-(4-bromothiazoi-2-yl)-2-((diphenylmethylene)amino)-3-oxopr opanoate (27 g, crude) as a solid. LCMS (ESI): m/z [M+H] calc’d for GaiHiyBrNaOaS 456.0; found 457.0.

Step 5, A solution of ethyl 3-(4-bromothiazol-2-yl)-2-((diphenylmethylene)amino)-3- oxopropanoate (20 g, 43.73 mmol) in THF (150 mL) at 0 °C, was added 1 M HCi (100 mL) and stirred at room temperature for 2 hh. The resulting solution was concentrated and washed with ethyl e†her(2QQ L x 2). The water phase was adjusted pH to 8 with sodium bicarbonate solution, then extracted with EtOAc (100 mL x 3). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford ethyl 2-amino-3-(4-bromothiazol-2-yl)-3-oxopropanoate as an oil (9 g, crude). LCMS (ESI): m/z [M+H] cale’d for CsHaBrNaOisS 292.0; found 292.9.

Step 6, A solution of ethyl 2-amino-3-(4-bromothiazol-2-yl)-3-oxopropanoate (10 g, 34.11 mmol) in MeOH (200 mL) at 0 °G, was added benzaldehyde (724 g, 68.23 mmol), zinc chloride (9.3 g, 68.23 mmol) and NaBHeCN (429 g, 68.23 mmol). The reaction was stirred at room temperature for 2 hh, then quenched with water (100 mL) and concentrated. The resulting mixture was extracted with EtOAc (100 ml x 3). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by siiiea gei chromatography to afford ethyl 3-(4-bromothiazol-2-yl)-2-(dibenzylamino)-3-oxopropanoate as a solid (8.4 g, 52 % yield). LCMS (ESI): m/z [M+H] ealc’d for G22H 2 iBrN 2 03S 472.1 ; found 473.0.

Step 7, A mixture of ethyl 3-(4-bromothiazol-2-yl)-2-(dibenzylamino)-3-oxopropanoate (5 g,

10.56 mmol) and (R,R)-TS-DENEB (1 375 g, 2.11 mmol) in DCM (100 mL), was added HCOOH (1 .99 mL, 43.29 mmol) and diethylamine (2.2 mL, 2.11 mmol) dropwise at room temperature under nitrogen atmosphere. The reaction mixture was stirred at 50 °G for 12 hh under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford ethyl (2S,3S)-3-(4-bromothiazol-2-yl)-2-(dibenzyiamino)-3-hydroxyp ropanoate (3.148 g, 60% yield) as an oil. LCMS (ESI): rn/z [M+H] calc’d for C 2 2H 2 3BrN 2 03S 474.1 ; found 475.0.

Step 8. A mixture of ethyl {2S,3S)-3-{4-bramothiazoi-2-yl)-2-(dibenzyiamino)-3- hydroxypropanoate (1 g, 2.1 mmol) and Ag2O (4.88 g, 2 .06 mmol) in acetonitrile (10 mL), was added iodomethane (3.58 g, 25.22 mmol) in portions. The reaction mixture was stirred at 50 °C for 12 hh, then filtered. The filter cake was washed with MeOH (50 mL x 2). The filtrate was concentrated under reduced pressure to afford ethyl (2S,3S)-3-(4-bromothiazol-2-yl)-2-(dibenzyiamino)-3-methoxyp ropanoate (1 .06 g, crude) as an oil. LCMS (ESI): m/z [M+H] eaic’d for C 2 3H 2 sBrN 2 O 3 S 488.1 ; found 489.3.

Step 9. A mixture of ethyl (2S,3S)-3-(4-bromothiazol-2-yl)-2-(dibenzylamino)-3- hydroxypropanoate (1 .06 g, 2.3 mmol) in MCI (10 ml, 8 M) was stirred at 80 °C for 12 hh and concentrated by reduced pressure. The residue was purified by reverse phase chromatography to afford (2S.3S)-3-(4-bromothiazol-2-yl)-2-(dibenzylamino)-3-methoxyp ropanoic acid (321 g, 31 .7% yield) as a solid LCMS (ESI): /z [M+H] calc’d for C 2 iH 2 iBrN 2 03S 460.1 ; found 461 .1 .

Step 10. A solution of (2S,3S)-3-(4-bromothiazol-2-yl)-2-(dibenzylamino)-3-methoxyp ropanoic acid (4.61 g, 10 mmol) in DCM (100 mL) at 0 °C was added methyl (35)-1 ,2-diazinane-3-carboxylate bis(triiluoroace†ie acid) salt (3.72 g, 15 mmol), NMM (10.1 mL. 100 mol), EDCI (3.8 g, 20 mmol) and HOBt (5.39 g, 39.89 mmol). The solution was warmed to room temperature and stirred for 1 h. The reaction was then quenched with H 2 O (100 mL) and was extracted with EtOAc (100 mL x 3). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressured. The residue was purified by silica gel column chromatography to give methyl (S)-1- ((2S,3S)-3-(4-bromothiazoi-2-yl)-2-(dibenzylamino)-3-methoxy propanoyi)hexahydropyridazine-3- carboxylate (5.11 g, 90% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 2 THgiBrN+O+S 587.1 ; found 586.1.

Step 11. A solution of methyl (S)-1-((2S,3S)-3-(4-bromothiazol-2-yl)-2-(dibenzylamino)-3- me†hoxypropanoyl)hexahydropyridazine-3-carboxylate (5.11 g, 9 mmol) in THE (100 mL)/H 2 O (100 L) was added LIOH (1 .81 g, 45 mmol) under N 2 atmosphere and the resulting mixture was stirred for 2 hh at 25 ,3 C. The resulting mixture was concentrated under reduced pressure, the residue was acidified to pH 5 with HCL (1 N). The aqueous layer was extracted with DCM (50 mL x 3). The combined organic phase was concentrated under reduced pressure to give (S)-1-((2S,3S)-3-(4-bromothiazol-2-yl)-2-(dibenzylamino)-3- methoxypropanoyi)hexahydropyrldazine-3-carboxylic acid (4.38 g , 85% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for CseH 2 gBri^CbS 572.1 ; found 573.1 .

Step 12. A mixture of (S)-1-((2S,3S)-3-(4-bromothiazol-2-yl)-2-(dibenzylamino)-3- methoxypropanoyi)hexahydropyrldazine-3-carboxylic acid (1.15 g, 2 mmol) and (S)-3-(1-ethyl-2-(2-(1- methoxyethyl)pyridin-3-yl)-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-1H-indol-3-yl)-2,2- dimethylpropan-1-ol (985 mg, 2 mmol ) in DCM (50 mL), was added DIEA (1.034 g, 8 mmol), EDCI (1.15 g, 558.3 mmol), HOBT (270.2 mg, 2 mmol). The reaction solution was stirred at 25 °C for 16 hh. The resulting mixture was diluted with DCM (200 mL), washed with water (50 ml. x 2) and brine (50 mL x 3) and dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford 3-(1-ethyl-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-5- (4,4,5,5-tetramethyM ,3,2-dioxaborolan-2-yl)-1H-indol-3-yl)-2,2-dimethylpropyl (S)-1-((2S,3S)-3-(4- bromothiazol-2-yl)-2-(dibenzylamino)-3-methoxypropanoyl)hexa hydropyridazine-3-carboxylate (1.13 g, 54% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 5 sHesBBrNeOzS 1046.4; found 1047.4,

Step 13. A mixture of 3-{1-ethyl-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-5-(4,4,5,5 -tetramethyl- 1 ,3,2-dioxaborolan-2-yl)-1 W-indol-3-yl)-2,2-dimethylpropyl (S)-1-((2S,3S)-3-(4-bromothiazol-2-yl)-2- (dibenzylamino)-3-methoxypropanoyl)hexahydropyridazine-3-car boxylate (250 mg, 0.24 mmol) and Pd(DtBPF)Cl2 (15.55 mg, 0.024 mmol) in dioxane (5 mL) and water (1 mL), was added K3PO4 (126.59 mg, 0.6 mmol) in portions under nitrogen atmosphere. The reaction mixture was stirred at 80 °C for 2 hh under nitrogen atmosphere. The resulting mixture was diluted with water (20 mL) and extracted with EtOAc (10mL. x 3), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford (6 3 S,3S,4S,Z)-4- (dibenzyiamino)-1 1 -ethyl-3-methoxy-1 ? -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl- 6’ ,6 2 ,6 3 ,6 4 ,6 6 ,6 s -hexahydro-1 1 H-8-oxa-2(4,2)-thiazoia-1 (5,3)-indola-6(1 ,3)-pyridazlnacycloundecaphane-

5.7-dione (137 mg, 44.38 %) as a solid. LCMS (ESI): rrt/z [M+H] calc’d for C 4 9H56N6O5S 840.4; found 841 5.

Step 14. A mixture of ((6 3 S,3S,4S,2)-4-(dibenzyiamino)-1 1 -ethyl-3-methoxy-1 2 -(2-((S)-1- mefhoxyethyl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 M-8-oxa-2(4,2)-thiazola-1 (5,3)- indola-6(1 ,3)-pyridazinacycloundecaphane-5,7-dione (100 mg, 0.12 mmol) and Pd/C (253.06 mg, 2.38 mmol) in MeOH (10 mL), was added HCGONPL (149.94 mg, 2.38 mmol) in portions. The reaction mixture was stirred at 60 °C for 6 hh under hydrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with MeOH (100 mL x 10). The filtrate was concentrated under reduced pressure to afford (6 3 S,3S,4S,2)-4-amino-1 1 -ethyl-3-methoxy-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl- 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-thiazola-1 (5,3) -indola -6(1 ,3)-pyridazinacycloundecaphane-

5.7-dlone (56 mg, crude) as an oil. LCMS (ESI): m/z [M+H] calc’d for C 3 5H44N15Q5S 660.3; found 661 .2.

Step 15. A mixture of (6 3 S,3S,48,Z)-4-amino-1 L ethyl-3-me†hoxy-1 2 -(2-((8)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-6 l ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 W-8-oxa-2(4,2)-thiazoia-1 (5,3)- indola-6(l ,3)-pyridazinacycloundecaphane-5,7-dione (56 g, 0.085 mmol) and N-(dimethylcarbamoyl)-N- methyl-L-valine (51 .42 mg, 0.25 mmol) in DMF (2 mL), was added 2-Chioro-1 ,3-dimethylimidazoiidinium hexafiuorophosphate (47.55 mg, 0 17 mmol) and DIEA (547.62 mg, 4.24 mmol) in portions. The reaction mixture was stirred for 12 hh. The resulting mixture was purified by reverse phase chromatography to afford (2S)-N-((6 3 S,3S,4S,2)-1 l -ethyl-3-methoxy-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-

5.7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,S s ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-thiazola-1 (5,3)-indola-6(1 ,3)- pyridazlnacycloundecaphane-4-yl)-3-methyl-2-(1 ,3,3-trimethylureido)butanamide (1 .5 mg, 2.06 % yield) as a solid. ' H NMR (400 MHz, Methanol-d4) d 8.74-8.77 (m, 1H), 8.61 (d, J - 1.6 Hz, 1H), 7.99 - 7.87 (m, 1H), 773 - 7.66 (m, 1H), 7.68 (s, 1H), 7.60 - 7.55 (m, 1H), 7.49 (d, J - 8.7 Hz, 1H), 7.31 (d, J - 51.0 Hz, OH), 5 89 (s, 1H), 495 (s, 1H), 4.43 (d, J - 13.0 Hz, 1H), 436 (q, J - 6.2 Hz, 1H), 4.33 - 4.19 (m, 2H),

4.10 - 4.03 (m, 1H), 4.03 (d, J = 11 .2 Hz, 1H), 3.78 - 3.67 (m, 2H), 3.65 (s, OH), 3.46 (s, 3H), 3.34 (s, 4H), 3.01 (d, J = 10.3 Hz, 1H), 2.93 (s, 6H), 2.88 - 2.81 (m, 1H), 2.78 (s, 3H), 2.70 - 2.60 (m, 1H), 2.23 - 2.01 (m, 2H), 2.03 (s, OH), 1.99 (d, J = 13.3 Hz, 1H), 1.91 - 1.74 (m, 1H), 1.69 - 1.54 (m, 1H), 1.45 (d, J = 6.2 Hz, 3H), 1.37 - 1.32 (m, 1H), 1.28 (s, 1H), 0.94 (p, J = 6.7 Hz, 12H), 0 51 (s, 3H), 0.10 (s, 1H). LCMS (ESI): m/z [M+H] calc’d for C 4 4H60N8G7 8444; found 845.4.

Example A286. The synthesis of (1 S,2S)-N -((6 3 S,4S ! Z)-T- e thyl-1 2 -(2-i(S)-1- methGxyethyl)pynd ! n-3-yf)-6 4 ,10,10-trimethyl-5,7 " d ! 0X0 8 1 ,6 2 ,6 3 ,6 4 ,8 5 ,8 6 -hexahydro-T i H-8-Gxa " 2(4,2)- thiazola”1(5,3) irsdGla-6(1,3)-pyridaz!nacyciGyndecaphane”4-yl)-2-methytey cloprGparie-1” carboxamide

Step 1. A solution of Intermediate 8 (8 g, 10.95 mmol) in HCI (200 rriL, 4M in 1 ,4-dioxane) was stirred at 0 °C for 2 hh, then concentrated under reduced pressure. The resulting mixture was diluted with DGM (60 mL) and saturated NaHC03 aqueous solution (40 mL). The organic phase was separated and washed with brine (50 mL x 2) and concentrated under reduced pressure to give (6 3 S 4S,2)-4-amino-1 1 - ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 W-8-oxa-2(4,2)- thiazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-5,7-dione (10.3 g, crude) as solid. LGMS (ESI): m/z [M+H] calc’d for C 3 4H42N6O4S 630.3; found 631 .2.

Step 2. A stirred solution of (6 3 S,4S,Z)-4-amino-1 1 -ethyl-1 2 -(2-((S)-l -me†hoxyethyl)pyridin-3-yl)- 10,10-dimethyl-6 1 ,6 2 ,6 3 6 4 6 s ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-thiazoia-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-5,7-dione (8 g, 12.68 mmol) In DMF (50 mL) at 0 °C, was added DlEA (9.83 g, 78.09 mmol), (1 S,2S)-2-methylcyclopropane-1-carboxylic acid (1 .52 g, 15.22 mmol) and HATU (14.47 g, 38.05 mmol). The reaction mixture was stirred af 0 “C for 2 hh and concentrated under reduced pressure. The residue was purified by reverse phase chromatography to afford (1 S,2S)-N -((6 3 S,4S,2)-1 ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5 ,7-dioxo-6 , ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hexahydro-1 1 H- 8- oxa-2(4,2)-thiazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)-2-methylcyclopropane-1- carboxamide (6.84 g, 56.37% yield) as a solid. 1 H NMR (400 MHz, DMSO-cfe) d 8.79 (dd, J- 4.7, 1 .9 Hz, 1H), 8.59 - 8.40 (m, 2H), 7.95 - 7.86 (m, 1H), 7.82 - 7.71 (m, 2H), 7.66 - 7.53 ( , 2H), 5.57 (t, J - 9.0 Hz, 1H), 5.07 (s, 1H), 4.41 - 4.28 (m, 2H), 4.25 (d, J- 12 4 Hz, 1H), 4.17 (d, J - 10.8 Hz, 1H), 4.09 (d, J - 7.2 Hz, 1H), 3.58 (s, 2H), 3 32 (d, J - 14.6 Hz, 1H), 3.28 (s, 3H), 3.16 (dd, J - 14.7, 9.1 Hz, 1H), 2.95 (d, J = 14.4 Hz, 1H), 2.75 (m, J = 12.1 , 7.1 Hz, 1H), 2.43 (d, J = 14.4 Hz, 1H), 2.13 - 2.00 (m, 1H), 1.76 (d, J = 22.0 Hz, 2H), 1.60 - 1.44 (m, 2H), 1.38 (d, J == 6.1 Hz, 3H), 1.07 (d, J = 1.9 Hz, 4H), 0.86 (dd, J = 14.1 , 7.1 Hz, 7H), 0.59 - 0.49 (m, 1H), 0.34 (s, 3H). LCMS (ESI): m/z [M+H] calc’d for Csg^sNeOsS 712 3; found 713.2.

Example A613, The synthesis of -{{2S)-1-({(8 3 S,4S,Z)-1 1 -ethyl 1 2 -{2-{(S)-1- rriethoxyethyl )pyndiri-3-yl) 10,10-di ethyl -S,7-dioxo-6 1 ,8 2 ,6 3 ,6 4 ,6 5 ,6 S hexahydro-1 1 #f-8 Oxa-2{4,2) thiazola-1 {5,3)-pyrrolo[3,2-b]jpyr!d a-6{1,3)-pyndaz!iiacyeloiirsdecapharie-4-yl)am o)-3-meihyl-1- oxobutan-2-yl)-3- ethoxy-M meihylazefidsrie-1-carboxamide t

A 0

Step 1. A mixture of methyl (S)-3-(4-bromothiazol-2-yl)-2-((tert- butoxycarbonyl)amino)propanoate (920 mg. 2.5mmol), 4,4,5,5-te†ramethyl-2-(4.4,5,5-tetramethyl-1 ,3,2- dioxaboroian-2-yl)-1 ,3,2-dioxaborolane (1.8 g, 6.3 mmol), x-Phos (180 g, 0.5 mmol), Pda(dba)3- chloroform (130 mg, 0.13 mmol) and potassium acetate (740 g, 7.5 mmol) in dioxane (25 mL) in a sealed tube under N? atmosphere, was stirred at 110 °C for 8 hh to afford crude methyl {S)-2-((tert- butoxycarbonyl)amino)-3-(4-(4,4,5,5-tetramethyl-l ,3.2-dioxaborolan-2-yl)thiazol-2-yl)propanoate as a solution. LCMS (ESI): m/z [M+H] calc’d for C 1 sHsaBNsOeS 412.2; found 331 .1 .

Step 2. A mixture of 5-chloro-1H-pyrroio[3,2-b]pyridine-3-carbaldehyde (7 g, 39 mmol) in MeOH (140 mL) under N? atmosphere, was added NaBI-U (2.9 g, 78 mmol) at 0 “C. The reaction mixture was stirred at 10 °C for 2 hh and concentrated under reduced pressure. The residue was diluted with EtOAc (200 mL), washed with brine (25 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gei column chromatography to afford (5-chloro-1 H- pyrrolo[3,2-£>]pyridln-3-yl)methanoi (3.5 g, 55% yield) as a solid LCMS (ESI): m/z [M+H] ealc’d for C8H7CIN 2 O 182.0; found 183.0.

Step 3. A mixture of (5-chloro-1H-pyrrolo[3,2-b]pyridin-3-yl)methanoi (3.5 g, 19 mmol) and ((1- methoxy-2-methylprop-1-en-1-yl)oxy)trimethylsilane (6.7 g, 38 mmol) in THF (50 mL), was dropwise added TMSOTf (3.8 g, 17.1 mmol) at 0 °C The reaction mixture was stirred at 5 °C for 2 hh, then diluted with EtOAc (100 mL). washed with saturated NaHCO 3 aqueous (50 mL), and brine (50 ml. x 2). The organic layer was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford methyl 3-(5-chloro-1H-pyrroio[3,2- i>]pyridin-3-yl)-2,2-dimethylpropanoate (3 g, 59% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 1 3H 15CIN 2 O 2 266.1 ; found 267.1 .

Step 4, A mixture of methyl 3-(5-chloro-1H-pyrrolo[3,2-b]pyridin-3-yl)-2,2-dimethylpropa noate (3 g, 11 mmol) in anhydrous THF (50 mL) at 0 °C, was added AgOTf (4.3g, 17 mmol) and I2 (2.9 g, 11 mmol). The reaction mixture was stirred at 0 °C for 2 hh, then quench with cone. Na2SC>3 (20 mL), diluted with EtOAc (50 mL) and filtered. The filtrate was washed with brine (50 mL). The organic layer was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified with silica gel column chromatography to aiford methyl 3-(5-chloro-2-iodo-1H-pyrroio[3,2-£j]pyridin-3-yl)-2,2- dimethylpropanoate (2.3 g, 52% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 1 3H14CIIN 2 O 2 393.0; found 392.0

Step 5. A mixture of methyl 3-(5-chloro-2-iodo-1H-pyrrclc[3,2-£>]pyridin-3-yl)-2,2- dimethylpropanoate (2.3 g, 5.9 mmol), 2-(2-(2-methoxyethyl)phenyl)-4,4,5,5-tetramethyl-1 ,3,2- dioxaborolane (1 .6 g, 7.1 mmol) and K 2 CO 3 (2.4 g, 18 mol) in dioxane (25 mL) and water (5 mL) under N 2 atmosphere, was added Pd(dppf)Cl2-DCM (480 mg, 0.59 mmol). The reaction mixture was stirred at 70 °C for 4 hh, then diluted with EtOAc (200 L) and washed with brine (25 mL). The separated organic layer was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford methyl (S)-3-(5-chloro-2-(2-(1-methoxyethyl)pyridin- 3-yl)-1H-pyrrolo[3,2-£>]pyridin-3-yl)-2,2-dimethylpropan oate (2 g, yield 84%) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 2 1H 2 4CIN3O 3 401 .2; found 402.2.

Step 6. A mixture of methyl (S)-3-(5-chloro-2-(2-(1-methoxyethyl)pyridin-3-yl)-1H-pyrrol o[3,2- b]pyridin-3-yl)-2,2-dimethylpropanoate (2 g, 5 mmol), cesium carbonate (3.3 g, 10 mmol) and Etl (1 .6 g, 10 mmol) in DMF (30 mL) was stirred at 25 °C for 10 hh. The resulting mixture was diluted with EtOAc (100 mL), washed with brine (20 mL x 4). The separated organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford methyl (S)-3-(5-chloro-1-ethyl-2-(2-(1-methoxyethyl)pyridin-3-yl)-1 H-pyrrolo[3,2- b]pyridin-3-yl)-2,2-dimethylpropanoate as two dlasteroomers (P1 : 0.7 g, 32% yield; P2: 0.6 g, 28% yield) both as a solid. LCMS (ESI): m/z [M+H] calc’d for C 2 3H 2 8CIN3O 3 429.2; found 430.2

Step 7. A mixture of methyl (S)-3-(5-chloro-1-ethyl-2-(2-(1-methoxyethyl)pyridin-3-yl)-1 H- pyrrolo[3,2-£)]pyridin-3-yl)-2,2-dimethylpropanoate (P2, 1 .2 g, 2.8 mmol) in anhydrous THF (20 mL) at 5 °C, was added LiBH-t (120 g, 5.6 mmol). The reaction mixture was stirred at 60 °C for 4 hh, then quenched with cone. NH4CI (20 mL), diluted with EtOAc (50 mL) and washed with brine (30 mL). The organic layer was separated, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified with silica gel column chromatography to afford (S)-3-(5-chloro-1-ethyl-2-(2-(1- methoxyethyl)pyridin-3-yl)-1H-pyrrolo[3,2-£>]pyridin-3-y l)-2,2-dimethylpropan-1-ol (1 g, 89% yield) as a solid LCMS (ESI): m/z [M+H] calc’d for GaaHssCiNaOa 401 .2; found 402 2.

Step 8, A mixture of solution from Step 1 (380 mg, crude, 1 mmol) in dioxane (10 ml..) and water (2 ml.), was added (S)-3-(5-chloro-1-ethyl-2-(2-(1-methoxyethyl)pyridin-3-yl)-1 H-pyrroio[3,2-b]pyridin-3-yl)-

2.2-dimethylpropan-1-ol (200 mg, 0 5 mmol), potassium carbonate (200 mg, 1 .5 mmol) and Pd-118 (30 mg, 0.05 mmol). This reaction mixture was stirred at 70 °C for 3 hh, then diluted with EtGAc (40 rrsL), filtered. The filtrate was washed with brine , dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified with silica gel column chromatography to afford methyl (S)-2- ((tert-butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1- methoxyethyl)pyridin-3-yl)-1H-pyrrolo[3,2-b]pyridin-5-yl)thi azol-2-yl)propanoate (300 mg, 65% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for Ca- sNsOeS 851 .3; found 652.3.

Step 9. A solution of methyl (S)-2-((fer/-butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3-hydrox y-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1H-py rrolo[3,2-b]pyridin-5-yl)thiazol-2- yl)propanoate (280 mg, 0.43 mmol) in MeOH (4 mL), was added a solution of lithium hydroxide (51 mg, 2.15 mmol) in water (2 mL) at 20 “C. The reaction was stirred at 20 c C for 5 hh, then adjusted to pH = 3~4 with HCI (1 N). The resulting mixture was diluted with water (30 mL) and extracted with EtOAc (15 L c 3). The combined organic phase was washed with brine (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (S)-2-((ierf-biJtoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3- hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin -3-yl)-l /--pyrrolo[3 s 2-b]pyridin-5-yl)thiazol-2- yi)propanoic acid (280 g, crude) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 3 3H43N5O15S 637.3; found 638.3.

Step 10. A solution of (S)-2-((tert-butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3-hydrox y-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1H-py rroio[3 s 2-b]pyrldln-5-yl)thiazol-2-yl)propanoic acid (274 mg, 0.43mmol) and methyl (S)-hexahydropyridazine-3-carboxylate (280 mg, 0.64 mmol) In DMF (3 mL) at 5 °C, was added a solution of HATU (245 mg, 0.64 mmol) and DlEA (555 mg, 4.3mmol) in DMF (2 mL). The reaction was stirred for 1 h, then diluted with EtOAc (20 mL) and water (20 mL). The organic layer was separated and washed with water (20 mL x 3) and brine (20 mL), dried over anhydrous sodium sulfate, filtered concentrated under reduced pressure. The residue was purified by silica gel chromatography to give methyl (S)-1-((S)-2-((feff-butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3 -hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1H-py rrolo[3,2-b]pyridin-5-yl)thiazol-2- yi)propanoyl)hexahydropyridazine-3-carboxylate (230 g, 70% yield) as solid. LCMS (ESI): m/z [M+H] calc’d for C 3 9H53N7O7S 763.4; found 764.3.

Step 11. A solution of methyl (S)-1-((S)-2-((tert-butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3 -hydroxy-

2.2-dimothylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl )-1H-pyrrolo[3,2-b]pyridin-5-yl)thiazol-2- yi)propanoyl)hexahydropyridazine-3-carboxylate (230 mg, 0.3 mmol) in DCE (3 ml), was added trimethyltin hydroxide (300 mg, 1.4 mmol) under N 2 atmosphere. The reaction was stirred at 65 °G for 16 hh, then concentrated under reduced pressure. The residue was diluted with EtOAc (20 mL), washed with water (20 mL) and brine (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford (S)-1-((S)-2-((teff-butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3 -hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-1H-py rrolo[3,2-b]pyridin-5-yl)thiazol-2- yl)propanoyi)hexahydropyridazine-3-carboxylic acid (200 mg, crude) as foam. LCMS (ESI): m/z [M+H] calc’d for CaeHsiNyO/S 749.4; found 750.3.

Step 12. A solution of (S)-1-((S)-2-((feff-butaxycarbonyl)amino)-3-(4-(1-ethyl-3-(3 -hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methGxyethyl)pyridin-3-yl)-1H-py rrolo[3.2-b]pyridin-5-yl)thiazol-2- yl)pr0panoyi)hexahydropyridazine-3-carboxylic acid (245 mg, 0.32rrsmol) in DCM (50 mL) at 5 °C, were added HOBt (432 mg, 3.2mmol), EDCI(1 .8 g, 9.6mmol) and DIEA (1 .85 g, 12.8mmol). The reaction mixture was stirred at 20 °C for 18 hh, then concentrated under reduced pressure. The residue was diiuted with EtOAc (20 ml.) and water (20 mL) The organic layer was separated and washed with water (30 mL x 3) and brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography to give tert-butyl ((6 3 S,4S,Z)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-S -dioxo-O 1 ,6 2 ,6 3 ,6 4 ,6 5 ,8 6 -hexahydro-1 1 H-8-oxa-2(4,2)- thiazoia-1 (5,3)-pyrroio[3,2-b]pyridina-6(1 ,3)-pyridazinacycloundecaphane-4-yl)carbamate (100 g, 43% yield) as solid. LCMS (ESI): m/z [M+H] calc’d for CssHigNvOsS 731 .4; found 732.3.

Step 13, A solution of terf-butyl ((6 3 S,4S,Z)-1 1 -ethyl-1 2 -(2-{(S)-1-me†hoxyethyl)pyridin-3-yl)- 10,10-dimethyl-s -dioxo-B 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-thiazola-1 (5,3)-pyrrolo[3,2- b]pyridina-6(1 ,3)-pyridazinacycloundecaphane-4-yl)carbamate (80 mg, 0.11 mmol) in TEA (0.2 mL) and DCM (0.6 L) was stirred at 20 “C for 1 h. The reaction was concentrated to afford (6 3 S,4S,Z)-4-amino- 1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-6 l ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 TT-S-oxa- 2(4,2)-thiazola-1 (5,3)-pyrroio[3,2-b]pyridina-6(1 ,3)-pyridazinacycloundecaphane-5,7-dione (72 mg, 95% yield) as a solid. LCMS (ESI): m/z [M+H] calc’d for C 3 3H41N7O4S 631 .3; found 832.3.

Step 14, A solution of (6 3 S,4S,Z)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10- dimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 s ,6 6 -hGxahydro-1 1 H-8-oxa-2(4,2)-thiazola-1 (5,3)-pyrrolo[3,2-b]pyridina-6(1 ,3)- pyridazinacycloundecaphane-5,7-dione (100 mg, 0.16 mmol) and (2S)-2-[(3-methoxyazetidin-1- yl)carbonyl(methyl)amino]-3-methylbutanoic acid (78 mg, 0.32 mmol) in DMF (5 mL) at 0 °C, was dropwise added a solution of DIEA (820 mg, 4.8 mmol) and HATU (91 g, 0.24 mmol) in DMF (5 mL).

The reaction mixture was stirred at 0°G for 2 hh, then diiuted with EtOAc (50 mL), washed with water (25 mL x 3), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford N-((2S)-1-(((6 3 S,4S,Z)-1 1 -ethyl-1 2 -(2- ((S)-1-methoxyethyl)pyridln-3-yl)-10,10-dimethyl-5,7-dioxo-6 , ,6 2 ,8 3 ,8 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)- thiazola-1 (5,3)-pyrrolo[3,2-b]pyridina-6(1 ,3)-pyridazlnacycloundecaphane-4-yl)amino)-3-methyl-1- oxobutan-2-yl)-3-methoxy-N-methylazetidlne-1-carboxamide (112.9 mg, 82% yield) as a solid. 1 H NMR (400 MHz, CD3OD) 6 8.77-8.75 (dd, J = 4.8, 1 .7 Hz, 1H), 7.96-7.94 (d, J = 8.6 Hz, 1H), 7.89-7.87 (dd, J = 8.4, 2.3 Hz, 2H), 7 77-7.74 (d, J = 8.6 Hz, 1H), 7.58-7.55 (dd, J = 7.8, 4.8 Hz, 1H), 5.73-5.70 (dd, J = 8.0, 2.7 Hz, 1H), 4.41-4.38 (dt, J = 8 5, 43 Hz, 2H), 4.33 - 4.26 (m, 3H), 4 24 - 4 17 (m, 3H), 4.04-4.01 (dd, J = 11 .9, 3.0 Hz, 1H), 3.99-3.96 (m, 1H), 3.89 - 3.83 (m, 2H), 3.53-3.49 (dd, J = 9.7, 7.3 Hz, 2H), 3.46-3.45 (d, J = 3.0 Hz, 1H), 3 35 (s, 3H), 3.34-3 33 (d, J = 4.5 Hz, 3H), 3.28 (s, 1H), 2 89 (s, 3H), 2 78-2.71 (td, J = 13.2, 3.4 Hz, 1H), 2.52-2.48 (d, J = 14.1 Hz, 1H), 2.23 - 2.20 (m, 1H), 2.19-2.11 (d, J = 10.2 Hz, 1H), 1.91-1.88 (d, J = 13.5 Hz, 1H), 1.73-1.70 (dd, J = 9.0, 3.9 Hz, 1H), 1.56 - 1 50 (m, 1H), 1.47-1.46 (d, J = 6.1 Hz, 3H), 0.98 - 0.91 (m, 9H), 0.88 (s, 3H), 0.45 (s, 3H). LCMS (ESI): m/z [M+H] calc’d for C 4 4H59NSO7S 857.4; found 858.3.

Example A579. The synthesis of -{(2¾-1-{({6 3 S,6 4 S,4S,2)-1 i -ethyl-1 2 -(2-({S)-1- methoxyethyl)pynd!n-3-yf)-6'^10,10-frimethyl-5,7 !0X0 8 1 ,6 2 ,6 3 ,6'\8 5 ,8 6 -hexahydro-1 1 H-8-Gxa 2(4,2)- thiaxøla”1(5,3) ø øla 6{1,3)-pyri a^:!nac ciGl!ødeca ane”4-yl)a rio)-3-met yl·1 Oxobl!ϊari-2-yl)-3- methoxy-M-methy aaetidine-1-carboxamide

Step 1, A solution of (S)-4-benzyloxazolidin-2-one (10 g, 56.43 mmol) in THF (100 mL) was purged with nitrogen, was added of n-butyllithium (24.83 mL, 62.08 mmol) at -78 °G under nitrogen atmosphere, then stirred for at -78 °G for 15 minutes. The reaction mixture was added 2-butenoyl chloride (6.49 g, 62.08 mmol). The resulting solution was stirred at -78 ,3 C for 30 minutes, then siowly warmed up to 0 °C and stirred for 15 minutes, quenched with saturated ammonium chloride solution (100 mL). The resulting soiution was extracted with EtOAc (100 mL x 3) and the combined organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford (4S)-4-benzyi-3-[(2£)-but-2-enoyl]-1 ,3-oxazolidin-2-one (1226 g, 88.57 % yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for C 1 4H15NO 3 245.1 ; found 246 1 .

Step 2, A soiution of CuBr-DMS (12.07 g, 58.71 mmol) in THF (120 ml) was purged and maintained nitrogen atmosphere, added of allylmagneslum bromide (58.71 mL, 58.71 mmol) at -78 °G The reaction was stirred at -60 °C for 30 minutes under nitrogen atmosphere followed by addition of (4 S)- 4-benzyl-3-[(2£)-but-2-enoyl]-1 ,3-oxazolidin-2-one (12 g, 48.92 mmol) at -78 °C. The resulting solution was stirred at -50 °G for 3 more hh, then quenched with saturated ammonium chloride soiution (100 ml.) and extracted with EtOAc (60 mL x 3). The combined organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford (S)-4-benzyl-3-((S)-3-methylhex-5-enoyi)oxazolidin-2-one (13.2 g, 93.89 % yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for C 1 7H 2 1 NO 3 287.2; found 288.2.

Step 3. A soiution of (S)-4-benzyi-3-((S)-3-methylhex-5-enoyl)oxazolidin-2-one ( 3.2 g, 45.94 mmol) in dioxane (200 mL) and water (200 mL), was added 2,4-Lutidine (9.84 g, 91 .87 mrnol) followed with K 2 OSO2H 2 O (1 .69 g, 4.59 mmol) at 0 °C. The reaction solution was stirred at 0 °C for 15 minutes, then was added NalC>4 (39.3 g, 183.74 mmol). The resulting mixture was stirred at 0 °C for 1 h, then extracted with EtOAc (150 mL x 3). The combined organic phase was hydrochloric acid (100 mL x 3), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford (S)-5-({8)-4- benzyl-2-oxooxazoiidin-3-yl)-3-methyl-5-oxopentanai (12.3 g, crude) as an oil. LCMS (ESI): m/z [M+H] calc'd for C 1 eHhsNC 289.1 ; found 290.1 .

Step 4. A solution of (S)-5-((S)-4-benzyl-2-oxooxazoiidin-3-yl)-3-methyl-5-oxopent anal (12.3 g, 42.51 mmol) in THF (200 ml) was purged and maintained with nitrogen atmosphere, then added borane- tetrahydrofuran complex (55.27 mL, 55.27 mmol) at 0 °C. The reaction was stirred at 0 °C for 30 minutes, then quenched with methanol (40 ml). The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford (8)-4-benzyi-3-((S)-5-hydroxy-3- methylpentanoyi)oxazolidin-2-one (9.6 g, 77.51 % yield) as an oil. LCMS (ESI): m/z [M+H] calc'd for C 1 6H 2 1NO4291.1 ; found 292.1.

Step 5, A solution of (S)-4-benzyl-3-((S)-5-hydroxy-3-methylpentanoyi)oxazolidin-2 -one (9.6 g, 32.95 mmol) and CB (16.39 g, 49.43 mmol) in DCM (120 mL) at 0 °C, was added triphenylphosphine (12.96 g, 49.41 mmol). The reaction was stirred at 0 °C for 1 h, then quenched with ice water (100 mL) and extracted with DCM (100 mL x 3). The combined organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford (S)-4-benzyl-3-((R)-5-bromo-3-methylpentanoyl)oxazoiidin-2-o ne (10 g, 85.67 % yield) as an oil. LCMS (ESI): m/z [M+H] calc’d for C 1 sHjoBrNCh 353.1 ; found 354.1

Step 6. A mixture of n-BuLi (2.26 mL, 5.65 mmol) and diisopropylamine (571 .3 mg, 5.65 mmol) in THF (10 mL) under nitrogen at -78 °C, was added a cooled (-78 °C) solution of (S)-4-benzyl-3-((R)-5- bromo-3-methylpentanoyi)oxazolidin-2-one (2 g, 5.65 mmol) In THF (9 mL). The reaction mixture was stirred at -78 c C for 30 minutes, then was added a solution of (£)-N-[(terFbutoxycarbonyl)imino](ierf- butoxy)formamide (1 .3 g, 5.65 mmol) in THF (10 mL), stirred for another 30 minutes at -78 °C. The resulting mixture was added DMPU (16 mL, 132.82 mmol) and warmed up to 0 °C and stirred for 90 minutes, followed by addition of a solution of LiOH H 2 O (1.18 g, 28.12 mmol) in water (20 L). Then THF was removed under reduced pressure. The residue was washed with DCM (80 L x 3). The aqueous phase was acidified to pH 5-6 with HCi (aq.). extracted with mixture of DCM/methancl (80 mL x 3, 10:1). The combined organic layers were dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure. The residue was purified by reverse phase chromatography to afford (3S,4S)-1 ,2- bis(tert-butaxycarbonyl)-4-methylhexahydropyridazine-3-carbo xylic acid (296 mg, 15.22 % yield) as a solid. LCMS (ESI): m/z [M-H] calc’d for C 1 sH 2 eNaOs 344.2; found 343.1 .

Step 7, A mixture of (38,48)-1 ,2-bis(tert-butoxycarbonyl)-4-methylhexahydropyridazine-3- carboxylic acid (289 mg, 0.84 mmol) and (S)-3-(1-ethyl-2-(2-(1-methoxyethyl)pyridin-3-yl)-5-(4.4,5,5 - tetramethyl-1 ,3,2-dioxaboiOian-2-yl)-1H-indol-3-yl)-2,2-dimethylpropan-1- ol (413.24 mg, 0.84 mmol) in DMF (10 mL) at 0 °C, was added DMAP (51 .26 mg, 0.42 mol) and DCC (692 53 mg, 3.36 mmol). The reaction solution was stirred at room temperature for 1 h, then quenched with water/ice (10 mL), extracted with EtOAc (15 mL x 3). The combined organic layers were washed with brine (50 mL x 3), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford 1 , 2 -d i - tert- b u ty I 3-(3-(1-ethyl-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)- 5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-1H-indol-3-yl)-2,2-dimethylpropyl) (3S,48)-4- methyltetrahydropyridazine-1 ,2,3-tricarboxylate (538 mg, 78.3 % yield) as a solid. LCMS (ESI): m/z [M-H] calc’d for C 4 5H67BN4O9818.5; found 819.4. Step 8. A solution of 1 ,2-di-tert-butyl 3-(3-(1-ethyl-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-5- (4,4,5,5-tetramethyM ,3,2-dioxaborolan-2-yl)-1H-indol-3-yl)-2,2-dimethylpropyl) (3S,4S)-4- methyltetrahydropyridazine-1 ,2,3-tricarboxylafe (508 mg, 0.62 mmol) in DCM (25 ml), was added TFA (25 ml.) at 0 °C. The reaction solution was stirred at room temperature for 1 h. The resulting mixture was concentrated to afford 3-(1-ethyl-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-5-(4,4,5,5 -fetramethyl-1 ,3,2- dioxaboroian-2-yl)-1H-indol-3-yl)-2,2-dimethylpropyl (3S,4S)-4-methylhexahydropyridazine-3-carboxylate (508 mg, crude) as an oil. LCMS (ESI): m/z [M-H] calc'd for C 3 5H51 BN4O5 618.4; found 619.3.

Step 9. A solution of 3-(1-ethyl-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-5-(4,4,5,5 -tetramethyl- 1 ,3,2-dioxaborolan-2-yl)-1H-indol-3-yl)-2,2-dimethylpropyl (3S,4S)-4-methylhexahydropyridazine-3- carboxylate (508 mg, 0.82 mmol) and (S)-3-(4-bromothiazol-2-yl)-2-((ferf- butoxycarbonyl)amino)propanoic acid (288.41 mg, 0.82 mmol) in DMF (50 mL) at 0 °C, was added DiEA (1061 .31 mg, 8.21 mmol), HATU (468.35 mg, 1 .23 mmol). The reaction solution was stirred at room temperature for 1 h, then quenched with ice water (30 mL) and extracted with EtOAc (30 mL x 3). The combined organic phase was washed with brine (50 mL x 3), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford 3-(1-ethyl-2-(2-((S)-1-methoxyethyl)pyndin-3-yl)-5-(4,4,5,5- te†ramethyl-1 ,3,2-dioxaborolan-2-yl)-1 H- indol-3-yl)-2,2-dimethylpropyl (3S,4S)-1-((S)-3-(4-bromofhiazol-2-yl)-2-((ie/†- butoxycarbonyl)amino)propanoyl)-4-methylhexahydropyridazine- 3-carboxylate (431 mg, 55.14 % yield) as a solid. LCMS (ESi): m/z [M-H] calc’d for C 4 sHs4BBrN608S 950.4; found 951 .3.

Step 10. A mixture of Pd(DTBpf)Cl2 (27.39 mg, 0.042 mmol) and K3PO4 (89.2 mg, 0.42 mmol) in dioxane (5 mL) and water (1 mL) was purged nitrogen, stirred at 60 °C for 5 minutes under nitrogen atmosphere, then added a solution of 3-(1-ethyl-2-(2-((S)-1-methoxyethyl)pyridin-3-yl)-5-(4,4,5,5 - tetramethyl-1 ,3,2-dioxaborolan-2-yl)-1H-indol-3-yl)-2,2-dimethylpropyl (3S,4S)-1-((S)-3-(4-bromothiazol-2- yl)-2-((tert-butoxycarbonyl)amino)propanoyl)-4-methylhexahyd ropyridazine-3-carboxylaie (200 mg, 0.21 mmol) in dioxane (5 mL) and water (1 mL) at 60 °C. The reaction mixture was stirred at 60 °C for 1 h, then quenched with ice water (5 mL), extracted with EtOAc (15 L x 3). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford terf-butyl ((6 3 S,6 4 S,4S,Z)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)pyridin-3-yl)-6 4 ,10,10-trimethyl-5,7-dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 'H-8-oxa-2(4,2)- thiazola-1 (5,3)-indola-6(1 ,3)-pyridazinacycloundecaphane-4-yl)carbamate (70 mg, 44.72 % yield) as a solid. LCMS (ESi): m/z [M-H] calc’d for C 4 oH 52 N 6 0 6 S 744.4; found 745.4.

Step 11. A solution of tert-butyl ((6 3 S,6 4 S,4S,Z)-1 '-ethyl-1 2 -(2-((S)-1-methoxyeihyl)pyridin-3-yl)-

6 4 .10.10-trimethyl-5,7-dioxo-6' ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-thiazola-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-4-yl)carbamate (70 mg, 0.094 mmol) in dioxane (5 ml), was added HCI in dioxane (5 mL, 4M). The reaction was stirred at room temperature for 1 h, then concentrated under reduced pressure to afford (6 3 S,6 4 S,4S,ZJ-4-amino-1 1 -ethyl-1 2 -{2-((S)-1-methoxyethyl)pyridin-3-yl)-

6 4 .10.10-trimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 W-8-oxa-2(4,2)-thiazoia-1 (5,3)-indola-6(1 ,3)- pyridazinacyclcundecaphane-5,7-dione (124 mg, crude) as an oil. LCMS (ESI): m/z [M-H] calc’d for C 3 6H45N5O4S 644.3; found 645.3.

Step 12. A mixture of (6 3 S,6 4 S,4S,Z)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)pyridin-3-yl)-

6 4 .10.10-trimethyl-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-2(4,2)-thiazola-1 (5,3)-indola-6(1 ,3)- pyridazinacyclcundecaphane-5,7-dione (112 mg, 0.17 mmol) and N-(3-methoxyazetidine-1-carbonyl)-N- methyl-L-valine (50.92 mg, 0.21 mmol) in DMF (3 mL) at 0 °C, was added DIEA (1 .795 g, 13.9 mmol), 2- chloro-1 ,3-dimethylimidazolidinium hexafluorophosphate (72.57 mg, 0.26 mmol). The reaction was stirred at room temperature for 1 h and then filtered. The filtrate was purified by reverse phase chromatography to afford N-((2S)-1-(((6 3 S,6 4 S,4S,2)-1 ’-ethyl-1 2 -(2-((S>1-methoxyethyl)pyridSn-3-yl)-6 4 ,10,10-trimethyl-5,7- dioxo-6 1 ,6 2 ,6 3 ,6 4 ,6 5 J 6 6 -hexahydro-1 ' H-8-oxa-2(4,2)-thiazola-1 (5,3)-indola-6(1 ,3)- pyridazinacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2 -yl)-3-methoxy-N-methylazetidine-1- carboxamide (25.6 mg, 16.92 % yield) as a solid. 1 H NMR (400 MHz, DMSO-ds) 6 8.76 (dd, J ----- 4.7, 1.8 Hz, 1H), 8.60 (s, 1H), 8.30 - 8.20 (m, 1H), 7.86 - 7.70 ( , 3H), 7.61 - 7.50 (m, 2H), 5.57 - 5 43 (m, 1H), 5.07 (d, J = 12.1 Hz, 1H), 439 -4.21 (m, 3H), 4.20 - 4.01 (m, 5H), 3 96 (d, J = 11.1 Hz, 1H), 3.82 (dd, J = 8.9, 3.6 Hz, 1H), 3.77 - 3.71 (m, 1H), 3.63 - 3.55 (m, 2H), 3.35 - 3.27 (m, 2H), 3.24 (s, 3H), 3.23 - 3.14

(m, 4H), 2.93 - 2.79 (m, 2H), 2.70 (s, 3H), 2.15 - 2.01 (m, 1H), 1.83 - 1.61 (m, 2H), 1.38 (d, J= 6.1 Hz, 4H), 0.98 (d, J = 6.4 Hz, 3H), 0.94 - 0.85 (m, 6H), 0.85 - 0.72 (rn, 6H), 0.43 (s, 3H). LCMS (ESI): m/z [M- H] calc’d for C 4 6H82N8O7S 870.4; found 871 .4. The following table of compounds (Table 3) were prepared using the aforementioned methods or variations thereof, as is known to those of skill in the art.

Table 3: Exemplary Compounds Prepared by Methods of the Present Invention

Blank = not determined

Biological Assays Potency assay: pERK The purpose of this assay is to measure the ability of test compounds to inhibit K-Ras in cells.

Activated K-Ras induces increased phosphorylation of ERK at Threonine 202 and Tyrosine 204 (pERK). This procedure measures a decrease in cellular pERK in response to test compounds. The procedure described below in NCi-H358 cells is applicable to K-Ras G12C.

Note: this protocol may be executed substituting other cell lines to characterize inhibitors of other RAS variants, including, for example, AsPC-1 (K-Ras G12D), Capan-1 (K-Ras G12V), or NCI-H1355 (K- Ras G13C).

NCI-H358 cells were grown and maintained using media and procedures recommended by the ATCC. On the day prior to compound addition, cells were plated in 384-well cell culture plates (40 mI/weli) and grown overnight in a 37°C. 5% C02 incubator. Test compounds were prepared in 10, 3-fold dilutions in DMSO, with a high concentration of 10 mM. On day of assay, 40 nl of test compound was added to each well of cell culture plate using an Echo550 liquid handler (LabCyte®). Concentrations of test compound were tested in duplicate. After compound addition, cells were Incubated 4 hours at 37°C, 5% C02 Following incubation, culture medium was removed and cells were washed once with phosphate buffered saline.

In some experiments, cellular pERK level was determined using the AlphaUSA SureFire Ultra p- ERK1/2 Assay Kit (PerkinElmer). Cells were lysed in 25 mI lysis buffer, with shaking at 600 RPM at room temperature. Lysate (10 mί) was transferred to a 384-well Opti-plate (PerkinElmer) and 5 mI acceptor mix was added. After a 2-hour Incubation in the dark, 5 mI donor mix was added, plate was sealed and incubated 2 hours at room temperature. Signal was read on an Envision plate reader (PerkinElmer) using standard AlphaLISA settings. Analysis of raw data was earned out in Excel (Microsoft) and Prism (GraphPad). Signal was plotted vs. the decadal logarithm of compound concentration, and IC 5 o was determined by fitting a 4-parameter sigmoidal concentration response model.

In other experiments, cellular pERK was determined by In-Cell Western. Following compound treatment, cells were washed twice with 200 ml iris buffered saline (TBS) and fixed for 15 minutes with 150 mί 4% paraformaldehyde in TBS. Fixed cells were washed 4 times for 5 minutes with TBS containing 0.1% Triton X-100 (TBST) and then blocked with 100 mI Odyssey blocking buffer (LI-COR) for 60 minutes at room temperature. Primary antibody (pERK, CST-4370, Cell Signaling Technology) was diluted 1 :200 in blocking buffer, and 50 mI was added to each well and incubated overnight at 4°C. Cells were washed 4 times for 5 minutes with TBST. Secondary antibody (IR-800CW rabbit, LI-COR, diluted 1 :8G0) and DNA stain DRAQ5 (LI-COR, diluted 1 :2G00) were added and Incubated 1-2 hours at room temperature. Cells were washed 4 times for 5 minutes with TBST. Plates were scanned on a Li-GOR Odyssey CLx Imager. Analysis of raw data was carried out in Excei (Microsoft) and Prism (GraphPad). Signal was plotted vs. the decadal logarithm of compound concentration, and IC 5 o was determined by fitting a 4-parameter sigmoidal concentration response model.

Determination of Ce!i Viability in RAS Mutant Cancer Ceil Lines Protocol: CellTiier-Gio© Ceil Viability Assay

Note The following protocol describes a procedure for monitoring ceil viability of K-Ras mutant cancer cell lines in response to a compound of the invention. Other RAS isoforms may be employed, though the number of ceils to be seeded will vary based on ceil line used.

The purpose of this cellular assay was to determine the effects of test compounds on the proliferation of throe human cancer cell lines (NCI-H358 (K-Ras G12C), AsPC-1 (K-Ras G12D), Capan-1 (K-Ras G12V)) over a 5-day treatment period by quantifying the amount of ATP present at endpoint using the GeilTiter-Glo® 2.0 Reagent (Promega).

Cells were seeded at 250 cells/well in 40 ml of growth medium in 384-well assay plates and incubated overnight in a humidified atmosphere of 5% GO? at 37°C. On the day of the assay, 10 mM stock solutions of test compounds were first diluted into 3 mM solutions with 100% DMSO. Well-mixed compound solutions (15 mI) were transferred to the next wells containing 30 ml of 100% DMSO, and repeated until a 9-concentration 3-fold serial dilution was made (starting assay concentration of 10 μM). Test compounds (132.5 nl) were directly dispensed into the assay plates containing cells. The plates were shaken for 15 seconds at 300 rpm, centrifuged, and incubated in a humidified atmosphere of 5% CO 2 at 37°C for 5 days. On day 5, assay plates and their contents were equilibrated to room temperature for approximately 30 minutes. CelITiter-Gio® 2.0 Reagent (25 pi) was added, and plate contents were mixed for 2 minutes on an orbital shaker before Incubation at room temperature for 10 minutes. Luminescence was measured using the PerkinEimer Enspire. Data were normalized by the following: (Sample signal/ Avg. DMSO) * 100. The data were fit using a four-parameter logistic fit.

The purpose of this biochemical assay was to measure the ability of test compounds to facilitate ternary complex formation between a nucleotide-loaded K-Ras isoform and Cyciophiiin A; the resuiting ternary complex disrupts binding to a BRAF RBD construct, inhibiting K-Ras signaling through a RAF effector. Data is reported as IC50 values. in assay buffer containing 25 mM HEPES pH 7.3, 0.002% Tween20, 0.1% BSA, 100 mM NaCl and 5 mM MgCl 2 , tagless Cyciophiiin A, His6-K-Ras-GMPPNP, and GST-BRAF RBD were combined in a 384-well assay plate at final concentrations of 25 μM, 12.5 nM and 50 nM, respectively. Compound was present in plate wells as a 10-point 3-fold dilution series starting at a final concentration of 30 μM. After incubation at 25°C for 3 hours, a mixture of Anii-His Eu-W1024 and anti-GST ailophycocyanin was then added to assay sample wells at final concentrations of 10 nM and 50 nM, respectively, and the reaction incubated for an additional 1 .5 hours TR-FRET signal was read on a micropiate reader (Ex 320 n , Em 885/615 nm). Compounds that facilitate disruption of a K-Ras:RAF complex were identified as those eliciting a decrease in the TR-FRET ratio relative to DMSO control weiis.

Table 4: Biological Assay Data for Representative Compounds of the Present Invention

Blank = not determined Additional H358 Ceil Viability assay data Key:

+++++: IC50 > 10 uM +4++: 10 uM > IC50 > 1 uM +: 1 uM > IC50 > 0.1 uM

++: 0.1 uM > IC50 > 0.01 uM +: IC50 < 0.01 uM

Table 5, H358 Cell Viability assay data (K-Ras G12C, IC50, uM): Key:

+++++: IC50 > 10 uM +4++: 10 uM > IC50 > 1 uM +: 1 uM > IC50 > 0.1 uM

++: 0.1 uM > iC5G > 0.01 uM +: IC50 < 0.01 uM

Table 6, Capan-1 Cell Viability assay data (K-Ras G12V, 1C50, ulVl):

Additional Ras-Raf disrupisorj/FRET/IVlOA assay data (IC5Q, uM)·, 'Key:

4-4-+++: IC50 > 10 uM ++++: 10 uM > IC50 > 1 uM

+++: 1 uM > IC50 > 0.1 uM ++: 0.1 uM > IC50 > 0.01 uM +: IC50 < 0.01 uM Table data

Table 8. KRAS G12C FRET data

Table 9. KRAS G12S FRET data

Table 10. KRAS G13C FRET data

Table 11. KRAS G12V FRET data

Table 12. KRAS WT FRET data

Table 13. KRAS G13D FRET data

Table 14. KRAS 061H FRET data

Table 15. NBAS G12C FRET data

Table 16. NBAS WT FRET data

Table 17. MBAS Q61K FRET data

Table 18. NBAS Q61R FRET data

In vitro Cell Proliferation Panels Potency for inhibition of cell growth was assessed at CrownBio using standard methods. Briefly, cell lines were cultured in appropriate medium, and then piated In 3D methylcellulose. Inhibition of celi growth was determined by CellTiter-Glo® after 5 days of culture with increasing concentrations of compounds. Compound potency was reported as the 50% inhibition concentration (absolute IC50).

The assay took place over 7 days. On day 1 , cells in 2D culture were harvested during logarithmic growth and suspended in culture medium at 1x105 cells/ml. Higher or lower celi densities were used for some cell lines based on prior optimization. 3.5 mi of celi suspension was mixed with 6.5% growth medium with 1% methylcellulose, resulting In a cell suspension in 0.65% methylceliuiose. 90 mί of this suspension was distributed in the wells of 2 96-well plates. One plate was used for day 0 reading and 1 plate was used for the end-point experiment. Plates were Incubated overnight at 37 C with 5% C02. On day 2, one plate (for to reading) was removed and 10 ml growth medium plus 100 mί CeilTifer-Glo® Reagent was added to each well. After mixing and a 10 minute incubation, luminescence was recorded on an EnVision Multi-Label Reader (Perkin Elmer). Compounds in DMSO were diluted in growth medium such that the final, maximum concentration of compound was 10 μM, and serial 4-fold dilutions were performed to generate a 9-point concentration series. 10 ml of compound solution at 10 times final concentration was added to wells of the second plate. Plate was then incubated for 120 hours at 37C and 5% C02. On day 7 the plates were removed, 100 ml GellTiter-Glo® Reagent was added to each well, and after mixing and a 10 minute incubation, luminescence was recorded on an EnVision Multi-Label Reader (Perkin Elmer). Data was exported to GeneData Sereener and modeled with a sigmoidal concentration response model In order to determine the IC50 for compound response.

Not all cell lines with a given RAS mutation may be equally sensitive to a RAS inhibitor targeting that mutation, due to differential expression of efflux transporters, varying dependencies on RAS pathway activation for growth, or other reasons. This has been exemplified by the cell line KYSE-410 which, despite having a KRAS G12C mutation, is insensitive to the KRAS G12C (OFF) inhibitor MRTX-849 (Hallin et al., Cancer Discovery 10:54-71 (2020)), and the cell line SW1573, which is insensitive to the KRAS G12C (OFF) inhibitor AMG510 (Canon et al., Nature 575:217-223 (2019)).

Table 19: IC50 values for various cancer cell lines with Compound B, Compound C, and low sensitivity: IC50 > 1 uM moderately sensitive: 1 uM > iC50 > 0.1 uM very sensitive: IC50 < 0.1 uM

vivo PD and Efficacy Data with Compound A, a compound of the present invention FIG. 1A:

Methods: The human pancreatic adenocarcinoma Capan-2 KRASG12V/wt xenograft model was used for a singie-day treatment PK/PD study (FIG. 1A). Compound A (Capan-2 pERK K-Ras G12D EC50: 0.0037 uM) was administered at 100 mg/kg as a single dose or bid (second dose administered 8 hours after first dose) orally administered (po). The treatment groups with sample collections at various time points were summarized in Table 20 below. Tumor samples were collected to assess RAS/ERK signaling pathway modulation by measuring the mRNA level of human DUSP6 in qPCR assay, while accompanylng blood plasma samples were collected to measure circulating Compound A levels.

Table 20. Summary of treatment groups, doses, and time points for single-dose PK/PD study using Capan-2 tumors. Results: In FIG. 1A, Compound A delivered at 100 mg/kg as a single dose inhibited DUSP8 mRNA levels in tumors >95% through 10 hours. A second dose of Compound A 8 hours following first administration maintained pathway modulation of 93% through 24 hours. These data indicate Compound A provides strong MARK pathway modulation with continued target coverage.

FIG 1B:

Methods: Effects of Compound A on tumor cell growth in vivo were evaluated In the human pancreatic adenocarcinoma Capan-2 KRASG12V/wt xenograft model using female BALB/c nude mice (6- 8 weeks old). Mice were Implanted with Capan-2 tumor cells in 50% Matrigel (4 x 106 cells/mouse) subcutaneously in the flank. Once tumors reached an average size of ~180 mm3, mice were randomized to treatment groups to start the administration of test articles or vehicle. Compound A was orally administered (po) twice dally at 100 mg/kg. A SHP2 inhibitor, RMC-4550 (commercially available), was administered orally every other day at 20 mg/kg. Body weight and tumor volume (using calipers) was measured twice weekly until study endpoints. Tumor regressions calculated as >10% decrease in starting tumor volume. All dosing arms were well tolerated.

Results: In FIG. 1B, single agent SHP2i RMC-4550 dosed every other day at 20 mg/kg po resulted in 39% TGI. Single-agent Compound A administered at 100 mg/kg po bid dally led to a TGI of 98%, with 4/10 (40%) individual animals achieving tumor regressions. Combination of Compound A and RMC-4550 resulted in total tumor regression of 35%, with individual tumor regressions observed in 7/9 (77.8%) individual animals at the end of treatment (Day 40 after treatment started) in Capan-2 CDX model with heterozygous KRASG12V. The anti-tumor activity of Compound A, and Combination arms was statistically significant compared with control group ( *** p<0.001 , ordinary One-way ANOVA with multiple comparisons via a post-hoc Tukey’s test), while RMC-4550 was not significant at these doses.

While the invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure come within known or customary practice within the art to which the invention pertains and may be applied to the essential features set forth herein.

AH publications, patents and patent applications are herein Incorporated by reference in their entirety to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety. APPENDIX D-1

C40-, C28-, AND C-32-LINKED RAPAMYCIN ANALOGS AS MTOR INHIBITORS

FIELD OF THE DISCLOSURE

[0001] The present disclosure relates to mTOR inhibitors. Specifically, the embodiments are directed to compounds and compositions inhibiting mTOR, methods of treating diseases mediated by mTOR, and methods of synthesizing these compounds.

BACKGROUND OF THE DISCLOSURE

[0002] The mammalian target of rapamyein (mTOR) is a serine-threonine kinase related to the lipid kinases of the phosphoinositide 3-kinase (PI3K) family. mTOR exists in two complexes, mTORC 1 and mTORC2, which are differentially regulated, have distinct substrate specificities, and are differentially sensitive to rapamyein. mTORC 1 integrates signals from growth factor receptors with cellular nutritional status and controls the level of cap-dependent mRNA translation by modulating the activity of key translational components such as the cap-binding protein and oncogene eIF4E.

[0003] mTOR signaling has been deciphered in increasing detail. The differing pharmacology of inhibitors of mTOR has been particularly informative. The first reported inhibitor of mTOR, Rapamyein is now understood to be an incomplete inhibitor of mTORC 1 . Rapamyein is a selective mTORC 1 inhibitor through the binding to the FK506 Rapamyein Binding (FRB) domain of mTOR kinase with the aid of FK506 binding protein 12 (FKBP12). The FRB domain of mTOR is accessible in the mTORC l complex, but less so in the xnTORC2 complex. Interestingly, the potency of inhibitory activities against downstream substrates of mTORC 1 by the treatment of Rapamyein is known to be diverse among the mTORC 1 substrates. For example, Rapamyein strongly inhibits phosphorylation of the mTORC 1 substrate S6K and, indirectly, phosphorylation of the downstream ribosomal protein S6 which control ribosomal biogenesis. On the other hand, Rapamyein shows only partial inhibitory activity against phosphorylation of 4E-BP1, a major regulator of eIF4E which controls the initiation of CAP-dependent translation. As a result, more complete inhibitors of mTORC 1 signaling are of interest.

[0004] A second class of “ATP- site” inhibitors of mTOR kinase were reported. This class of mTOR inhibitors will be referred to as TORI (ATP site TOR inhibitor). The molecules compete with ATP, the substrate for the kinase reaction in the active site of the rnTOR kinase (and are therefore also rnTOR active site inhibitors). As a result, these molecules inhibit downstream phosphorylation of a broader range of substrates.

[0005] Although mTOR inhibition may have the effect of blocking 4E-BP1 phosphorylation, these agents may also inhibit mTORC2, which leads to a block of Akt activation due to inhibition of phosphorylation of Akt S473.

[0006] Disclosed herein, inter alia, are mTOR inhibitors. In some embodiments, compounds disclosed herein are more selective inhibitors of mTORC 1 versus mTORC2. In some embodiments, compounds disclosed herein are more selective inhibitors of mTORC2 versus mTORC 1 . In some embodiments, compounds disclosed herein exhibit no selectivity difference between mTORC 1 and mTORC2.

SUMMARY OF THE DISCLOSURE

[0007] The present disclosure relates to compounds capable of inhibiting the activity of TOR. The present disclosure further provides a process for the preparation of compounds of the present disclosure, pharmaceutical preparations comprising such compounds and methods of using such compounds and compositions in the management of diseases or disorders mediated by mTOR.

[0008] The present disclosure provides a compound of Formula Ic: or a pharmaceutically acceptable salt or tautomer thereof, wherein: R 32 is 11. =O, -OR 3 , -Ns, or -O-C(=Z 1 )-R 32a ;

R 28 is -H, (Ci-C 6 )alkyl, or -C(=Z 1 )-R 28a ;

R 40 is -H or -C(=Z 1 )-R 40a ; wherein when R 28 and R 40 are H, then R 32 is not =O; each Z 1 is independently O or S;

R 283 , R 32a , and R 40 * are independently -AALAA-B; -AAA-B; -LAAAAAAAB; -O-(Ci-C 6 )alkyl; or -0-(C 6 -Cio)aryl; wherein the aryl of -O-(C 6 -Cio)aryl is unsubstituted or substituted with 1-5 substituents selected from -NO 2 and halogen;

A 1 and A 2 are independently absent or are independently selected from wherein the bond on the left side of A 1 , as drawn, is bound to -C(=Z')- or L 2 ; and wherein the bond on the right side of the A 2 moiety as drawn, is bound to B or L 3 ; each Q is independently 1 to 3 rings selected from arylene, eyeloalkylene, heteroarylene, and heteroeyclylene; each X is independently absent or 1 to 2 rings selected from arylene, cyeloalkylene, heteroarylene, and beterocyclylene; each X 1 is independently a heteroarylene or heteroeyclylene ring; each W is independently absent or 1 to 2 rings selected from arylene, cyeloalkylene, heteroarylene, and heteroeyclylene; each W l is independently a heteroarylene or heteroeyclylene ring; each G is independently absent or a ring selected from arylene, cyeloalkylene, heteroarylene, and heteroeyclylene; each G 1 and G 2 are independently heteroarylene or heteroeyclylene ring; each L 1 is independently selected from

L 2 and L 3 are independently absent or are independently selected from each B is independently selected from n n nd on the left side of B J , as drawn, is bound to A 2 , L 3 , or L 1 ; and wherein the heteroarylene, heterocyclylene, and arylene are each independently optionally substituted with alkyl, hydroxy alkyl, haloalkyl, alkoxy, halogen, or hydroxyl; each R J is independently H or (Ci-Cc alkyl; each R 4 is independently H, (C 1 -C 6 )alkyl, halogen, 5-12 memhered heteroaryl, 5-12 membered heterocyclyl, (C 6 -Cio)aryl, wherein the heteroaryl, heterocyclyl, and aryl are each independently optionally substituted with -N(R ) 2 , -OR 3 , halogen, (C 1 -C 6 )alkyl, -(Ci-C 6 )alkylene-heteroaryl, -(C 1 -Cc alkylene-CN, -C(O)NR J -heteroaryl, or -C(O)NR 3 -heterocyclyl; each R 5 is independently H, (C 1 -C 6 )alkyl, -C(O)0R 3 , or -N(R ) 2 , wherein the alkyl of (C 1 -C 6 jalkyl is each independently optionally substituted with -N(R 3 ) 2 or -OR 3 ; each R 6 is independently H, (C 1 -C 6 lalkyl, -C(O)0R 4 , or -N(R 5 ) 2 , wherein the alkyl of (C 1 -C 6 ialkyl is each independently optionally substituted with N(R S2 or -OR J ; each R ; is independently H, (C 1 -C 6 )aikyl, -C(O)0R 3 , or-N , wherein the alkyl of (C 1 -C t Oalkyl is each independently optionally substituted with N(R 3 ) 2 or -OR 3 ; each R 8 is independently H, (C 1 -C 6 lalkyl, -C(O)0R 4 , or N(R 3 ) 2 , wherein the alkyl of (C 1 -C 6 ialkyl is each independently optionally substituted with -N(R 3 ) 2 or -OR 3 ; each Y is independently -C(R 3 ) 2 or a bond; each n is independently an integer from one to 12; each o is independently an integer from zero to 30; each p is independently an integer from zero to 12; each q is independently an integer from zero to 30; and each r is independently an integer from one to 6.

[0009] The present disclosure provides a compound of Formula la:

5UB5TITUTE SHEET (RULE 26) da) or a pharmaceutically acceptable salt or tautomer thereof, wherein:

R 32 is -H, =O, -OR 3 -N 3 , or -O-C(=Z 1 )-R 32a ;

R 28 is -H, (Ci-C 6 )alkyl, or -C(=Z J )-R 28a ; wherein when R 28 and R 40 are H, then R 32 is not =O; each Z 1 is independently O or S;

R 28a , R 32a , and R 40® are independently -A^IAA^B; -A A 2 -B; -IAA^ILA LB; -O-(Ci-C 6 )alkyl; or -O-(C 6 -Cio)aryl; wherein the aryl of --O-(C 6 -Cio)aryl is unsubstituted or substituted with 1-5 substituents selected from -NO ?, and halogen;

A s and A 2 are independently absent or are independently selected from wherein the bond on the left side of A 1 , as drawn, is bound to -C(=Z 1 )- or L 2 ; and wherein the bond on the right side of the A 2 moiety, as drawn is bound to B or L 5 ; each Q is independently 1 to 3 rings selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; each X is independently absent or 1 to 2 rings selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; each X 1 independently is a heteroarylene or heterocyclylene ring; each W is independently absent or 1 to 2 rings selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; each W 1 independently is a heteroarylene or heterocyclylene ring; each G is independently absent or a ring selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; each G s and G 2 are independently heteroarylene or heterocyclylene ring; each L 1 is independently selected from L 2 and L 3 are independently absent or are independently selected from each B is independently selected from each B 1 is independently selected from

- | -NR 3 -(C(R 3 ) 2 )n-(C 6 -C 1 o)aiylene-(C(R 3 ) 2 )n-, -*-NR 3 -(C(R 3 ) 2 )n-heteroaiylene-, -¾-(C 6 -Cio)aiylene-, i-NR 3 -(C(R > ) 2 ) n -heteroarylene-heterocyclylene-(C 6 -Cio)arylene-,

^ heteroarylene-hete r ocy C lylene-(C 6 -C 1 o)arylene-, ^C-(C(R 3 )2 ) P - c-(C(R 3 ) 2 ) p -heteroarylene- ? mo — anc j ^ ~ NR -(C(R 3 ) 2 ) u -S(O) 2- ar 'lene-C(O)-, wherein the Y bond on the left side of B 1 , as drawn, is bound to A 2 , Ld, or L 1 ; and wherein the heteroarylene, heterocyclylene, and arylene are each independently optionally substituted with alkyl, hydroxyaikyl, haloalkyl, alkoxy, halogen, or hydroxyl; each R is independently H or (C 1 -C 6 jalkyl; each R 4 is independently H, (C 1 -C 6 )alkyl, halogen, 5-12 membered heteroaryl, 5-12 membered heterocyclyl, (C 6 -Cioiaryl, wherein the heteroaryl, heterocyclyl, and aryd are each independently optionally substituted with -N(R 3 ) 2 , -OR 3 , halogen, (C 1 -C t palkyl, -(Ci-C 6 )alkylene-heteroaryl, -(€ i -C 6 lalkylene-CN, -C(O)NR 3 -heteroaryl, or -C(O)NR 3 -heterocyclyl; each R 5 is independently H, (C 1 -CDalkyl, -C(O)OR 3 , or -N(R ) 2 , wherein the alkyl of (C -C 6 ialkyl is each independently optionally substituted with -N(R ) 2 or -OR 3 ; each R 6 is independently H, (C -C & )alkyl, -C(O)OR 3 , or -N(R J ) 2 , wherein the alkyl of (C 1 -C 6 lalkyl is each independently optionally substituted with -N(R 3 ) 2 or -OR 3 ; each R' is independently H, (C 1 -C 6 )alkyl, -C(O)OR 3 , or -N , wherein the alkyl of (C 1 -C 6 )alkyl is each independently optionally substituted with -N(R 3 ) 2 or -OR 3 ; each R 8 is independently H, (C 1 -C 6 jalkyl, -C(O)0R 3 , or -N(R 3 ) 2 , wherein the alkyl of (C 1 -C 6 )alkyl is each independently optionally substituted with -N or -OR 3 ; each Y is independently Ci Y· or a bond; each n is independently an integer from one to 12; each o is independently an integer from zero to 30; each p is independently an integer from zero to 12; each q is independently an integer from zero to 30; and each r is independently an integer from one to 6.

[0010] The present disclosure provides a compound of Formula I: or pharmaceutical] y acceptable salt or tautomer thereof, wherein:

R 32 is -H, =O , or -OR 3 ;

R 40 is H or -C(=Z 1 )-R 40a ; wherein at least one of R 28 and R 40 is not H;

Z 1 is independently O or S;

R 28a and R 40a are independently -A^I A^B ; -A 1 -A 2 -B ; L 2 -A 1 -L 1 -A 2 -L 3 -B ; -O-(C 1 -C 6 )alkyl; or -O-fC 6 -Cu aryl; wherein the aryl of -O-(C 6 -Cio)aryl is unsubstituted or substituted with 1-5 substituents selected from -NO 2 and halogen;

A 1 and A 2 are independently absent or are independently selected from s:

wherein the bond on the left side of A 1 , as drawn, is bound to -C(=Z')- or L 2 ; and wherein the bond on the right side of the A 2 moiety, as drawn, is bound to B or L 3 ; each Q is independently 1 to 3 rings selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; each X is independently absent or 1 to 2 rings selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; each X s is independently a heteroarylene or heterocyclylene ring; each W is independently absent or 1 to 2 rings selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; each W l independently is a heteroarylene or heterocyclylene ring; each G is independently absent or a ring selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; each G 1 and G 2 are independently heteroarylene or heterocyclylene ring; each L 1 is independently selected from

1 on

L 2 and L 3 are independently absent or are independently selected from

1 ΊO O each B is independently selected from

^ ~ NR 3 -(C(R 3 ) 2 ) n -S(O) 2- ar lene-C(())-, wherein the bond on the left side of B 1 , as drawn, is bound to A 2 , L3, or L 1 ; and wherein the heteroarylene, heterocyclylene, and arylene are each independently optionally substituted with alkyl, hydroxyalkyl, haloalkyl, alkoxy, halogen, or hydroxyl; each R 4 is independently H or (C 1 -C 6 Salkyl; each R 4 is independently H, (C 1 -Ccdalkyl, halogen, 5-12 membered heteroaryl, 5-12 membered heterocyclyl, (C 6 -Cio)aryI, wherein the heteroaryl, heterocyclyl, and aryl are each independently optionally substituted with -N(R 3 ) 2 , -OR 3 halogen, (CTC t palkyl, -(Ci-C t Oalkylene-heteroaryl, -(C 1 -C 6 lalkylene-CN -C(O)NR 3 -heteroaryl, or -C(O)NR 3 -heterocyclyl; each R 5 is independently H, (C 1 -C 6 )alkyl, -CiOiOR '. or-N(R 3 ) 2 , wherein the alkyl of (C -Gpalkyl is optionally substituted with -N(R ) 2 or -OR 3 ; each R 6 is independently H, (C 1 -C 6 jaikyl, -C(O)GR 3 , or -N(R 3 ) 2 , wherein the alkyl is of (Cj-C 6 )alkyl optionally substituted with -N(R 3 ) 2 or -OR 3 ; each R' is independently H, (C 1 -Ccdalkyl, -C(O)0R J , or -N(R 3 ) 2 , wherein the alkyl of (C 1 -C 6 )alkyl is optionally substituted with -N(R 3 ) 2 or -OR 3 ; each R 8 is independently H, (C 1 -C 6 )alkyl, -C(O)0R 3 , or -N(R ) 2 , wherein the alkyl of (C 1 -C jalkyl is optionally substituted with -N(R J ) 2 or -OR 3 ; each Y is independently -C(R 3 ) 2 or a bond; each n is independently an integer from one to 12; each o is independently an integer from zero to 30; each p is independently an integer from zero to 12; each q is independently an integer from zero to 30; and each r is independently an integer from one to 6.

[0011] The present disclosure provides a compound of Formula II: or a pharmaceutically acceptable salt or tautomer thereof, wherein:

R 32 is 11. =O or -OR 3 ;

R 28 is -H or -C(=Z 1 )-R 28a ;

R 40 is H or -C(=Z 1 )-R 40a ; wherein at least one of R 28 and R 40 is not H;

Z 1 is independently O or S;

R 28a and R 40a are independently -A^I A^B; -A^A^B; -O-(Ci-C 6 )alkyl; or -C C 6 - Cio)aryl; wherein the aryl of -O-(C 6 -Cio)aryl is unsubstituted or substituted with 1-5 substituents selected from --NO 2 and halogen;

A 1 and A 2 are independently absent or are independently selected from

wherein the bond on the left side of A 1 , as drawn, is bound to -C(=Z 1 )-; and wherein the bond on the right side of the A 2 moiety, as drawn, is bound to B; each Q is independently 1 to 3 rings selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; each X is independently absent or 1 to 2 rings selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; each X 1 is independently a heteroarylene or heterocyclylene ring; each W is independently absent or 1 to 2 rings selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; each W 1 is independently a heteroarylene or heterocyclylene ring; each G is independently absent or a ring selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; each G 1 and G 2 are independently heteroarylene or heterocyclylene ring; each L 1 is independently selected from

each B is independently selected from each B 3 is independently selected from NR 3 -(C(R 3 ) 2 ) n -, ~NR 3 -(C(R 3 ) 2 ) n -S(O) 2- aryIene-C(O)- , wherein the £ ~ bond on the left side of B 1 , as drawn, is bound to A 2 or L 1 ; and wherein the heteroarylene, heterocyclylene, and arylene are each independently optionally substituted with alkyl, hydroxyalkyl, haloalkyl, alkoxy, halogen, or hydroxyl; each R 3 is independently H or (Ci-C 6 lalkyl; each R 4 is independently H, (C 1 -C 6 jalkyl, halogen, 5-12 membered heteroaryl, 5-12 membered heterocyclyl, (C 6 -C t o)aryl, wherein the heteroaryl, heterocyclyl, and aryl are each independently optionally substituted with -N(R 3 ) 2 , -OR 3 , halogen, (C 1 -C 6 lalkyl, -(C 1 -C 6 lalkylene-heieroaryl, -(C 1 -C 6 lalkylene-CN, -C(O)NR -heteroaryl, or -C(O)NR 3 -heterocyclyl; each R 1 ' is independently H, (C -C 6 )alkyl, -C(O)OR 3 , or -N(R 3 ) 2 , wherein the alkyl of (C 1 -C 6 lalkyl is optionally substituted with N{R or -OR 3 ; each R 6 is independently H, (C 1 -Ccdalkyl, -C(G)0R 3 , or -N(R 3 ) 2 , wherein the alkyl of (C 1 -C fi lalkyl is optionally substituted with -N 2 or -OR 3 ; each R 7 is independently H, (C 1 -C 6 jalkyl, -C(O)GR 3 , or -N(R 3 ) 2 , wherein the alkyl of (C 1 -C 6 )alkyl is optionally substituted with -N(R 3 ) 2 or -OR 3 ; each R 8 is independently H, (C 1 -C 6 ialkyl, -C(O)OR 3 , or -N(R 5 ) 2 , wherein the alkyl of (C 1 -C 6 lalkyl is optionally substituted with --N(R 3 ) 2 or -OR ; each Y is independently C(R 3 ) 2 or a bond; each n is independently an integer from one to 12; each o is independently an integer from zero to 30; each p is independently an integer from zero to 12; each c] is independently an integer from zero to 30; and each r is independently an integer from one to 6.

[0012] In some embodiments, a compound of Formula I or II is represented by the structure of Formula 1-28: or a pharmaceutically acceptable salt or tautomer thereof.

[0013] In some embodiments, a compound of Formula la, Ic, I, or II is represented by the structure of Formula I-28b: or a pharmaceutically acceptable salt or a tautomer thereof.

[0014] In some embodiments, a compound of Formula I or II is represented by the structure of Formula 1-40:

1 nnr

or a pharmaceutically acceptable salt or tautomer thereof.

[0015] In some embodiments, a compound of Formula la, Ic, I or II is represented by the structure of Formula I-4Qb: or a pharmaceutically acceptable salt or a tautomer thereof

[0016] In some embodiments, a compound of Formula la, Ic, I or II is represented by the structure of Formula I-32b:

or a pharmaceutically acceptable salt or a tautomer thereof.

[0017] The present disclosure provides a method of treating a disease or disorder mediated by mTOR comprising administering to the subject suffering from or susceptible to developing a disease or disorder mediated by mTOR a therapeutically effective amount of one or more disclosed compounds. The present disclosure provides a method of preventing a disease or disorder mediated by mTOR comprising administering to the subject suffering from or susceptible to developing a disease or disorder mediated by mTOR a therapeutically effective amount of one or more disclosed compounds. The present disclosure provides a method of reducing the risk of a disease or disorder mediated by mTOR comprising administering to the subject suffering from or susceptible to developing a disease or disorder mediated by mTOR a therapeutically effective amount of one or more disclosed compounds.

[0018] Another aspect of the present disclosure is directed to a pharmaceutical composition comprising a compound of Formula I, la, lb, Ic, II, or lib, or a pharmaceutically acceptable salt or tautomer of any of the foregoing, and a pharmaceutically acceptable carrier. The pharmaceutically acceptable carrier can further comprise an excipient, diluent, or surfactant. The pharmaceutical composition can be effective for treating, preventing, or reducing the risk of a disease or disorder mediated by mTOR in a subject in need thereof.

[0019] Another aspect of the present disclosure relates to a compound of Formula I, la, lb, Ic, II, or lib, or a pharmaceutically acceptable salt or tautomer of any of the foregoing, for use in treating, preventing, or reducing the risk of a disease or disorder mediated by mTOR in a subject in need thereof.

[0020] Another aspect of the present disclosure relates to the use of a compound of Formula I, la, lb, Ic, II, or lib, or a pharmaceutically acceptable salt or tautomer of any of the foregoing, in the manufacture of a medicament for in treating, preventing, or reducing the r sk of a disease or disorder mediated by mTOR in a subject in need thereof.

[0021] The present disclosure also provides compounds that are useful in inhibiting mTOR.

DETAILED DESCRIPTION OF THE DISCLOSURE

[0022] The present disclosure relates to mTOR inhibitors. Specifically, the embodiments are directed to compounds and compositions inhibiting mTOR, methods of treating diseases mediated by mTOR, and methods of synthesizing these compounds.

[0023] The details of the disclosure are set forth in the accompanying description below. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present disclosure, illustrative methods and materials are now described. Other features, objects, and advantages of the disclosure will be apparent from the description and from the claims. In the specification and the claims, the singular forms also may include the plural unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. All patents and publications cited in this specification are incorporated herein by reference in their entireties.

Terms

[0024] The articles “a” and “an” are used in this disclosure and refers to one or more than one (i.e., to at least one) of the grammatical object of the article, unless indicated otherwise. By way of example, “an element” may mean one element or more than one element, unless indicated otherwise.

[0025] The term “or” means “and/or” unless indicated otherwise. The term “and/or” means either “and” or “or”, or both, unless indicated otherwise.

[0026] The term “optionally substituted” unless otherwise specified means that a group may be unsubstituted or substituted by one or more (e.g., 0 1, 2, 3, 4, or 5 or more, or any range derivable therein) of the substituents listed for that group in w'hich said substituents may be the same or different. In an embodiment, an optionally substituted group has 1 substituent. In another embodiment an optionally substituted group has 2 substituents. In another embodiment an optionally substituted group has 3 substituents. In another embodiment an optionally substituted group has 4 substituents. In another embodiment an optionally substituted group has 5 substituents.

[0027] The term “alkyl,” by itself or as part of another substituent, means, unless otherwise stated, a straight (he., unbranched) or branched non-cyclic carbon chain (or carbon), or combination thereof, which may be fully saturated, mono- or polyunsaturated and can include di-and multivalent radicals, having the number of carbon atoms designated (he., Ci-C 1 o means one to ten carbons). Examples of saturated hydrocarbon radicals include, but are not limited to, groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, (cyclohexyl)methyl, homologs and isomers of, for example, n-pentyl, n-hexyl, n- heptyl, n-octyl, and the like. An unsaturated alkyl group is one having one or more double bonds or triple bonds. Examples of unsaturated alkyl groups include, but are not limited to, vinyl, 2-propenyl, crotyl, 2-isopentenyl, 2-(butadienyl), 2,4-pentadienyl, 3-(1,4-pentadienyl), ethynyl, 1- and 3-propynyl, 3-butynyl, and the higher homologs and isomers.

[0028] The term “alkylene,” by itself or as part of another substituent, means, unless otherwise stated, a divalent radical derived from an alkyl. Typically, an alkyl (or alkylene) group will have from 1 to 24 carbon atoms such as those groups having 10 or fewer carbon atoms.

[0029] The term “alkenyl” means an aliphatic hydrocarbon group containing a carbon — carbon double bond and which may be straight or branched having about 2 to about 6 carbon atoms in the chain. Certain alkenyl groups have 2 to about 4 carbon atoms in the chain. Branched may mean that one or more lower alkyl groups such as methyl, ethyl, or propyl are atached to a linear alkenyl chain. Exemplary alkenyl groups include ethenyl, propenyl, n- butenyl, and i-butenyl. A C :·( " · > alkenyl group is an alkenyl group containing between 2 and 6 carbon atoms.

[0030] The term “alkenylene,” by itself or as part of another substituent, means, unless otherwise stated, a divalent radical derived from an alkene.

[0031] The term “alkynyl” means an aliphatic hydrocarbon group containing a carbon — carbon triple bond and which may be straight or branched having about 2 to about 6 carbon atoms in the chain. Certain alkynyl groups have 2 to about 4 carbon atoms in the chain. Branched may mean that one or more lower alkyl groups such as methyl, ethyl, or propyl are attached to a linear· alkynyl chain. Exemplary alkynyl groups include ethynyl, propynyl, n- butynyl, 2-butynyl, 3-methylbutynyl, and n-pentynyl. A CVC 6 alkynyl group is an alkynyl group containing between 2 and 6 carbon atoms.

[0032] The term “alkynylene,” by itself or as part of another substituent, means, unless otherwise stated, a divalent radical derived from an alkyne.

[0033] The term “cycioalkyl” means a monocyclic or polycyclic saturated or partially un saturated carbon ring containing 3-18 carbon atoms. Examples of cycloalkyl groups include, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptanyl, cyclooctanyl, norboranyl, norborenyl, bicyclo[2.2.2]octanyI, or bicyclo[2.2.2]octenyl. A C 3 - Cg cycloalkyl is a cycloalkyl group containing between 3 and 8 carbon atoms. A cycioalkyl group can be fused (e.g., decalin) or bridged (e.g., norbornane).

[0034] A “cycloalkylene," alone or as part of another substituent, means a divalent radical derived from a cycioalkyl.

[0035] The terms “heterocyclyl” or “heterocycloalkyl” or “heterocycle” refers to a monocyclic or polycyclic 3 to 24-xnembered ring containing carbon and at least one heteroatom selected from oxygen, phosphorous, nitrogen, and sulfur and wherein there is not delocalized p electrons (aromaticity) shared among the ring carbon or heteroatom(s). Heterocyclyl rings include, but are not limited to, oxetanyl, azetadinyl, tetrahydrofuranyl, pyrrolidinyl, oxazolinyl, oxazolidinyl, thiazolinyl, thiazolidinyl, pyranyl, thiopyranyl, tetrahydropyranyl, dioxalinyl, piperidinyl, morphoiinyl, thiomorpholinyl, thiomorpholinyl S- oxide, thiomorpholinyl S-dioxide, piperazinyl, azepinyl, oxepinyl, diazepinyl, tropanyl, and homotropanyl. A heterocyclyl or heterocycloalkyl ring can also be fused or bridged, e.g., can be a bicyclic ring.

[0036] A “heterocyclylene” or “heterocycloalky lene,” alone or as part of another substituent, means a divalent radical derived from a “heterocyclyl” or “heterocycloalkyl” or “heterocycle.”

[0037] The term “aryl” means, unless otherwise stated, a polyunsaturated, aromatic, hydrocarbon substituent, which can be a single ring or multiple rings (preferably from 1 to 3 rings) that are fused together (i.e., a fused ring aryl) or linked covalently A fused ring aryl may refer to multiple rings fused together wherein at least one of the fused rings is an aryl ring

[0038] An “arylene,” alone or as part of another substituent, means a divalent radical derived from an aryl. [0039] The term “heteroaryl” refers to an aryl group (or rings) that contains at least one heteroatom such as N, O, or S, wherein the nitrogen and sulfur atom(s) are optionally oxidized, and the nitrogen atoxn(s) is optionally quatemized. Thus, the term “heteroaryl” includes fused ring heteroaryl groups (i.e, multiple rings fused together wherein at least one of the fused rings is a heteroaromatic ring). A 5,6-fused ring heteroarylene refers to two rings fused together, wherein one ring has 5 members and the other ring has 6 members and wherein at least one ring is a heteroaryl ring. Likewise, a 6,6-fused ring heteroarylene refers to two rings fused together, wherein one ring has 6 members and the other ring has 6 members, and wherein at least one ring is a heteroaryl ring. And a 6,5-fused ring heteroarylene refers to two rings fused together, wherein one ring has 6 members and the other ring has 5 members, and wherein at least one ring is a heteroaryl ring. A heteroaryl group can be attached to the remainder of the molecule through a carbon or heteroatom. Non- limiting examples of aryl and heteroaryl groups include phenyl, 1-naphthyl, 2-naphthyl, 4- biphenyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 3-pyrazolyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl, 4-oxazolyl, 2-phenyl-4-oxazolyl, 5-oxazolyl, 3-isoxazolyl, 4- isoxazolyl, 5-isoxazolyi, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-fury I, 3-furyl, 2-thienyl, 3- thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-benzothiazolyI, purinyl, 2-benzimidazolyl, 5-indolyl, 1-isoquinolyl, 5-isoquinolyl, 2-quinoxalinyl, 5-quinoxalinyl, 3- quinolyl, and 6-quinolyl. Substituents for each of the above noted aryl and heteroaryl ring systems are selected from the group of acceptable substituents described herein.

[0040] The term “heteroaryl” may also include multiple condensed ring systems that have at least one such aromatic ring, which multiple condensed ring systems are further described below'. The term may also include multiple condensed ring systems (e.g., ring systems comprising 2, 3 or 4 rings) wherein a heteroaryl group, as defined above, can be condensed with one or more rings selected from heteroaryls (to form for example a naphihyridinyl such as 1,8-naphthyridinyl), heterocycles, (to form for example a 1, 2, 3, 4- tetrahydronaphthyridinyl such as 1, 2, 3, 4-tetrahydro-1,8-naphthyridinyl), carbocycles (to form for example 5,6,7, 8-tetrahydroquinolyl) and aryls (to form for example indazolyl) to form the multiple condensed ring system. The rings of the multiple condensed ring system can be connected to each other via fused, spiro and bridged bonds when allowed by valency requirements. It is to be understood that the individual rings of the multiple condensed ring system may be connected in any order relative to one another. It is also to be understood that the point of attachment of a multiple condensed ring sy stem (as defined above for a heteroaryl) can be at any position of the multiple condensed ring system including a heteroaryl, heterocycle, and or carbocycle portion of the multiple condensed ring system and at any suitable atom of the multiple condensed ring system including a carbon atom and heteroatom (e.g., a nitrogen)

[0041] A “heteroarylene,” alone or as part of another substituent, means a divalent radical derived from a heteroaryl

[0042] Non-limiting examples of aryl and heteroaryl groups include pyridinyl, pyrimidinyl, thiophenyl, thienyl, furanyl, indolyl, benzoxadiazolyl, benzodioxolyl, henzodioxanyl, thianaphthanyl, pyrrolopyridinyl, indazolyl, quinolinyl, quinoxalinyl, pyridopyrazinyl, quinazolinony 1, benzol soxazolyl , imidazopyridinyl, benzo furan l, benzothienyl, benzothiophenyl, phenyl, naphthyl, biphenyl, pyrrolyl, pyrazolyl, imidazolyl, pyrazinyl, oxazolyl, isoxazolyl, thiazolyl, furylthienyl, pyridyl, pyrimidyl, benzothiazolyl, purinyl, benzimidazolyl, isoquinolyl, fhiadiazolyl, oxadiazolyl, pyrrolyl, diazolyl, triazolyl, tetrazolyl, benzothiadiazolyl, isothiazolyl, pyrazolopyrimidinyl, pyrrolopyrimidinyl, benzotriazolyl, benzoxazolyl, or quinoiyl. The examples above may be substituted or unsubstituted and divalent radicals of each heteroaryl example above are non-limiting examples of heteroarylene. A heteroaryl moiety may include one ring heteroatom (e.g., O, N, or S). A heteroaryl moiety may include two optionally different ring heteroatoms (e.g., O, N, or S). A heteroaryl moiety may include three optionally different ring heteroatoms (e.g., O,

N, or S). A heteroaryl moiety may include four optionally different ring heteroatoms (e.g., O, N, or S). A heteroaryl moiety may include five optionally different ring heteroatoms (e.g., O, N, or S). An aryl moiety may have a single ring. An aryl moiety may have two optionally different rings. An aryl moiety may have three optionally different rings. An aryl moiety may have four optionally different rings. A heteroaryl moiety may have one ring. A heteroaryl moiety may have two optionally different rings. A heteroaryl moiety may have three optionally different rings. A heteroaryl moiety may have four optionally different rings. A heteroaryl moiety may have five optionally different rings.

[0043] The terms “halo” or “halogen,” by themselves or as part of another substituent, mean, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom. Additionally, terms such as “haloalkyl” may include monohaloalkyl and polyhaloalkyl. For example the term “halo(C 1 -C 4 )alkyl” may include, but is not limited to, fluoromethyl, dilluoromethyl, trifluoromethyk 2,2,2-trifluoroethyL 4-chlorobutyl, 3-bromopropyl, 1-fluoro-2-bromoethyl, and the like [0044] The term “hydroxyl,” as used herein, means -OH.

[0045] The term “hydroxyalkyl” as used herein, means an alkyl moiety as defined herein, substituted with one or more, such as one, two or three, hydroxy groups. In certain instances, the same carbon atom does not carry more than one hydroxy group. Representative examples include, but are not limited to, hydroxymethyl, 2-hydroxyethyl, 2-hydroxypropyl, 3- hydroxypropyl, 1-(hydroxymethyl)-2-methylpropyl, 2-hydroxybntyL 3-hydroxybutyl, 4- hydroxybutyl, 2,3-dihydroxypropyL 2-hydroxy-1-hydroxymethylethyl, 2,3-dihydroxybutyl, 3,4-dihydroxybutyl and 2-(hydroxymethyl)-3-hydroxypropyl.

[0046] The term “oxo,” as used herein, means an oxygen that is double bonded to a carbon atom.

[0047] A substituent group, as used herein, may be a group selected from the following moieties:

(A) oxo, halogen, -CF 3 , -CN, -OH, -OCH 3 , -N¾, -COOH, -CONH 2 , -NO 2 , -SH, -SO 3 H, -SO4H, -SO 2 NH 2 , -NHNH 2 , -ONH 2 , -NHC=(O)NHNH 2 , -NHC=(O)NH 2 , -NHSO 2 H, -NHC=(O)H, -NHC(O)-OH, -NHOH. -OCF 3 , -OCHF2, -OCH 2 F, unsubstituted alkyl, un substituted cycloalkyl, unsubstituted heterocycloalkyl, unsubstituted aryl, unsubstituted heteroaryl, and

(B) alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, substituted with at least one substituent selected from:

(i) oxo, halogen, -CF 3 , -CN, -OH, -OCH 3 , -NH 2 , -COOH, -CONH 2 , -NO 2 , -SH, -SO 3 H, -SO4H, -SO 2 NH2, -NHNH 2 , -ONH 2 , -NHC=(O)NHNH 2 , -NHC=(O)NH 2 , -NHSO 2 H, -NHC=(O)H, -NHC(O)-OH, -NHOH, -OCF 3 , -OCHF 2 , -OCH 2 F, unsubstituted alkyl unsubstituted heteroalkyl, unsubstituted cycloalkyl, unsubstituted heterocycloalkyl, unsubstituted aryl, unsubstituted heteroaryl, and

(ii) alkyl, heteroalky cycloalkyl, heterocycloalkyL aryl, heteroaryl, substituted with at least one substituent selected from:

(a) oxo, halogen, -Cl·,. -CN, -OH, -OCH 3 , -NH 2 , -COOH, -CONH 2 , -NO 2 , -SH, -S0 3 H, -SO4H, -SO 2 NH 2 , -NHNH 2 , -ONH 2 , -NHC=(O)NHNH 2 , -NHC=(O)NH 2, -NHSO 2 H, -NHC=(O)H, -NHC(O)-OH, -NHOH, -OCF3, -OCHF2, -OCH 2 F, unsubstituted alkyl, unsubstituted heteroalkyl, unsubstituted cycloalkyl, unsubstituted heterocycloalkyl, un substituted aryl, unsubstituted heteroaryl, and

(h) alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, substituted with at least one substituent selected from: oxo, halogen, -GET, -CN, -OH, -OCH 3 , -NH 2 , -COOH -CONH2, -NO 2 , -SH, -SO 3 H, -SO4H, -SO 2 NH 2 , -NHNH 2 , -ONH 2 , -NHC=(O)NHNH 2 , -NHC=(O)NH 2 , -NHSO 2 H, -NHC=(O)H, -NHC(O)-OH, -NHOH, -OCF3, -OCHF2, -OCH 2 F, unsubstituted alkyl, unsubstituted heteroalkyl, imsubstituted cycloalkyl, imsubstituted heterocycloalkyl, unsubstituted aryl, unsubstituted heteroaryl.

[0048] An “effective amount” when used in connection with a compound is an amount effective for treating or preventing a disease in a subject as described herein.

[0049] The term “carrier”, as used in this disclosure, encompasses carriers, excipients, and diluents and may mean a material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting a pharmaceutical agent from one organ, or portion of the body, to another organ, or portion of the body of a subject .

[0050] The term “treating” with regard to a subject, refers to improving at least one symptom of the subject’s disorder. Treating may include curing, improving, or at least partially ameliorating the disorder.

[0051] The term “prevent” or “preventing” with regard to a subject refers to keeping a disease or disorder from afflicting the subject. Preventing may include prophylactic treatment. For instance, preventing can include administering to the subject a compound disclosed herein before a subject is afflicted with a disease and the administration will keep the subject from being afflicted with the disease.

[0052] The term “disorder” is used in this disclosure and means, and is used interchangeably with, the terms disease, condition, or illness, unless otherwise indicated.

[0053] The term “administer”, “administering”, or “administration” as used in this disclosure refers to either directly administering a disclosed compound or pharmaceutically acceptable salt or tautomer of the disclosed compound or a composition to a subject, or administering a prodrug derivative or analog of the compound or pharmaceutically acceptable salt or tautomer of the compound or composition to the subject, which can form an equivalent amount of active compound within the subject’s body.

[0054] A “patient” or “subject” is a mammal, e.g., a human, mouse, rat, guinea pig, dog, cat horse, cow, pig, or non-human primate, such as a monkey, chimpanzee, baboon or rhesus.

Compounds

[0055] The present disclosure provides a compound having the structure of Formula Ic, or a pharmaceutically acceptable salt or tautomer thereof, wherein R 32 , R 28 , and R 40 are described as above.

[0056] The present disclosure provides a compound having the structure of Formula la. or a pharmaceutically acceptable salt or tautomer thereof, wherein R 32 , R 28 , and R 40 are described as above.

[0057] The present disclosure provides a compound having the structure of Formula I, or a pharmaceutically acceptable salt or tautomer thereof, wherein R 32 , R 28 , and R 40 are described as above The present disclosure provides a compound having the structure of Formula lb: or a pharmaceutically acceptable salt and or tautomer thereof, wherein R 3 R 28 , and R 40 are described as above for Formula I

The present disclosure provides a compound having the structure of Formula II, or a pharmaceutically acceptable salt or tautomer thereof, wherein R 32 , R 28 , and R 40 are described as above.

[0060] The present disclosure provides a compound having the structure of Formula lib: or a pharmaceutically acceptable salt or tautomer thereof, wherein R 32 , R 28 , and R 40 are described as above for Formula II.

In certain embodiments, a compound has the following formula: or a pharmaceutically acceptable saltor tautomer thereof.

[0062] In certain embodiments, R 32 is ~ 0. In certain embodiments, R 2 is -OR 3 . In certain embodiments, R 32 is H. In certain embodiments, R 32 is -N3.

[0063] As described above, each R is independently H or (C 1 -C 6 lalkyl. In certain embodiments, R 3 is H. In certain embodiments, R 3 is (CTC 6 ialkyl. In certain embodiments, R 3 is methyl.

[0064] In certain embodiments, R 28 is H. In certain embodiments, R 28 is (C -C 6 jalkyl. In certain embodiments, R 40 is H.

[0065] In certain embodiments, a compound is represented by the structure of Formula I-

40b: or a pharmaceutically acceptable salt or tautomer thereof, wherein R 32 and R 40 are described as above for Formula la, Ic, I, or II. [0066] In certain embodiments, a compound is represented by the structure of Formula I-

40: or a pharmaceutically acceptable salt or tautomer thereof, wherein R 32 and R 40 are described as above.

[0067] In certain embodiments, R 40 is -C(=Z 1 )-R 40a . In certain embodiments, Z 1 is O. In certain embodiments, Z 1 is S.

[0068] In certain embodiments, R a is -O-fCi-C 6 jalkyl or -O-(C 6 -Cio)aryl; wherein the aryl is unsubstituted or substituted with 1-5 substituents selected from NO 2 and halogen.

[0069] In certain embodiments, R 40a is -A 1 -L 1 -A 2 -B. In certain embodiments, R 40a is-A 1 -A 2 -B. In certain embodiments, R 40a is --L 2 -A 1 -L 1 -A 2 -L 3 -B.

[0070] In certain embodiments, R 4 a is -A 1 -L 1 -A 2 -B, wherein A 1 and A 2 are absent. In certain embodiments, R 40 * is -A l -L l -A 2 -B, wherein A 2 is absent. In certain embodiments R 40a is A 1 -L 1 -A 2 -B, wherein A 1 is absent. In certain embodiments, R 40a is A 1 -L 1 -A 2 -B. In certain embodiments, R 40a is -A J -A 2 -B. In certain embodiments, R 40a is -L 2 -A 3 -L 3 -A 2 -L 3 -B, wherein L 2 and A 1 are absent. In certain embodiments, R 40a is -I/-A 1 -L 1 -A 2 -L 3 -B, wherein L 2 is absent. In certain embodiments, R 40a is -L 2 -A f -L f -A 2 -L/-B, wherein L 3 is absent

[0071] In certain embodiments, a compounds is represented by the structure of Formula

1-28b: or a pharmaceutically acceptable salt or tautomer thereof, wherein R 32 and R 28 are described as above for Formula la, Ic, I, or II.

[0072] In certain embodiments, a compound is represented by the structure of Formula I-

28: or a pharmaceutically acceptable salt or tautomer thereof, wherein R 32 and R 28 are described as above.

[0073] In certain embodiments, R 28 is -Ci ^ Zlj-R 283 . In certain embodiments, Z 1 is O. In certain embodiments, Z f is S.

[0074] In certain embodiments, R 28a is -O-(Ci-C 6 )alkyl or -O-(C 6 -Cio)aryi; wherein the aryl is unsubstituted or substituted with 1-5 substituents selected from NO 2 and halogen.

[0075] In certain embodiments, R 28a is --A 1 -I, 1 -A 2 -B. In certain embodiments, R z8a is _A { -A 2 -B. In certain embodiments, R 28a is -L 2 A i -L i -A 2 -L 5 -B. [0076] In certain embodiments, R iSa is -A 1 -L 1 -A 2 -B, wherein A 3 and A 2 are absent. In certain embodiments, R 28a is --A 1 -I, 1 -A 2 -B, wherein A 2 is absent. In certain embodiments, R 28a is -A 1 -L 1 -A 2 -B, wherein A 3 is absent. In certain embodiments, R 28a is -A f -L f -A 2 -B. In certain embodiments, R 2 a is -A 3 -A 2 -B. In certain embodiments, R 28a is -L 2 -A 3 -L 3 -A 2 -L 3 -B, wherein L 2 and A 1 are absent. In certain embodiments, R 8a is -L 2 - A 3 -L 3 -A 2 -L -B , wherein L 2 is absent. In certain embodiments R 28a is -L 2 -A 1 -L 1 -A 2 -L 3 -B, wherein L 3 is absent.

[0077] In certain embodiments, the compounds are represented by the structure of Formula I -32b: or a pharmaceutically acceptable salt or tautomer thereof, wherein R 32 is described as above for Formula la, Ic, I, or II.

[0078] In certain embodiments, R 32 is -O-C(=Z 1 )-R 3 ^. In certain embodiments, Z 1 is O. In certain embodiments, Z 1 is S.

[0079] In certain embodiments, R 2a is -O-CCi-CcOalkyl or -O-(C 6 -Cio)aryl; wherein the aryl is un substituted or substituted with 1-5 substituents selected from NO 2 and halogen.

[0080] In certain embodiments, R 32a is -A 3 -L 3 -A 2 -B. In certain embodiments, R 32a is -A 3 -A 2 -B In certain embodiments, R 32a is -L 2 -A l -L l -A 2 -L 3 -B.

[0081] In certain embodiments, R 32a is -A 3 -L 3 -A 2 -B, wherein A 1 and A 2 are absent. In certain embodiments, R 3 a is -A J -L J -A 2 -B, wherein A 2 is absent. In certain embodiments, R 32a is -A l -L l -A 2 -B, wherein A 3 is absent. In certain embodiments, R 32a is -A 1 -L 1 -A 2 -B. In certain embodiments, R 32a is -A 1 -A 2 -B. In certain embodiments, R 32a is -L z -A 1 -L 1 -A 2 -L 3 -B, wherein L 2 and A 1 are absent. In certain embodiments, R 32a is -L 2 -A 1 -L 1 -A 2 -L 3 -B, wherein L 2 is absent. In certain embodiments, R Za is -L 2 -A 1 -L 1 -A 2 -L -B, wherein L 3 is absent. [0082] As described above, each L 1 is independently selected from

[0083] As described above for Formula la, each L 1 is independently selected from 84] As described above for Formula Ic, each L 1 is independently selected from

)85] In certain embodiments, L 1 i iss

[0086] in certain embodiments,

In certain embodiments, L 1 is In certain . ents, L 1 is In certain embodiments, certain embodiments,

)89] In certain embodiments, L f i ; c s. . In certain

O

Ϋ'n-^ O^A embodiments, L 1 is O in certain embodiments L 1 is O

In certain embodiments,

As described above, L 2 and L 3 are independently absent or are independently selected from l 1 L

[0091] As described above for Formula la and Ic, L 2 and L 3 are independently absent or are independently selected from

[0092] In certain embodiments, L, 2 is absent. In certain embodiments, L, 2 is ,

In certain embodiments, certain embodiments, a certa n embodiments, L certain embodiments,

3 . In certain

O embodiments, L 2 is . In certain embodiments, L 2 is O ,

In certain embodiments, certain embodiments I, 3 is In certain embodiments, L certain embodiments, In certain embodiments, L 3 is . In certain

O embodiments, . In certain embodiments, L 3 is In certain embodiments, L 2 is In certain embodiments, L 2 is ,

[0099] In certain embodiments,

As described above, A 1 and A 2 are independently absent or are independently selected from each Q is independently 1 to 3 rings selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; each X is independently absent or 1 to 2 rings selected from arylene, cyeloalkylene, heteroarylene, and beterocyclylene; each X 1 is independently a heteroarylene or heteroeyclylene ring; each W is independently absent or 1 to 2 rings selected from arylene, cyeloalkylene, heteroarylene, and heteroeyclylene; each W l is independently a heteroarylene or heteroeyclylene ring; each G is independently absent or a ring selected from arylene, cyeloalkylene, heteroarylene, and heteroeyclylene; each G 1 and G 2 are independently heteroarylene or heteroeyclylene ring.

[00103] As described above for Formula la, A 1 and A 2 are independently absent or are independently selected from each Q is independently 1 to 3 rings selected from arylene, cycloalkylene, heteroarylene, and beterocyclylene; each X is independently absent or 1 to 2 rings selected from arylene, cycloalkylene heteroarylene, and heterocyclylene; each X 1 is independently a heteroarylene or lieterocyclylene ring; each W is independently absent or 1 to 2 rings selected from arylene cycloalkylene, heteroarylene, and lieterocyclylene; each W 1 is independently a heteroarylene or lieterocyclylene ring; each G is independently absent or a ring selected from arylene, cycloalkylene, heteroarylene, and lieterocyclylene; each G 1 and G 2 are independently heteroarylene or lieterocyclylene ring.

[00104] As described above for Formula Ic, A 1 and A 2 are independently absent or are independently selected from

each Q is independently 1 to 3 rings selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; each X is independently absent or 1 to 2 rings selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; each X s is independently a heteroarylene or heterocyclylene ring; each W is independently absent or 1 to 2 rings selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; each W 1 is independently a heteroarylene or heterocyclylene ring; each G is independently absent or a ring selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; each G 1 and G 2 are independently heteroarylene or heterocyclylene ring;

[00105] For Formula I, the bond on the left side of A 1 , as drawn, is bound to -C(—Z 1 )- or L 2 ; and the bond on the right side of the A 2 moiety, as drawn, is bound to B or L 3 . For Formula II, the bond on the left side of A 1 , as drawn, is bound to and the bond on the right side of the A 2 moiety, as drawn, is bound to B. For Formula la and Ic, the bond on the left side of A 1 , as drawn, is bound to C(=Z 1 )- or L 2 ; and wherein the bond on the right side of the A 2 moiety, as drawn, is bound to B or L 3 .

[00106] In certain embodiments, A 1 is absent. In certain embodiments. A 1 is , certain embodiments. certain embodiments. A 1 is

[00107] In certain embodiments, AMs s 3 P P , wherein each Q is independently 1 to 3 rings selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene.

[0010S] In certain embodiments, A 1 is , wherein each Q is independently 1 to 3 rings selected from aryiene, cycloalkylene, heteroaryiene, and heterocyclylene .

[00109] In certain embodiments, A 1 wherein each X is independently absent or 1 to 2 rings selected from arylene, cycloalk lene, heteroarylene, and heterocyclylene; and each X 1 is a heteroarylene or heterocyclylene ring

In certain embodiments, A is , wherein each W is independently absent or 1 to 2 rings selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; and each W 3 is a heteroarylene or heterocyclylene ring. wherein each W is independently absent or 1 to 2 rings selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; and each W 3 is a heteroarylene or heterocyclylene ring.

! ~ N G 1 N-|-

[00112] In certain embodiments, A 1 Is — , wherein each G Is independently absent or a ring selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene.

[00113] In certain embodiments, A 3 wherein each G is independently absent or a ring selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; and each G 3 and G 2 are independently heteroarylene or heterocyclylene ring. In certain embodiments, A is , , certain embodiments, certain embodiments, A 2 Is

[00117] In certain embodiments. A 2 is , wherein each Q is independently 1 to 3 rings selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene.

[00118] In certain embodiments, A 2 is , wherein each Q is independently 1 to 3 rings selected from aryiene, cycloalkylene, heteroaryiene, and heterocyclylene .

?N-|C(R 3 ) 2 | - X - l·- X 1 N-f-

[00119] In certain embodiments, A 2 is p , wherein each X is independently absent or 1 to 2 rings selected from arylene, cycloalk lene, heteroarylene, and heterocyclylene; and each X 1 is independently a heteroarylene or heterocyclylene ring , wherein each W is independently absent or 1 to 2 rings selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; and each W 1 is independently a heteroarylene or heterocyclylene ring.

In certain embodiments, A 2 is p , wherein each W is independently absent or 1 to 2 rings selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; and each W 1 is independently a heteroarylene or heterocyclylene ring.

[00122] In certain embodiments, A 2 is , wherein each G is independently absent or a ring selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene.

-1-N G 1 N — G~N G 2 N-f-

In certain embodiments, A 2 is wherein each G is independently absent or a ring selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; and each G 1 and G are independently a heteroarylene or heterocyciylene ring. In certain embodiments, A 2 is ,

As described above, each B is independently selected from

[00128] In certain embodiments, ,

[00130] As described above, each B 1 is independently selected from ^ NR 3 -(C(R 4 ) 2 ) n -, ¾ NR 3 -(C(R 3 ) 2 )n-(C 6 -C 1 o)arylene-(C(R 3 ) 2 ) -, ¾-NR 3 -(C(R 3 ) 2 )n-heteroarylene-, (C 6 -C t o) ar lene- , -heteroarylene- heterocyclylene-(C 6 -Cio)arylene-, ^ heteiOarylene-heterocyclylene-(C 6 -Cio)arylene-, heterocyclyiene— arylene and ^ ~ NR 3 -(C(R 3 ) 2 ) n -S(O) 2- ar lene-C(O)-, wherein the bond on the left side of B 1 , as drawn, is bound to A 2 or L 1 ; and wherein the heteroarylene, heterocyclylene, and arylene are each independently optionally substituted with alkyl, hydroxyalkyi, haloalkyl, allcoxy, halogen, or hydroxyl.

[00131] As described above for Formula Ic, each B 1 is independently selected front )aiylene-(C(R 3 ) 2 )n-, s~ NR 3 -(C(R 3 ) 2 )n- heteroarylene-, 5- NR 3 -(C(R 3 ) 2 )n-heteroarylene-(C(R 3 ) 2 ) -, ^ (Cfi-C 1 olarylene-,

-i- NR 3 -(C(R 3 ) 2 )n-NR 3 C(O)-, ^NR 3 -(C(R 3 ) 2 )n-heteroarylene-heterocyclylene-(C 6 -

Ciojarylene·, > heteroarylene-heterocyclylene-(C 6 -C io)arylene- , ,

S(O) 2- arylene-C(O)-, and ^ NR 3 -(C(R 3 ) 2 )n-S(O) 2 ---arylene-(C(R 3 ) 2 ) wherein the bond on the left side of B 1 , as drawn, is bound to A 2 , L 3 , or L 1 ; and wherein the heteroarylene, heterocyclylene, and arylene are each independently optionally substituted with alkyl, hydroxyalkyl, haloalkyl, alkoxy, halogen, or hydroxyl

[00132] In certain embodiments,

[§§133] In certain embodiments, B 1 is _ jn cer†a ] n embodiments,

B is w ¾ erein arylene is optionally substituted with haloalkyl.

[00134] In certain embodiments,

Cio)arylene-(C(R J ) 2 )n-, ^ NR 3 -(C(R 3 ) 2 )n-heteroarylene-, ^ (C 6 -Cio)arylene-, ^ NR 3 -

(C(R 3 ) 2 ) n -NR 3 C(O)-, ^ NR 3 -(C(R 3 ) 2 ) n -heteroarylene-heterocyclylene-(C 6 -C o)arylene-, or

^ heteroarylene-heterocyclylene-(C 6 -C io)arylene- . In certain embodiments, B 5 is [00135] In certain embodiments, B 1 is . In certain

[00136] In certain embodiments, B 1 is ~ ^ NR 3 -(C(R ) 2 )n-heteroarylene-(C(R ) 2 )n-. hi certain embodiments, certain embodiments, B 1 is In certain embodiments, B 1 is arylene-(C(R 3 ) 2 )n [00137] In certain embodiments, in B 1 , the heteroaryl, heterocyclyl, and arylene are optionally substituted with alkyl, hydroxyalkyl, haloalkyl, alkoxy, halogen, or hydroxyl.

[00138] In certain embodiments, R ¾ is H. In certain embodiments, R 3 is (C 1 -C 6 lalkyl.

[00139] In certain embodiments, R 4 is H. In certain embodiments, R 4 is (C 1 -C 6 jalkyl. In certain embodiments, R 4 is halogen. In certain embodiments, R 4 is 5-12 membered heteroaryl, 5-12 membered heterocyclyl, or (C 6 -C 1 o)aryl, wherein the heteroaryl, heterocyclyl, and aryl are optionally substituted with -N(R ) 2 , -OR 3 , halogen, (C 1 -C 6 )alkyl, - (C 1 -C 6 lalkylene -heteroaryl, -(Ci-C 6 lalkylene-CN, or -C(O)NR 3 -heteroaryl. In certain embodiments, R 4 is -C(O)NR 3 -heterocyclyl. In certain embodiments, R 4 is 5-12 membered heteroaryl, optionally substituted with -N(R 3 ) 2 or -OR 3 .

[00140] As described above, each R 5 is independently H, (C 1 -Cklalkyl, -C(O)OR 3 , or -N(R 3 ) 2 , wherein the alkyl of (Ci-CcOalkyl is optionally substituted with -N(R 3 j2 or -OR 3 . In certain embodiments, R 5 is H. In certain embodiments, R 5 is (C 1 -Crpalkyl, wherein the alkyl is optionally substituted with -N(R 3 ) 2 or -OR 5 . In certain embodiments, R 3 is -C(O)OR 5 . In certain embodiments, R 5 is -N(R ) 2.

[00141] As described above, each R 6 is independently H, (C 1 -Cklalkyl, -C(O)OR 3 , or -N(R 5 ) 2 , wherein the alkyl of (C 1 -C 6 lalkyl is optionally substituted with --N(R 3 ) 2 or -OR 3 . In certain embodiments, R 6 is H. In certain embodiments, R 6 is (C 1 -Crpalkyl, wherein the alkyl is optionally substituted with -N(R 3 ) 2 or -OR 5 . In certain embodiments, R° is -C(O)OR 5 . In certain embodiments, R b is -N(R 3 ) 2 .

[00142] As described above, each R' is independently H, (Ci-Crpalkyl, -C(O)OR 3 , or -N(R 5 ) 2 , wherein the alkyl of (C 1 -C 6 lalkyl is optionally substituted with --N(R 3 ) 2 or -OR 3 . In certain embodiments, R 7 is H. In certain embodiments, R 7 is (C 1 -Crpalkyl, wherein the alkyl is optionally substituted with -N(R 3 ) 2 or -OR 5 . In certain embodiments, R·' is -C(O)OR 5 . In certain embodiments, R 7 is -N(R 3 ) 2 .

[00143] As described above, each R 8 is independently H, (Ci-Crpalkyl, -C(O)OR 3 , or -N(R 5 ) 2 , wherein the alkyl of (C 1 -C 6 lalkyl is optionally substituted with --N(R 3 ) 2 or -OR 3 . In certain embodiments, R 8 is H. In certain embodiments, R 8 is (C 1 -Crpalkyl, wherein the alkyl is optionally substituted with -N(R 3 ) 2 or -OR 5 . In certain embodiments, R 8 is -C(O)OR 5 . In certain embodiments, R 8 is -N(R 3 ) 2 . [00144] As described above, each Y is independently Ct R V or a bond. In certain embodiments, Y is C(R 3 ) 2 In certain embodiments, Y is €¾. In certain embodiments, Y is a bond.

[00145] In certain embodiments, n is 1, 2, 3, 4, 5, 6, 7, or 8, or any range derivable therein. In certain embodiments, n is 1, 2, 3, or 4. In certain embodiments, n is 5, 6, 7, or 8. In certain embodiments, n is 9, 10, 11, or 12.

[00146] In certain embodiments, o is an integer from zero to 10, or any range derivable therein. In certain embodiments, o is 0, 1, 2, 3, 4, or 5. In certain embodiments, o is 6, 7, 8, 9, or 10. In certain embodiments, o is one to 7. In certain embodiments, o is one to 8. In certain embodiments, o is one to 9. In certain embodiments, o is 3 to 8.

[00147] In certain embodiments, o is an integer from zero to 30, or any range derivable therein. In certain embodiments, o is an integer from zero to 30, 29, 28, 27, or 26. In certain embodiments, o is an integer from zero to 25, 24, 23, 22, or 21. In certain embodiments, o is an integer from zero to 20, 19, 18, 17, or 16. In certain embodiments, o is an integer from zero to 15, 14, 13, 12, or 11.

[00148] In certain embodiments, p is 0, 1, 2, 3, 4, 5, or 6, or any range derivable therein.

In certain embodiments, p is 7, 8, 9, 10, 11, or 12. In certain embodiments, p is 0, 1, 2, or 3.

In certain embodiments, p is 4, 5, or 6.

[00149] In certain embodiments, q is an integer from zero to 10, or any range derivable therein. In certain embodiments, q is 0, 1, 2, 3, 4, or 5. In certain embodiments, q is 6, 7, 8, 9, or 10. In certain embodiments, q is one to 7. In certain embodiments, q is one to 8. In certain embodiments, q is one to 9. In certain embodiments, q is 3 to 8.

[00150] In certain embodiments, q is an integer from zero to 30, or any range derivable therein. In certain embodiments, q is an integer from zero to 30, 29, 28, 27, or 26. In certain embodiments, q is an integer from zero to 25, 24, 23, 22, or 21. In certain embodiments, q is an integer from zero to 20, 19, 18, 17, or 16. In certain embodiments, q is an integer from zero to 15, 14, 13, 12, or 11.

[06151] As described above, r is an integer from one to 6. In certain embodiments, r is one. In certain embodiments, r is 2. In certain embodiments, r is 3. In certain embodiments, r is 4. In certain embodiments, r is 5. In certain embodiments, r is 6. [00152] As described above, when R 28 and R 40 are H, then R 32 is not =O. In certain embodiments, the compound is not rapamycin, as shown below:

In certain embodiments, in Formula la or le, R 3 is -O-C(=Z i )-R :>2a . In certain embodiments, R 32 is -O-C(=Z 1 )-R 32a ; wherein R 32a is -A^-A^B; -A 3 -A 2 -B; or L 2 -A l -L l -A 2 -L 3 -B. In certain embodiments, in Formula la or Ic, R 28 is-C^Z^-R 283 . In certain embodiments, R 28 is-C(=Z 1 )-R 28a ; wherein R 28a is -A 1 -L 1 -A 2 -B; -A 5 -A 2 -B; or -L 2 -A 1 -L 1 -A 2 -L 3 -B . In certain embodiments, in Formula la or Ic, R 40 is -C(=Z 1 )-R 4ila . In certain embodiments, R 40 is -€(=Z 1 )-R 40a . wherein R 40a is -A^ΐLA^B; -A 1 - A 2 -B ; or -L 2 - A 1 -L 1 - A 2 -L 3 -B

[00153] The present disclosure provides a compound of Formula la or Ic, or a pharmaceutically acceptable salt or tautomer thereof, having one, two, or three of the following features: a) R 32 is -O-C(=Z 1 )-R 32a ; b) R 28 is--C(=Z i )-R 28a ; c) R 40 is-C(=Z 1 )-R 40a .

[00154] The present disclosure provides a compound of Formula la or Ic, or a pharmaceutically acceptable salt or tautomer thereof, having one, two, or three of the following features: a) R 32 is -O-C^Z^-R 323 ; wherein R >2a is -A 1 -L 1 -A 2 -B; -A l -A 2 -B; or L 2 -A 1 -L 1 -A 2 -L 3 -B; b) R 28 is-C(=Z 3 )-R 28a ; wherein R 28a is -A 1 -!. 1 - A 2 -B ; -A 3 -A 2 -B; or -L 2 -A 1 -L 1 -A 2 -L 3 -B; c) R 40 is-C(=Z 1 )-R 40a . wherein R 408 is -A^L^A^B; -A 3 -A 2 -B; or

-L 2 -A 1 -L 1 -A 2 -L 3 -B. [00155] The present disclosure provides a compound of Formula la or Ic, or a pharmaceutically acceptable salt or tautomer thereof, having one, two, or three of the following features: a) R 40 is -C(=Z 1 )-R 40a ; b) R 40a is -A 1 -ΐ A 2 -B; -A l -A 2 -B; -I A^Ϊ A^-B; c) R 32 is -OR 3 , such as -OH.

[00156] The present disclosure provides a compound of Formula la or Ic, or a pharmaceutically acceptable salt or tautomer thereof, having one, two, three, or four of the following features: a) one of R 28a , R 32a , and R 40a is -A’-l -A^B; b) A 1 is absent; c) A 2 is absent; g) R 4 is 5-12 membered heteroaryl, optionally substituted with -N(R 3 4 ) 2 or -OR >3 J .

[00157] The present disclosure provides a compound of formula: or a pharmaceutically acceptable salt or tautomer thereof, having one, two, three, or four of the following features: a) Z 1 is O; b) A J is absent; f) R is 5-12 membered heteroaryl optionally substituted with -N(R 3 ) 2 or -OR 3 ; and g) R 32 is =O .

In the above R 40 * can be -A^ΐ A^B; -A 1 -A 2 -B; or -L 2 -A 1 -L 1 -A 2 -L 3 -B

[00158] The present disclosure provides a compound of formula: or a pharmaceutically acceptable salt or tautomer thereof, having one two, three, or four of the following features: a) Z 1 is O; b) A 1 is absent; f) R 4 is 5-12 membered heteroaryl, optionally substituted with - N(R 3 ) 2 or -OR and g) R 32 is -OH.

In the above, R 40a can be -A^ΐ A^B; -A 1 -A 2 -B; or -L 2 - A i -L 1 - A 2 -L 3 -B . [00159] The present disclosure provides a compound of formula: or a pharmaceutically acceptable salt or tautomer thereof, having one two, three, or four of the following features: a) Z 1 is O; f) R 4 is 5-12 membered heteroaryl, optionally substituted with -N(R 3 ) 2 or -OR 3 ; and g) R 32 is =O.

In the above, R 40a can be -A 1 -L 1 -A 2 -B; -A 1 -A 2 -B; or -L 2 -A i -L 1 -A 2 -L 3 -B.

[00160] The present disclosure provides a compound of formula: or a pharmaceutically acceptable salt or tautomer thereof, having one, two three, or four of the following features: a) Z 1 is O; b) A 1 is absent;

^ NR 3 --(C(R :S ) 2 ) r -S(O) 2--- arylene-C(O)--, wherein the arylene is optionally substituted with alkyl, hydroxyalkyl, haloalkyl, alkoxy, halogen, or hydroxyl; f) R 4 is 5-12 membered heteroaryl, optionally substituted with -N or -OR 3 ; and g) R 32 is -OH.

In the above, R 40a can be -A 1 -L 1 -A 2 -B; -A 1 -A 2 -B; or -L 2 -A i -L 1 -A 2 -L 3 -B.

[00161] The present disclosure provides a compound of formula: or a pharmaceutically acceptable salt or tautomer thereof, having one, two, three, or four of the following features: a) 7) is O; r S(O) 2--- arylene-C(O)---, wherein the arylene is optionally substituted with alkyl, hydroxyalkyl, haloalkyl, alkoxy, halogen, or hydroxyl; g) R 4 is 5-12 memhered heteroaryl, optionally substituted with -N or -OR 3 ; and h) R 32 is -OH.

In the above, R 40a can be -A 1 -L 1 -A 2 -B; A 1 -A 2 -B; or -L 2 -A i -L 1 -A 2 -L 3 -B. [00162] In certain embodiments, in Formula la or Ic, R 40a is any organic moiety, which may have a molecular weight (e.g. the sum of the atomic masses of the atoms of the substituent) of less than 15 g/mol, 50 g/mol, 100 g/mol, 150 g/mol, 200 g/mol, 250 g/mol, 300 g/mol, 350 g/mol, 400 g / mol, 450 g/mol, or 500 g/mol.

[0(1163] In certain embodiments, the present disclosure provides for a compound selected from below or a pharmaceutically acceptable salt or tautomer thereof. 't

 

c _

[00164] In certain embodiments, the present disclosure provides for a compound selected from below or a pharmaceutically acceptable salt or tautomer thereof,

a a

a s:

ŭ o »

s;

[00165] In certain embodiments, the present disclosure provides for a compound selected from below or a pharmaceutically acceptable salt or tautomer thereof,

1 s c

ss:

[00166] The compounds of the disclosure may include pharmaceutically acceptable salts of the compounds disclosed herein. Representative “pharmaceutically acceptable salts” may include, e.g., water-soluble and water-insoluble salts, such as the acetate, amsonate (4,4- diaminostilbene-2, 2-disulfonate), benzenesulfonate, benzonate, bicarbonate, bisulfate, bitartrate, borate, bromide, butyrate, calcium, calcium edetate, camsylate, carbonate, chloride, citrate, elavulariate, dihydrochloride, edetate, edisylate, estolate, esylate, fiunarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexafluorophosphate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, sethionate, lactate, lactobionate, laurate, magnesium, malate, maleate, mandelate, mesylate, methy [bromide, methylnitrate, methylsulfate, mucate, napsylate, nitrate, N-methylglucamine ammonium salt 3-hydroxy-2-naphthoate, oleate, oxalate, palmitate, pamoate, 1,1-methene-bis-2-hydroxy-3- naphthoate einbonate, pantothenate, phosphate/diphosphate picrate, polygalacturonate, propionate, p-toluenesulfonate, salicylate, stearate, subacetate, succinate, sulfate, sulfosalicylate, suramate, tannate, tartrate, teoclate, tosylate, triethiodide, and valerate salts.

[00Ϊ67] “Pharmaceutically acceptable salt” may also include both acid and base addition salts. “Pharmaceutically acceptable acid addition salt” may refer to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which may be formed with inorganic acids such as, but are not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as, but not limited to, acetic acid, 2,2-dichioroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 4- acetamidobenzoic acid, camphoric acid, camphor-10- sulfonic acid, capric acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, dodecylsulfuric acid, ethane-1,2-cii sulfonic acid, ethanesulfonic acid, 2-hydroxyethanesulfonic acid, formic acid, fumaric acid, gaiactaric acid, gentisic acid, glucoheptonic acid, gluconic acid, glucuronic acid, glutamic acid, glutaric acid, 2-oxo-glutaric acid, glycerophosphoric acid, glycolic acid, hippuric acid, isobutyric acid, lactic acid, lactobionic acid, lauric acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesulfonic acid, mucic acid, naphthalene-1, 5- disulfonic acid, naphthalene-2-sulfonic acid, 1-hydroxy-2-naphthoic acid, nicotinic acid, oleic acid, orotic acid, oxalic acid, palmitic acid, parnoic acid, propionic acid, pyroglutamic acid, pyruvic acid, salicylic acid, 4- aminosalicylic acid, sebacic acid, stearic acid, succinic acid, tartaric acid, thiocyanic acid, p-toluenesulfonic acid, trifluoroacetic acid, undecylenic acid, and the like.

[00168] “Pharmaceutically acceptable base addition salt” may refer to those salts that retain the biological effectiveness and properties of the free acids, which are not biologically or otherwi se undesirable. These salts may be prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases may include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. For example, inorganic salts may include, but are not limited to, ammonium, sodium, potassium, calcium, and magnesium salts. Salts derived from organic bases may include, but are not limited to, salts of primary, secondary', and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, diethanolamine, ethanolamine, deanol, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine, methylglueamine, theobromine, triethanolamine, tromethamine, purines, piperazine, piperidine, N-ethylpipexidine, polyamine resins and the like.

[00169] Unless otherwise stated, structures depicted herein may also include compounds which differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structure except for the replacement of a hydrogen atom by s deuterium or tritium, or the replacement of a carbon atom by i3 C or i4 C, or the replacement of a nitrogen atom by 15 N, or the replacement of an oxygen atom with 17 0 or 18 0 are within the scope of the disclosure. Such isotopically labeled compounds are useful as research or diagnostic tools.

[00170] In some embodiments, one or more deuterium atoms may be introduced into the PEG moiety of any compound of the present invention. Mechanisms for such modifications are known in the art starting from commercially available starting materials, such as isotopically enriched hydroxylamine building blocks. In some embodiments, a tritium or a deuterium may be introduced at the C 3 2 position of compounds of the present invention using, for example, a commercially available isotopically pure reducing agent and methods known to those in the art. In some embodiments, 14 C may be introduced into the C40 carbamate moiety of compounds of the present invention using commercially available materials and methods known to those of skill in the art. In some embodiments, an isotope such as deuterium or tritium may be introduced into the R 40a substituent of a compound of Formula la, Ic, I or II, using commercially available starting materials and methods known to those of skill in the art.

Methods of Synthesizing Disclosed Compounds

[00171] The compounds of the present disclosure may be made by a variety of methods, including standard chemistry. Suitable synthetic routes are depicted in the schemes given below.

[00172] The compounds of any of the formulae described herein may be prepared by methods known in the art of organic synthesis as set forth in part by the following synthetic schemes and examples. In the schemes described below', it is well understood that protecting groups for sensitive or reactive groups are employed where necessary in accordance with general principles or chemistry. Protecting groups are manipulated according to standard methods of organic synthesis (T. W. Greene and P. G. M. Wuts, "Protective Groups in Organic Synthesis”, Third edition, Wiley, New' York 1999). These groups are removed at a convenient stage of the compound synthesis using methods that are readily apparent to those skilled in the ait. The selection processes, as well as the reaction conditions and order of their execution, shall be consistent with the preparation of compounds of Formula I, la, l b, II, or lib, or a pharmaceutically acceptable salt or tautomer of any of the foregoing. [00173] The compounds of any of the formulae described herein may be prepared by methods which avoid the use of metal -mediated cycloaddition reactions which require the use of azide-containing compounds. Azide containing compounds present potential safety hazards associated with their preparation and storage (e.g., explosion due to high energy decomposition). Also, the reaction schemes herein can avoid the use of copper or ruthenium metals in the penultimate or ultimate synthetic steps, which can be advantageous. Avoiding the use of copper or ruthenium metals in the penultimate or ultimate synthetic steps reduces the potential for contamination of the final compounds with undesirable metal impurities.

[00174] As rapamycin can be an expensive starting material, good yields on reactions are advantageous. The reaction schemes herein provide better yields than other reaction schemes. In the reaction schemes herein, there is no need to alkylate at the C40-hydroxyl of rapamycin, which is advantageous for providing as much as a 5-fold improved overall yield in preparing bivalent compounds from rapamycin compared to other reaction schemes.

[00175] There is an additional synthetic improvement associated with better yields. Avoiding the need to alkylate at the C40-hydroxyl gives as much as a 5-fold improved overall yield in preparing bivalent compounds from rapamycin.

[00176] Those skilled in the art will recognize if a stereocenter exists in any of the compounds of the present disclosure. Accordingly, the present disclosure may include both possible stereoisomers (unless specified in the synthesis) and may include not only racemic compounds but the individual enantiomers and/or diastereomers as well. When a compound is desired as a single enantiomer or diastereomer, it may be obtained by stereospecific synthesis or by resolution of the final product or any convenient intermediate. Resolution of the final product, an intermediate, or a starting material may be effected by any suitable method known in the art. See, for example, "Stereochemistry of Organic Compounds" by E.

L. Eliel, S. H. Wilen, and L. N. Mander (Wiley-lnterscience, 1994).

Preparation of Compounds

[00177] The compounds described herein may be made from commercially available starting materials or synthesized using known organic, inorganic, and/or enzymatic processes.

[00178] The compounds of the present disclosure can be prepared in a number of ways well known to those skilled in the art of organic synthesis. By way of example, compounds of the disclosure can be synthesized using the methods described below, together with synthetic methods known in the art of synthetic organic chemistry, or variations thereon as appreciated by those skilled in the art. These methods may include but are not limited to those methods described below.

[00179] The term “tautomers” may refer to a set of compounds that have the same number and type of atoms, but differ in bond connectivity and are in equilibrium with one another. A “tautomer” is a single member of this set of compounds. Typically a single tautomer is drawn but it may be understood that this single structure may represent ail possible tautomers that might exist. Examples may include enol-ketone tautomerism. When a ketone is drawn it may be understood that both the enol and ketone forms are part of the disclosure.

[00180] In addition to tautomers that may exist at all amide, carbonyl, and oxime groups within compounds of Formula I, la, ft, Ic, II, or lib, compounds in this family readily interconvert via a ring-opened species between two major isomeric forms, known as the pyran and oxepane isomers (shown below). This interconversion can be promoted by magnesium ions, mildly acidic conditions, or alkyl amine salts, as described in the following references: i) Hughes, P. F.; Musser, J.; Conklin, M ; Russo, R. 1992. Tetrahedron Lett. 33(33): 4739-32. ii) Zhu, T. 2007. U.S. Patent 7, 241, 771; Wyeth iii) Hughes, P.F. 1994. U.S. Patent 5,344,833; American Home Products Corp. The scheme below shows an interconversion between the pyran and oxepane isomers in compounds of Formula i, la, lb,

Ic, II, or lib.

Ring-opened species

[00181] As this interconversion occurs under mild condition, and the thermodynamic equilibrium position may vary between different members of compounds of Formula I, la, ft, Ic, II, or lib, both isomers are contemplated for the compounds of Formula I, la, ft, Ic, II, or lib. For the sake of brevity, the pyran isomer form of ail intermediates and compounds of Formula I, la, lb, Ic, ΪΪ, or Ilh is shown.

General Assembly Approaches For Bifunctional Rapalogs

[§§182] With reference to the schemes below, rapamycin is Formula RAP, where R 16 is -OCH3; R 26 is =O; R 28 is -OH; R 32 is =O; and R 40 is -OH. A “rapalog” refers to an analog or derivative of rapamycin. For example, with reference to the schemes below, a rapalog can be rapamycin that is substituted at any position, such as R 16 , R 2 °, R 28 , R 32 , or R 40 . An active site inhibitor (AS inhibitor) is an active site mTOR inhibitor. In certain embodiments, AS inhibitor is depicted by B, in Formula I, la, lb, Ic, II, or lib.

Series 1 bifunctional rapalogs

[00183] A general structure of Series 1 bifunctional rapalogs is shown in Scheme 1 below. For these types of bifunctional rapalogs, the linker may include variations where q - 0 to 30, such as q = 1 to 7, and r = 1 to 6. The linker amine can include substitutions, such as R = H and C1-C6 alky] groups. The carbamate moiety, where Z 1 - O or S, can be attached to the rapalog at R 40 or R 8 (Formula I, la, lb, Ic, II, or lib), including variations found in Table 1 in the Examples Section. An mTOR active site inhibitor can attach to the linker via a primary or secondary amine, and may include variations found in Table 2 in the Examples Section.

Scheme 1. Series 1 bifnnctional rapalogs. Series 2 hifunctional rapalogs

[00184] A general structure of Series 2 bifunctional rapalogs is shown in Scheme 2 below. For these types of bifunctional rapalogs, the linker may include variations where q - 0 to 30, such as q = 1 to 7. The linker amine can include substitutions, such as R = H and C1-C6 alkyl groups. The pre-linker amine can include substitutions, such as R 2 = H, C1-C6 alkyl groups, and cycloalkyl including 4 to 8-membered rings. The carbamate moiety, where Z 1 = O or S can be attached to the rapalog at R 4t ' or R 28 (Formula I la, lb, Ic, II, or lib), including variations found in Table 1 in the Examples Section. An mTOR active site inhibitor can attach to the linker via a primary or secondary amine, and may include variations found in Table 2 in the Examples Section.

Scheme 2. Series 2 hifunctional rapalogs. amine containing pre-linker

Series 3 hifunctional rapalogs

[00185] A general structure of Series 3 bifunctional rapalogs is shown in Scheme 3 below. For these types of hifunctional rapalogs, the linker may include variations where q - 0 to 30, such as q = 1 to 7. The linker amine can include substitutions, such as R = H and C1-C6 alkyl groups. The post-linker amine can include substitutions, such as R 2 = H, C1-C6 alkyl groups, and cycloalkyl including 4 to 8-membered rings. The carbamate moiety, where Z 1 = O or S, can be attached to the rapalog at R 40 or R 28 (Formula I, la, lb, Ic, II, or lib), including variations found in Table 1 in the Examples Section. An mTOR active site inhibitor can attach to the linker via a primary or secondary amine, and may include variations found in Table 2 in the Examples Section.

Scheme 3. Series 3 bifunctional rapalogs amine containing post-linker

Series 4 bifunctional rapalogs

[00186] A general structure of Series 4 bifunctional rapalogs is shown in Scheme 4 below. For these types of bifunctional rapalogs, the linker may include variations where q = 0 to 30, such as q - 1 to 7. The linker amine can include substitutions, such as R - H and C1-C6 alkyl groups. The pre- and post-linker amines can each include substitutions, such as R 2 = H, C1-C6 alkyl groups, and cycloalkyl including 4 to 8-membered rings. The carbamate moiety, where Z f = O or S, can be attached to the rapaiog at R 40 or R 28 (Formula I, la, lb, Ic, II, or lib), including variations found in Table 1 in the Examples Section. An mTOR active site inhibitor can atach to the linker via a primary or secondary amine, and may include variations found in Table 2 in the Examples Section.

Scheme 4. Series 4 bifunctional rapalogs amine containing amine containing pre-linker

[00187] A general structure of Series 5 bifunctional rapalogs is shown in Scheme 5 below. For these types of bifunctional rapalogs, the pre-linker amine can include substitutions, such as R 2 = H, C1-C6 alkyl groups, and cycloalkyl including 4 to 8-membered rings. The carbamate moiety, where Z 1 - O or S, can be attached to the rapaiog at R 40 or R 28 (Formula I, la, lb, Ic, II, or lib), including variations found in Table 1 in the Examples Section. An mTOR active site inhibitor can attach to the linker via a primary or secondary amine, and may include variations found in ' Fable 2 in the Examples Section. Scheme 5. Series 5 bifunctional rapalogs amine containing

Series 6 bi functional rapalogs

[00188] A general structure of Series 6 bifunctional rapalogs is shown in Scheme 6 below'. For these types of bifunctional rapalogs, the linker may include variations where q = 0 to 30, such as q = 1 to 7. The linker amines can include substitutions, such as R = H and C1-C6 alkyl groups. The post-linker amine can include substitutions, such as R - H, C 1 -C6 alkyl groups, and cycloalkyl including 4 to 8-membered rings. The carbamate moiety, where Z 1 = O or S, can be attached to the rapalog at R 40 or R 28 (Formula I, la, ft, Ic, II, or lib), including variations found in Table 1 in the Examples Section. An mTOR active site inhibitor can attach to the linker via a primary' or secondary ' - amine, and may include variations found in Table 2 in the Examples Section.

Scheme 6. Series 6 bifunctional rapalogs. amine containing post-linker

Series 7 bifunctional rapalogs

[00189] A general structure of Series 7 bifunctional rapalogs is shown in Scheme 7 below. For these types of bifunctional rapalogs, the linker may include variations where q = 0 to 30, such as q - 1 to 7. The linker amine can include substitutions, such as R - H and C 1 -C6 alkyl groups. The pre- and post-linker amines can each include substitutions such as R 2 = H, C1-C6 alkyl groups, and cycloalkyl including 4 to 8-membered rings. The carbamate moiety, where Z 1 = C) or S, can be attached to the rapalog at R 40 or R 28 (Formula I, la, lb, Ic, II, or s; lib), including variations found in Table 1 in the Examples Section. An mTOR active site- inhibitor can attach to the linker via a primary or secondary amine, and may include variations found in Table 2 in the Examples Section.

Scheme 7. Series 7 bifnnclional rapalogs

Series 8 bifunctional rapalogs

[§§190] A general structure of Series 8 bifunctional rapalogs i shown in Scheme 8 below. For these types of bifunctional rapalogs, the linker may include variations where q = 0 to 30, such as q = 1 to 7 The linker amine can include substitutions, such as R = H and C1---C6 alkyl groups. The post-linker amine can include substitutions, such as R 2 = H, C1-C6 alkyl groups, and cycloalkyl including 4 to 8-membered rings. The carbamate moiety, where Z 1 ----- O or S, can be attached to the rapalog at R 40 or R 28 (Formula I, la, lb, Ic, II, or lib), including variations found in Table 1 in the Examples Section. An mTOR active site inhibitor can attach to the linker via a primary or secondary ' · amine, and may include variations found in Table 2 in the Examples Section.

Scheme 8. Series 8 bifunctional rapalogs amine containing post-tinker amine containing post-tinker

Pharmaceutical Compositions

[§§191] Another aspect provides a pharmaceutical composition including a pharmaceutically acceptable excipient and a compound of the present invention, or pharmaceutically acceptable salt or tautomer thereof. [00192] In embodiments of the pharmaceutical compositions, a compound of the present invention, or a pharmaceutically acceptable salt or tautomer thereof, may be included in a therapeutically effective amount.

[00193] Administration of the disclosed compounds or compositions can be accomplished via any mode of administration for therapeutic agents. These modes may include systemic or local administration such as oral, nasal, parenteral, transdermal, subcutaneous, vaginal, buccal, rectal, topical, intrathecal, or intracranial administration modes.

[00194] In certain embodiments, administering can include oral administration, administration as a suppository, topical contact, intravenous, parenteral, intraperitoneal, intramuscular, intralesional, intrathecal, intracranial, intranasal or subcutaneous administration, or the implantation of a slow-release device, e.g., a mini-osmotic pump, to a subject. Administration can be by any route, including parenteral and transmucosal (e.g., buccal, sublingual, palatal, gingival, nasal, vaginal, rectal, or transdermal). Parenteral administration includes, e.g., intravenous, intramuscular, intra-arteriole, iniradermal, subcutaneous, intraperitoneal, intraventricular, and intracranial. Other modes of delivery include, but are not limited to, the use of liposomal formulations, intravenous infusion, transdermal patches, etc. The compositions of the present disclosure can be delivered by transdermally, by a topical route, formulated as applicator sticks, solutions, suspensions, emulsions, gels, creams, ointments, pastes, jellies, paints, powders, and aerosols. Oral preparations include tablets, pills, powder, dragees, capsules, liquids, lozenges, cachets, gels syrups, slurries, suspensions, etc., suitable for ingestion by the patient. Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories, and dispersible granules. Liquid form preparations include solutions, suspensions, and emulsions, for example, water or water/propylene glycol solutions. The compositions of the present disclosure may additionally include components to provide sustained release and/or comfort. Such components include high molecular weight, anionic rnucomimet.ic polymers, gelling polysaccharides and finely-divided drag carrier substrates. These components are discussed in greater detail in U.S. Pat. Nos. 4,911,920; 5,403,841; 5,212,162; and 4,861,760. The entire contents of these patents are incorporated herein by reference in their entirety for al l purposes. The compositions of the present disclosure can also be delivered as microspheres for slow release in the body. For example, microspheres can be administered via iniradermal injection of drug-containing micro spheres, which slowly release subcutaneously (see Rao, J. Biomater Set Polym. Ed. 7:623-645, 1995; as biodegradable and injectable gel formulations (see, e.g., Gao Pharm. Res. 12:857-863, 1995); or, as microspheres for oral administration (see, e.g , Ey!es, J. Pharm. Pharmacol. 49:669-674, 1997). In another embodiment, the formulations of the compositions of the present disclosure can be delivered by the use of liposomes which fuse with the cellular membrane or are endocytosed, i.e., by employing receptor ligands attached to the liposome, that bind to surface membrane protein receptors of the cell resulting in endocytosis. By using liposomes, particularly where the liposome surface carries receptor ligands specific for target cells, or are otherwise preferentially directed to a specific organ, one can focus the delivery of the compositions of the present invention into the target cells in vivo. (See, e.g., Al-Muhammed, J. Microencapsul. 13:293-306, 1996; Chonn, Curr. Opin. Biotechnol. 6:698-708, 1995; Ostro, Am. J. Hosp. Pharm. 46: 1576- 1587, 1989). The compositions of the present disclosure can also be delivered as nanoparticles.

[00195] Depending on the intended mode of administration, the disclosed compounds or pharmaceutical compositions can be in solid, semi-solid or liquid dosage form, such as, for example, injectables, tablets, suppositories, pills, time-release capsules, elixirs, tinctures, emulsions, syrups, powders, liquids, suspensions, or the like, sometimes in unit dosages and consistent with conventional pharmaceutical practices. Likewise, they can also be administered in intravenous (both bolus and infusion), intraperitoneal, intrathecal, subcutaneous or intramuscular form, and all using forms well known to those skilled in the pharmaceutical arts.

[00196] Illustrative pharmaceutical compositions are tablets and gelatin capsules comprising a compound of the disclosure and a pharmaceutically acceptable carrier, such as a) a diluent, e.g., purified water, triglyceride oils, such as hydrogenated or partially hydrogenated vegetable oil, or mixtures thereof, com oil, olive oil, sunflower oil, safflower oil, fish oils, such as EPA or DHA, or their esters or triglycerides or mixtures thereof, omega- 3 fatty acids or derivatives thereof, lactose, dextrose, sucrose, mannitol, sorbitol, cellulose, sodium, saccharin, glucose and/or glycine; b) a lubricant, e.g., silica, talcum, stearic acid, its magnesium or calcium salt, sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and/or polyethylene glycol; for tablets also; c) a binder, e.g., magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, magnesium carbonate, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, waxes and/or polyvinylpyrrolidone, if desired; d) a dismtegrant, e.g., starches, agar, methyl cellulose, bentonite, xanthan gum, alginic acid or its sodium salt, or effervescent mixtures; e) absorbent, colorant, flavorant and sweetener; 1) an emulsifier or dispersing agent, such as Tween 80, Labrasol, HPMC, DOSS, caproyl 909, labrafac, labrafil, peceol transcutol, capmul MCM, capmul PG-12, captex 355, gelucire, vitamin E TOPS or other acceptable emulsifier; and/or g) an agent that enhances absorption of the compound such as cyclodextrin, hydroxypropyl-cyclodextrin, PEG400, PEG200.

[00197] Liquid, particularly injectable, compositions can, for example, be prepared by dissolution, dispersion, etc. For example, the disclosed compound is dissolved in or mixed with a pharmaceutically acceptable solvent such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like, to thereby form an injectable isotonic solution or suspension. Proteins such as albumin chylomicron particles, or serum proteins can be used to solubilize the disclosed compounds.

[00198] The disclosed compounds can be also formulated as a suppository that can be prepared from fatty emulsions or suspensions; using polyalkylene glycols such as propylene glycol, as the carrier.

[00199] The disclosed compounds can also be administered in the form of liposome delivery systems such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles. Liposomes can be formed from a variety of phospholipids, containing cholesterol stearylamine or phosphatidylcholines. In some embodiments, a film of lipid components is hydrated with an aqueous solution of drug to a form lipid layer encapsulating the drug, as described for instance in U.S. Pat. No. 5,262,564, the contents of which are hereby incorporated by reference.

[00200] Disclosed compounds can also be deli vered by the use of monoclonal antibodies as individual carriers to which the disclosed compounds are coupled. The disclosed compounds can also be coupled with soluble polymers as targetable drug earners. Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol, polyhydroxyethylaspanamidephenol, or polyethyleneoxidepolylysine substituted with palmitoyl residues. Furthermore, the disclosed compounds can be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, poly lactic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross-linked or amphipathic block copolymers of hydrogel s. In one embodiment, disclosed compounds are not covalently bound to a polymer, e.g., a polycarboxylic acid polymer, or a polyacrylate.

[00201] Parenteral injectable administration is generally used for subcutaneous, intramuscular or intravenous injections and infusions. Injectables can be prepared in conventional forms, either as liquid solutions or suspensions or solid forms suitable for dissolving in liquid prior to injection.

[00202] Another aspect of the disclosure relates to a pharmaceutical composition comprising a compound, or a pharmaceutically acceptable salt or tautomer thereof, of the present disclosure and a pharmaceutically acceptable carrier. The pharmaceutically acceptable carrier can further include an excipient, diluent, or surfactant.

[00203] Compositions can be prepared according to conventional mixing, granulating or coating methods, respectively, and the present pharmaceutical compositions can contain from about 0.1% to about 99%, from about 5% to about 90%, or from about 1% to about 20% of the disclosed compound by weight or volume. mTOR ami Methods of Treatment

[00204] The term “mTOR” refers to the protein “mechanistic target of rapamycin (serine/threonine kinase)” or “mammalian target of rapamycin.” The term “mTOR” may include both the wild-type form of the nucleotide sequences or proteins as well as any mutants thereof. In some embodiments, “mTOR” is wild-type mTOR. In some embodiments, “mTOR” is one or more mutant forms. The term “mTOR” XYZ may refer to a nucleotide sequence or protein of a mutant mTOR wherein the Y numbered amino acid of mTOR that normally has an X amino acid in the wildtype, instead has a Z amino acid in the mutant. In embodiments, an mTOR is the human mTOR.

[00205] The term “mTORC 1 ” refers to the protein complex including mTOR and Raptor (regulatory -associated protein of mTOR). mTORC 1 may also include ML8T8 (mammalian lethal with SEC 13 protein 8), PRAS40, and/or DEPTOR. mTORC 1 may function as a nutrient/energy/redox sensor and regulator of protein synthesis. The ter “mTORC 1 pathway” or “mTORC 1 signal transduction pathway” may refer to a cellular pathway including mTORC 1 . An mTORC 1 pathway includes the pathway components upstream and downstream from mTORC 1 . An mTORC 1 pathway is a signaling pathway that is modulated by modulation of mTORC 1 activity. In embodiments, an mTORC 1 pathway is a signaling pathway that is modulated by modulation of mTORC 1 activity but not by modulation of n o t mT0RC2 activity. In embodiments, an mTORC 1 pathway is a signaling pathway that is modulated to a greater extent by modulation of mTORC l activity than by modulation of mTORC2 activity.

[00206] The term “mTORC2” refers to the protein complex including mTOR and RICTOR (rapamyein-insensitive companion of mTOR). mTORC2 may also include GpL, mSINl (mammalian stress·· activated protein kinase interacting protein 1), Protor 1/2, DEPTOR, TTI1, and/or TEL2. mTORC2 may regulate cellular metabolism and the cytoskeleton. The term “mTORC2 pathway” or “mTORC2 signal transduction pathway” may refer to a cellular pathway including mTORC2. An mTORC2 pathway includes the pathway components upstream and downstream from mTORC2. An mTORC2 pathway is a signaling pathway that is modulated by modulation of mTORC2 activity. In embodiments, an mTORC2 pathway is a signaling pathway that is modulated by modulation of mTORC2 activity but not by modulation of mTORC 1 activity. In embodiments, an mTORC2 pathw-ay is a signaling pathway that is modulated to a greater extent by modulation of mTORC2 activity than by modulation of mTORC 1 activity.

[00207] The term “rapamycin” or “sirolimus” refers to a macrolide produced by the bacteria Streptomyces hygroseopicus. Rapamycin may prevent the activation of T cells and B cells. Rapamycin has the IUPAC name (3S,6/i,7E,9/i, 10 R, 12R, 145, 15E, 17E, 19E,215,235,26R,27RJ4a5)- 9, 10, 12, 13, 14 21,22,23,24,25 26,27,32,33,34,34a- hexadecahydro-9,27 -dihydroxy-3 -[(LR)-2-[( 1 5,3 R,4R )-4-hydroxy-3 -methoxycyclohexyl] - 1 -methylethyl] - 10,21 -dimethoxy-6,8, 12, 14,2026-hexamethyl-23,27-epoxy-3H-pyrido[2, l-c][1,4]-oxaazacyclohentriacontine-l,5, 1 l,28,29(4H,6H,31 )-pentone. Rapamycin has the CAS number 53123-88-9. Rapamycin may be produced synthetically (e.g , by chemical synthesis) or through use of a production method that does not include use of Streptomyces hygroseopicus.

[00208] “Analog” is used in accordance with its plain ordinary meaning within chemistry and biology and refers to a chemical compound that is structurally similar to another compound (i.e., a so-called “reference” compound) but differs in composition e.g., in the replacement of one atom by an atom of a different element, or in the presence of a particular functional group, or the replacement of one functional group by another functional group, or the absolute stereochemistry of one or more chiral centers of the reference compound, including isomers thereof. [00209] The term “rapamycin analog” or “rapalog” refers to an analog or derivative (e.g., a prodrug) of rapamycin

[00210] The terms “active site mTOR inhibitor” and “ATP mimetic” refers to a compound that inhibits the activity of mTOR (e.g., kinase activity) and binds to the active site of mTOR (e.g., the ATP binding site, overlapping with the ATP binding site, blocking access by ATP to the ATP binding site of mTOR). Examples of active site mTOR inhibitors include, but are not limited to, GNK128, PP242, PP12I, MLNQ128, AZD8055, AZD2014, NVP-BEZ235, BGT226, SF1126, Toxin 1, Torin 2, WYE 687, WYE 687 salt (e.g., hydrochloride), PF04691502, PI-103, CC-223, 081-027, XL388. KU-0063794, GDC-0349, and PKI-587. In embodiments, an active site TOR inhibitor is an asTORi. In some embodiments, “active site inhibitor” may refer to “active site mTOR inhibitor.”

[00211] The term “FKBP” refers to the protein Peptidyl-prolyl cis-trans isomerase. For non-limiting examples of FKBP, see Cell Mol Life Sci. 2013 Sep;70(18):3243-75. In embodiments, “FKBP” may refer to “FKBP-12” or “FKBP 12” or “FKBP 1 A.” In embodiments, “FKBP” may refer to the human protein. Included in the term “FKBP” is the wildtype and mutant forms of the protein. In embodiments, “FKBP” may refer to the wildtype human protein. In embodiments, “FKBP” may refer to the wildtype human nucleic acid. In embodiments, the FKBP is a mutant FKBP In embodiments, the mutant FKBP is associated with a disease that is not associated with wildtype FKBP. In embodiments, the FKBP includes at least one amino acid mutation (e.g , 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 mutations, or any range derivable therein) compared to wildtype FKBP.

[00212] The term “FKBP-12” or “FKBP 12” or “FKBP1A” may refer to the protein “Peptidyl-prolyl cis-trans isomerase FKBP 1 A.” In embodiments, “FKBP-12” or “FKBP 12” or “FKBP 1 A” may refer to the human protein included in the term “FKBP-12” or “FKBP 12” or “FKBP 1 A” are the wildtype and mutant forms of the proleinln embodiments, the reference numbers immediately above may refer to the protein, and associated nucleic acids, known as of the dale of filing of this application. In embodiments, “FKBP-12” or “FKBP 12” or “FKBP 1 A” may refer to the wildtype human protein. In embodiments, “FKBP-12” or “FKBP 12” or “FKBP1A” may refer to the wildtype human nucleic acid. In embodiments, the FKBP-12 is a mutant FKBP-12. In embodiments, the mutant FKBP-12 is associated with a disease that is not associated with wildtype FKBP-12. In embodiments, the FKBP-12 may include at least one amino acid mutation (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 mutations, or any range derivable therein) compared to wildtype FKBP-12. In embodiments, the FKBP-12 has the protein sequence corresponding to reference number 01:206725550.

[00213] The term “4E-BRG’ or “4EBP1” or “EIF4EBP1” refers to the protein “Eukaryotic translation initiation factor 4E-binding protein 1.” In embodiments, “4E-BP1” or “4EBP1” or ΈIR4EBR G’ may refer to the human protein. Included in the term “4E-BP 1” or “4EBP 1” or ΈIE4EBRG’ are the wildtype and mutant forms of the protein. In embodiments, the reference numbers immediately above may refer to the protein, and associated nucleic acids, known as of the date of filing of this application. In embodiments, “4E-BP 1” or “4EBP1” or “EIF4EBP1” may refer to the wildtype human protein. In embodiments, “4E-BP1” or “4EBP1” or “EIF4EBP1” may refer to the wildtype human nucleic acid. In embodiments, the 4EBP1 is a mutant 4EBP1. In embodiments, the mutant 4EBP1 is associated with a disease that is not associated with wildtype 4EBP1 In embodiments, the 4EBP1 may include at least one amino acid mutation (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 mutations, or any range derivable therein) compared to wildtype 4EBP1. In embodiments, the 4EBP1 has the protein sequence corresponding to reference number GL4758258.

[00214] The term “Akt” refers to the serine/threonine specific protein kinase involved in cellular processes such as glucose metabolism, apoptosis, proliferation, and other functions, also known as “protein kinase B” (PKB) or “Aktl.” In embodiments, “Akt” or “AM” or “PKB” may refer to the human protein. Included in the term “Akt” or “Aktl” or “PKB” are the wildtype and mutant forms of the protein. In embodiments, the reference numbers immediately above may refer to the protein, and associated nucleic acids, known as of the date of filing of this application. In embodiments, “Akt” or “Aktl” or “PKB” may refer to the wildtype human protein. In embodiments, “Akt” or “Aktl” or “PKB” may refer to the wildtype human nucleic acid. In embodiments, the Akt is a mutant Akt. In embodiments, the mutant Akt is associated with a disease that is not associated with wildtype Akt. In embodiments, the Akt may include at least one amino acid mutation (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 mutations, or any range derivable therein) compared to wildtype Akt. In embodiments, the Akt has the protein sequence corresponding to reference number GI: 62241011.

Methods of Modulating mTOR [00215] In some embodiments, compounds disclosed herein are more selective inhibitors of niTORC 1 versus mTGRC2. In some embodiments, compounds disclosed herein are more selective inhibitors of mTORC2 versus xnTORC 1 . In some embodiments, compounds disclosed herein exhibit no selectivity difference between mTORC 1 and niTORC2.

[00216] In another aspect is provided a method of modulating mTORC 1 activity in a subject in need thereof, including administering to the subject an effective amount of a compound as described herein, or a pharmaceutically acceptable salt thereof. In embodiments, the method includes inhibiting mTORCl activity. In embodiments, the method includes inhibiting mTORC 1 activity and not inhibiting mTGRC2 activity.

[00217] In embodiments, the method includes inhibiting mTORC 1 activity more than inhibiting mTORC2 activity. In embodiments, the method includes inhibiting mTORC 1 activity at least 1.1 fold as much as inhibiting mTORC2 activity (e.g., at least 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200,

300 400, 500 600, 700 800, 900 1000, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000, 10000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000 or 1000000 fold).

[00218] In another aspect is provided a method of modulating mTORC2 activity in a subject in need thereof, including administering to the subject an effective amount of a compound as described herein, or a pharmaceutically acceptable salt thereof. In embodiments, the method includes inhibiting mTORC2 activity in embodiments, the method includes inhibiting mTORC2 activity and not inhibiting mTORC 1 activity.

[00219] In embodiments, the method includes inhibiting mTORC2 activity more than inhibiting mTORC 1 activity. In embodiments, the method includes inhibiting mTORC2 activity at least 1.1 fold as much as inhibiting mTORC 1 activity (e.g., at least 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, 3, 4 5, 6, 7, 8 9, 10, 20 30, 40, 50, 6 ( 3, 70, 80, 9 ( 3, 100 20 ( 3,

300, 400, 500, 600, 700, 800, 900, 1000, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000, 10000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, or 1000000 fold).

[00220] In some embodiments, the mTOR is in a cell. In some embodiments, the cell is a mammalian cell, such as a human cell. The cell may be isolated in vitro, form part of a tissue in vitro, or may form part of an organism.

Exemplary Embodiments o ct [00221] Some embodiments of this disclosure are Embodiment I, as follows: [00222] Embodiment I-1. A compound of Formula I: or a pharmaceutically acceptable salt or tautomer thereof wherein:

R 32 is -H, =O or -OR 3 ;

R 28 is H or -C(=Z 1 )-R 28a ;

R 40 is -H or -C(=Z 1 )-R 40a ; wherein at least one of R 28 and R 40 is not H;

Z 1 is O or S;

R iSa an(j j ^ 4 o a are independently -A l -L l -A 2 -B; -A l -A 2 -B; . L 2 -A l -L l - A 2 -IAB ; -O-(Ci-C 6 )alkyl; or -0-(C 6 -Cio)aryl; wherein the aryl is unsubstituted or substituted with 1-5 substituents selected from -NO 2 and halogen;

A 1 and A 2 are independently absent or are independently selected from

1 o

wherein the bond on the left side of A 1 , as drawn, is bound to -C(=Z 1 )- or L 2 ; and wherein the bond on the light side of the A 2 moiety, as drawn, is bound to B or L 3 ; each Q is independently 1 to 3 rings selected from arylene, eycloalkylene, heteroarylene, and heierocyclylene; each X is independently absent or 1 to 2 rings selected from arylene, eycloalkylene, heteroarylene, and heteroeyclylene; each X 1 is a heteroarylene or heteroeyclylene ring; each W is independently absent or 1 to 2 rings selected from arylene eycloalkylene, heteroarylene, and heteroeyclylene; each W 1 is a heteroarylene or heteroeyclylene ring; each G is independently absent or a ring selected from arylene, eycloalkylene, heteroarylene, and heteroeyclylene; o each G J and G 2 are independently heteroarylene or heterocyclylene ring;

L 2 and L J are independently absent or are independently selected from

/ L t-N _ ^lM— heteroarylene heterocyclylene— arylene — an( j |SfR .. (C(R :i ) 2 ) r -S(O) 2- arylene-C(O) wherein the bond on the left side of B s as drawn, is bound to A 2 , L 3 , or L 1 ; and wherein the heteroarylene, heterocyclylene, and arylene are optionally substituted with alkyl, hydroxyalkyl, haloalkyl, alkoxy, halogen, or hydroxyl; each R 3 is independently H or (Ci-C 6 )alkyl; each R 4 is independently H, (Cj-C 6 ialkyl, halogen, 5-12 memhered heteroaryl, 5-12 membered heterocyclyl, (C 6 -Cioiaryl, wherein the heteroaryl, heterocyclyl, and aryl are optionally substituted with -N(R ) 2 , -OR 3 , halogen, (C 1 -C 6 jalkyi, -(C 1 -QOalkylene- heteroaryl -(C 1 -C 6 ialkylene-CN, -C(O)NR 3 -heteroaryI; or -C(O)NR 3 -heterocyelyl; each R 3 is independently H, (Cj-C & )alkyl, -C(O)0R 3 , or -N(R 3 ) 2 , wherein the alkyl is optionally substituted with -OR 3 ; each R 6 is independently H, (C 1 -C 6 )alkyl, -C(O)0R 3 , or -NiR 3 ) ? ., wherein the alkyl is optionally substituted with -N or -OR 3 ; each R 7 is independently H, (C 1 -C 6 jalkyL -C(O)GR 3 , or -N(R 3 ) 2 , wherein the alkyl is optionally substituted with -NiR 3 ) ? or -OR 3 ; each R s is independently H, (C 1 -Ccdalkyl, -C(O)0R 3 , or -N(R 3 ) 2 , wherein the alkyl is optionally substituted with -N(R 3 ) 2 or -OR 3 ; each Y is independently C(R 3 ) 2 or a bond; each n is independently a number from one to 12; each o is independently a number from zero to 30; each p is independently a number from zero to 12; each q Is independently a number from zero to 30; and each r is independently a number from one to 6.

Embodiment 1-2. A compound of Formula II:

or a pharmaceutically acceptable salt or tautomer thereof wherein:

R 32 is -H, =O or -OR 3 ;

R 28 is -H or -C(=Z 1 )-R 28a ;

R 40 is H or -C(=Z 1 )-R 40a ; wherein at least one of R 28 and R 40 is not H;

Z 1 is O or S;

R 2Sa and R 40a are independently --A 1 -L 1 -A 2 -B; A 1 -A -B; -O-(C 1 -C 6 )alkyl; or -O-(C 6 - C 1 o)aryl; wherein the aryl is unsubstituted or substituted with 1-5 substituents selected from -NO 2 and halogen;

A 1 and A 2 are independently absent or are independently selected from

wherein the bond on the left side of A 1 , as drawn, is bound to -Ci-Z 1 )-; and wherein the bond on the right side of the A 2 moiety, as drawn, is bound to B; each Q is independently 1 to 3 rings selected from arylene, cycloalkylene, heteroarylene, and heterocyclyiene; each X is independently absent or 1 to 2 rings selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; each X 1 is a heteroarylene or heterocyclylene ring; each W is independently absent or 1 to 2 rings selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; each W 1 is a heteroarylene or heterocyclyiene ring; each G is independently absent or a ring selected from arylene, cycloalkylene, heteroarylene, and heterocyclylene; each G 1 and G 2 are independently heteroarylene or heterocyclyiene ring; ¾ NR 3 -(C(R J ) 2 ) n -heteroarylene-heterocyclylene-(C 6 -C 1 o)arylene-, heterocyclylene— arylene — anf j

^ NR 3 -(C(R J ) 2 ) n -S(O) 2 arylene-C(O) wherein the bond on the left side of Bh as drawn, is bound to A 2 or L 1 : and wherein the heteroarylene, heterocyclylene, and arylene are optionally substituted with alkyl, hydroxyalkyl, haloalkyl, alkoxy, halogen, or hydroxyl; each R 4 is independently H or (C 1 -C 6 lalkyl; each R 4 is independently H, (C 1 -Ccdalkyl, halogen, 5-12 membered heteroaryl, 5-12 membered heterocyclyl, (C 6 -Cio)aryl, wherein the heteroaryl, heterocyclyl, and aryl are optionally substituted with N{R -OR 4 , halogen, (C 1 -C 6 lalkyl, - (Ci- Chalky lene- heteroaryl, -(C 1 -C 6 lalkylene-CN, -C(O)NR 3 -heteroaryl; or -C(O)NR 3 -heterocyclyl; each R 5 is independently H, (C 1 -C 6 lalkyl, -C(O)OR 4 , or -N(R 4 ) 2 , wherein the alkyl is optionally substituted with -NCR 3 ) ? or -OR 3 ; each R 6 is independently H, (C 1 -C 6 )aikyl, -C(O)OR 3 , or -N(R 3 ) 2 , wherein the alkyl is optionally substituted with N(R S: or -OR 4 ; each R' is independently H, (C 1 -C 6 lalkyl, -C(O)OR 4 , or -N(R 4 ) 2 , wherein the alkyl is optionally substituted with -N(R 3 ) 2 or -OR 3 ; each R 8 is independently H, (C 1 -C 6 )alkyl, -C(O)OR 4 , or -N 2 , wherein the alkyl is optionally substituted with -N(R 3 ) 2 or -OR 3 ; each Y is independently C 2 or a bond; each n is independently a number from one to 12; each o is independently a number from zero to 30; each p is independently a number from zero to 12; each c] is independently a number from zero to 30; and each r is independently a number from one to 6. [00224] Embodiment 1-3. The compound of Embodiment I-1 or 1-2, wherein R Z is =O.

[00225] Embodiment 1-4. The compound of Embodiment I-1 or 1-2, wherein R 3z is -OR 3 .

[00226] Embodiment 1-5. The compound of any one of Embodiments I-1 to 1-4, wherein the compounds are represented by the structure of Formula 1-40: or a pharmaceutically acceptable salt or tautomer thereof.

[00227] Embodiment 1-6. The compound of Embodiment 1-5, wherein Z ! is O.

[00228] Embodiment 1-7. The compound of Embodiment 1-5, wherein Z 1 is S.

[00229] Embodiment 1-8. The compound of any one of Embodiments 1-5 to 1-7, wherein

R 40a is -A f -L f -A 2 -B, wherein A 1 and A 2 are absent.

[00230] Embodiment 1-9. The compound of any one of Embodiments 1-5 to 1-7, wherein

R 40S j s _AELEA 2 -B, wherein A 2 is absent.

[00231] Embodiment I-10. The compound of any one of Embodiments 1-5 to 1-7, wherein R 4 ° a j s - AELEA 2 -B, wherein A 1 is absent.

[00232] Embodiment I-11. The compound of any one of Embodiments 1-5 to 1-7, wherein

R 40a is -AELEA 2 -B.

[00233] Embodiment I-12. The compound of any one of Embodiments 1-5 to 1-7, wherein R 40a is -AEA 2 -B.

[00234] Embodiment I-13. The compound of any one of Embodiments 1-5 to 1-7, wherein , wherein L 2 and A J are absent.

[00235] Embodiment I-14. The compound of any one of Embodiments 1-5 to 1-7, wherein R 40a is — L 2 -A 1 -L 1 -A 2 -L -B, wherein L 2 is absent. [00236] Embodiment I-15. The compound of any one of Embodiments 1-5 to 1-7, wherein R 40S j s — L 2 -A 1 -L 1 -A 2 -L > -B, wherein L 3 is absent.

[00237] Embodiment I-16. The compound of any one of Embodiments 1-5 to 1-7, wherein 4° a j s -O-(Ci-C 6 )alkyl or -O-iC 6 -C 1 oiaryl; wherein the aryl is unsubstituted or substituted with 1-5 substituents selected from -NO 2 and halogen.

[00238] Embodiment I-17. The compound of an one of Embodiments I-1 to 1-4, wherein the compounds are represented by the structure of Formula 1-28: or a pharmaceutically acceptable salt or tautomer thereof.

[00239] Embodiment I-18. The compound of Embodiment I-17, wherein Z 3 is O.

[00240] Embodiment I-19. The compound of Embodiment I-17, wherein Z 1 is S.

[00241] Embodiment 1-20. The compound of any one of Embodiments I-17 to I-19, wherein R 28a is -A 3 -L 3 -A 2 -B, wherein A 1 and A 2 are absent.

[00242] Embodiment 1-21. The compound of any one of Embodiments I-17 to I-19, wherein R 28a is -A 3 -L 3 -A 2 -B, wherein A 2 is absent.

[00243] Embodiment 1-22. The compound of any one of Embodiments I-17 to I-19, wherein R 2Sa is -A 1 -L 1 -A 2 -B, wherein A 1 is absent.

[00244] Embodiment 1-23. The compound of any one of Embodiments I-17 to I-19, wherein R 2Sa is -A i -L i -A 2 -B.

[00245] Embodiment 1-24. The compound of any one of Embodiments I-17 to I-19, wherein R 28a is -A 3 -A 2 -B. [00246] Embodiment 1-25. The compound of any one of Embodiments I-17 to I-19, wherein R 28a is — L--A 1 -L 1 -A 2 -L 3 -B, wherein L 2 and A 1 are absent.

[00247] Embodiment 1-26. The compound of any one of Embodiments I-17 to I-19, wherein R 2Sa is . L - A i -L i -A 2 --L --B, wherein L 2 is absent.

[00248] Embodiment 1-27. The compound of any one of Embodiments I-17 to I-19, wherein R 28a is -- L 2 -A 1 -L 1 -A 2 -L i -B, wherein L 3 is absent.

[00249] Embodiment 1-28. The compound of any one of Embodiments I-17 to I-19, wherein R 28a is -O-(Ci-C 6 )alkyl or -O-(C 6 -Cio)aryl; wherein the aryl is unsubstituted or substituted with 1-5 substituents selected from -NC and halogen.

[00250] Embodiment 1-29. The compound of any one of Embodiments I-1 to I-11, I-13 to I-15, I-17 to 1-23, and 1-25 to 1-27, wherein

[00251] Embodiment 1-30. The compound of any one of Embodiments I-1 to I-11, I-13 to I-15, I-17 to 1-23, and 1-25 to 1-27, wherein L

[00252] Embodiment 1-31. The compound of any one of Embodiments I-1 to I-11, I-13 to I-15, I-17 to Ϊ-23, and 1-25 to 1-27, wherein L 1 is

[00253] Embodiment 1-32. The compound of any one of Embodiments I-1 to I-11, I-13 to I-15, I-17 to 1-23, and 1-25 to 1-27, wherein

[00254] Embodiment 1-33. The compound of any one of Embodiments I-1 to I-11, I-13 to I-15, I-17 to 1-23, and 1-25 to 1-27, wherein [00255] Embodiment 1-34. The compound of any one of Embodiments I-1 to 1-7, I-15, I-17 to I-19, and 1-27, wherein L [00256] Embodiment 1-35. The compound of any one of Embodiments I-1 to 1-7, I-13 to I- [00262] Embodiment 1-41. The compound of any one of Embodiments I-1 to 1-7, T9, 1-11 to I-12, I-14 to I-15, I-17 to I-19, 1-21, 1-23 to 1-24, 1-26 to 1-27, and 1-29 to 1-35, wherein A 1 , , ,

5UB5TITUTE SHEET (RULE 26) 8] Embodiment 1-47. The compound of any one of Embodiments I-1 to 1-7, T IC) to I-

15, I-17 to I-19, 1-22 to 1-27 and 1-29 to 1-44, wherein

Embodiment 1-48. The compound of any one of Embodiments I-1 to 1-7, 1-10 to T

15, 1-17 to I-19, 1-22 to 1-27 and 1-29 to 1-44, wherein

Embodiment 1-49. The compound of any one of Embodiments I-1 to 1-7, 1-10 to I-

15, I-17 to I-19, 1-22 to 1-27 and 1-29 to 1-44, wherein

Embodiment 1-50. The compound of any one of Embodiments T1 to 1-7, 1-10 to I-

15, I-17 to I-19, 1-22 to 1-27 and 1-29 to 1-44, wherein A 2 is.

Embodiment 1-51. The compound of any one of Embodiments T1 to 1-7, 1-10 to I-

15, 1-17 to I-19, 1-22 to 1-27 and 1-29 to 1-44, wherein

[00273] Embodiment 1-52. The compound of any one of Embodiments I-1 to 1-7, 1-10 to I-

15, I-17 to I-19, 1-22 to 1-27 and 1-29 to 1-44, wherein A 2 is. [00274] Embodiment 1-53. The compound of any one of Embodiments I-1 to 1-7, I-10 to I-

15, I-17 to I-19, 1-22 to 1-27 and 1-29 to 1-44,

[00275] Embodiment 1-54. The compound of any one of Embodiments I-1 to 1-53, wherein

[00279] Embodiment 1-58. The compound of any one of Embodiments I-1 to 1-57, wherein R 4 is 5-12 membered heteroaryl, optionally substituted with -N(R J ) 2 , -OR 3 , halogen, (Ci- C b ialkyl, -(C 1 -C 6 )alkylene-heteroaryl, -(Ci-C 6 ialkylene-CN, or -C(O)NR 3 -heteroaryl.

[00280] Embodiment 1-59. The compound of any one of Embodiments I-1 to 1-58, or a pharmaceutically acceptable salt or tautomer thereof, wherein compound has the following formula: Embodiment 1-60. A compound selected from the group consisting of: 1 or a pharmaceutically acceptable salt or isomer thereof.

[00282] Embodiment 1-61. A pharmaceutical composition comprising a compound of any one of Embodiments I-1 to 1-60, or a pharmaceutically acceptable salt thereof, and at least one of a pharmaceutical ly acceptable carrier, diluent, or excipient.

[00283] Embodiment 1-62. A method of treating a disease or disorder mediated by mTOR comprising administering to the subject suffering from or susceptible to developing a disease or disorder mediated by mTOR a therapeutically effective amount of one or more compounds of any one of Embodiments I-1 to 1-60, or a pharmaceutically acceptable salt thereof.

[00284] Embodiment 1-63. A method of preventing a disease or disorder mediated by mTOR comprising administering to the subject suffering from or susceptible to developing a disease or disorder mediated by mTOR a therapeutically effective amount of one or more compounds of any one of Embodiments I-1 to 1-60, or a pharmaceutically acceptable salt thereof

[00285] Embodiment 1-64. A method of reducing the risk of a disease or disorder mediated by m'TOR comprising administering to the subject suffering from or susceptible to developing a disease or disorder mediated by niTOR a therapeutically effective amount of one or more compounds of any one of Embodiments I-1 to 1-60, or a pharmaceutically acceptable salt thereof.

[00286] Embodiment 1-65. The method of any one of Embodiments 1-62 to 1-64, wherein the disease is cancer or an immune- mediated disease.

[00287] Embodiment 1-66. The method of Embodiment 1-65, wherein the cancer is selected from brain and neurovascular tumors, head and neck cancers, breast cancer, lung cancer, mesothelioma, lymphoid cancer, stomach cancer, kidney cancer, renal carcinoma, liver cancer, ovarian cancer, ovary endometriosis, testicular cancer, gastrointestinal cancer, prostate cancer, glioblastoma, skin cancer, melanoma, neuro cancers, spleen cancers, pancreatic cancers, blood proliferative disorders, lymphoma, leukemia, endometrial cancer, cervical cancer, vulva cancer, prostate cancer, penile cancer, bone cancers, muscle cancers, soft tissue cancers, intestinal or rectal cancer, anal cancer, bladder cancer, bile duct cancer, ocular cancer, gastrointestinal stromal tumors, and neuro-endocrine tumors.

[00288] Embodiment 1-67. The method of Embodiment 1-65, wherein the immune mediated disease is selected from resi tance by transplantation of heart, kidney, liver, medulla ossium, skin, cornea, lung, pancreas, intestinum tenue, limb, muscle, nerves, duodenum, small-bowel, or pancreatic-islet-cell; graft-versus-host diseases brought about by medulla ossium transplantation; rheumatoid arthritis, systemic lupus erythematosus, Hashimoto's thyroiditis, multiple sclerosis, myasthenia gravis, type I diabetes, uveitis, allergic encephalomyelitis, and glomerulonephriti .

[00289] Embodiment 1-68. A method of treating cancer comprising administering to the subject a therapeutically effective amount of one or more compounds of any one of Embodiments I-1 to 1-60, or a pharmaceutically acceptable salt thereof.

[00290] Embodiment 1-69. The method of Embodiment 1-68, wherein the cancer is selected from brain and neurovascular tumors, head and neck cancers, breast cancer, lung cancer, mesothelioma, lymphoid cancer, stomach cancer, kidney cancer, renal carcinoma, liver cancer, ovarian cancer, ovary endometriosis, testicular cancer, gastrointestinal cancer, prostate cancer, glioblastoma, skin cancer, melanoma, neuro cancers, spleen cancers, pancreatic cancers, blood proliferative disorders, lymphoma, leukemia, endometrial cancer, cervical cancer, vulva cancer, prostate cancer, penile cancer, bone cancers, muscle cancers, soft tissue cancers intestinal or rectal cancer, anal cancer, bladder cancer, bile duct cancer, ocular cancer, gastrointestinal stromal tumors, and neuro-endocrine tumors.

[00291] Embodiment 1-70. A method of treating an immune -mediated disease comprising administering to the subject a therapeutically effective amount of one or more compounds of any one of Embodiments I-1 to 1-60, or a pharmaceutically acceptable salt thereof.

[00292] Embodiment 1-71 The method of Embodiment 1-70, wherein the immune- mediated disease is selected from resi tance by transplantation of heart kidney, liver, medulla ossium, skin, cornea, lung pancreas, intestinum tenue, limb, muscle nerves, duodenum, small-bowel, or pancreatic-islet-cell; graft-versus-host diseases brought about by medulla ossium transplantation; rheumatoid arthritis, systemic lupus erythematosus, Hashimoto's thyroiditis multiple sclerosis, myasthenia gravis, type I diabetes, uveitis, allergic encephalomyeliti s , and glomerulonephriti s .

[00293] Embodiment 1-72. A method of treating an age related condition comprising administering to the subject a therapeutically effective amount of one or more compounds of any one of Embodiments I-1 to 1-60, or a pharmaceutically acceptable salt thereof.

[00294] Embodiment 1-73 The method of Embodiment 1-72, wherein the age related condition is selected from sarcopenia, skin atrophy muscle wasting, brain atrophy, atherosclerosis, arteriosclerosis, pulmonary emphysema, osteoporosis, osteoarthritis, high blood pressure, erectile dysfunction, dementia, Huntington’s disease, Alzheimer's disease, cataracts, age-related macular degeneration, prostate cancer, stroke, diminished life expectancy, impaired kidney function and age-related hearing loss, aging-related mobility disability (e.g., frailty), cognitive decline, age-related dementia, memory impairment, tendon stiffness, heart dysfunction such as cardiac hypertrophy and systolic and diastolic dysfunction, immunosenescence, cancer, obesity and diabetes.

[00295] Embodiment 1-74. A compound of any one of Embodiments I-1 to 1-60, or a pharmaceutically acceptable salt thereof, for use in treating, preventing, or reducing the risk of a disease or condition mediated by mTOR. [00296] Embodiment 1-75. Use of a compound of any of Embodiments I-1 to 1-60, or a pharmaceut cally acceptable salt thereof, in the manufacture of a medicament for treating, preventing, or reducing the risk of a disease or disorder mediated b mTOR.

[00297] Embodiment 1-76. A compound of any one of Embodiments I-1 to 1-60, or a pharmaceutically acceptable salt thereof, for use in treating cancer.

[00298] Embodiment 1-77. U se of a compound of any one of Embodiments 1-1 to Ϊ-60, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating cancer.

[00299] Embodiment 1-78. A compound of any one of Embodiments I-1 to 1-60, or a pharmaceutically acceptable salt thereof, for use in treating an immune-mediated disease.

[00300] Embodiment 1-79. Use of a compound of any one of Embodiments I-1 to 1-60, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating an immune-mediated disease .

[00301] Embodiment 1-80. A compound of any one of Embodiments I-1 to 1-60, or a pharmaceutically acceptable salt thereof, for use in treating an age related condition.

[00302] Embodiment 1-81. Use of a compound of any one of Embodiments I-1 to 1-60, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating an age related condition.

[00303] Some embodiments of this disclosure are Embodiment II, as follows:

[00304] Embodiment II-1. A compound of Formula Ic: or a pharmaceutically acceptable salt or tautomer thereof, wherein:

R 28 is -H, (C -C 6 )alkyl, or -C(=Z 1 )-R 28a ;

R 40 is -H or -C(=Z 1 )-R 40a ; wherein when R 28 and R 40 are H then R 32 is not =O; each Z 1 is independently O or S;

R 28a , R 32a , and R 40a are independently -A 1 -L 1 -A 2 -B; -A 1 -A 2 -B; -L 2 - A 1 -L 1 -A 2 -L 3 -B ; -O-iC 1 -C t Oalkyl; or -O-(C 6 -Cjo)aryl; wherein the aryl of -O-(C 6 -Cio)aryl is unsubstituted or substituted with 1-5 substituents selected from -NO 2 and halogen;

A 1 and A 2 are independently absent or are independently selected from wherein the bond on the left side of A 1 , as drawn, is bound to -C(=Z 1 )- or L 2 ; and wherein the bond on the right side of the A 2 moiety, as drawn is bound to B or L 3 ; each Q is independently 1 to 3 rings selected from arylene, eycloalkylene, heteroarylene, and heierocyclylene; each X is independently absent or 1 to 2 rings selected from arylene, eycloalkylene, heteroarylene, and heterocyclylene; each X 1 is independently a heteroarylene or heterocyclylene ring; each W is independently absent or 1 to 2 rings selected from arylene eycloalkylene, heteroarylene, and heterocyclylene; each W 1 is independently a heteroarylene or heterocyclylene ring; each G is independently absent or a ring selected from arylene, eycloalkylene, heteroarylene, and heterocyclylene; each G 1 and G 2 are independently heteroarylene or heterocyclylene ring; each L 1 is independently selected from

L 2 and L 3 are independently absent or are independently selected from

and -l NR 3 -(C(R 3 ) 2 ) « -

S(O) 2- arylene-(C(R 3 ) 2 ) H wherein the bond on the left side of B 1 , as drawn, is bound to A 2 , L 3 , or L 1 ; and wherein the heleroarylene, heterocyclylene, and arylene are each independently optionally substituted with alkyl, hydroxyalkyl, haloalkyl, alkoxy, halogen, or hydroxyl; each R 3 is independently H or (Ci-C 6 )alkyl; each R 4 is independently H, (C 1 -C 6 jalkyl, halogen, 5-12 membered heteroaryl, 5-12 membered heteroeyelyl, (C 6 -C t o)aryl, wherein the heteroaryl, heterocyelyl, and aryl are optionally substituted with -N(R 3 ) 2 , -OR 3 , halogen, (C 1 -C 6 jalkyL -(Ci-Ct alkylene- heteroaryl, -(C 1 -C oj alkylene-CN, -C(O)NR -heteroaryl, or -C(O)NR -he†eroeyclyI; each R 5 is independently H, (C 1 -C 6 )alkyl, -C(O)0R 3 , or -N(R ) 2 , wherein the alkyl of (C 1 -C jalkyl is optionally substituted with -N(R 3 ) 2 or -OR 3 ; each R 6 is independently H, (C 1 -C 6 lalkyl, -C(O)OR 3 , or --N(R ) 2 , wherein the alkyl of (C 1 -C 6 lalkyl is optionally substituted with N ( R ) 2 or -OR 3 ; each R / is independently H, (C t -C 6 )alkyl, -C(O)0R 3 , or -N(R 3 ) 2, wherein the alkyl of (C 1 -C t Oalkyl is optionally substituted with -N(R 3 ) 2 or -OR 3 ; each R 8 is independently H, (C -C 6 lalkyl, -C(O)OR 3 , or -N(R 3 ) 2 , wherein the alkyl of (C 1 -C 6 lalkyl is optionally substituted with -N(R ) 2 or -OR 3 ; each Y is independently C(R ) 2 or a bond; each n is independently an integer from one to 12; each o is independently an integer from zero to 30; each p is independently an integer from zero to 12; each q is independently an integer from zero to 30; and each r is independently an Integer from one to 6.

[00305] Embodiment II-1A. A compound of Formula la: or a pharmaceutically acceptable salt or tautomer thereof wherein:

R 32 is -H, =O, -OR 3 , -N3, or -O-C(=Z 1 )-R 32a ;

R 28 is H, (Ci-C 6 ) lkyl, or -C(=Z l )-R 28a ;

R 40 is -H or -C(=Z 1 )-R 40a ; wherein when R 28 and R 40 are H, then R 32 is not =O; each Z 1 is independently O or S;

R 2S ‘\ R 32a , and R 40 * are independently -A’-LAAB; -AAAB; -LAAAAAAAB; -O-(C 1 -C 6 )alkyl; or -0-(C 6 -Cio)aryl; wherein the aryl of -O-(C 6 -Cio)aryl is unsubstituted or substituted with 1-5 substituents selected from -NO 2 and halogen;

A 1 and A 2 are independently absent or are independently selected from

wherein the bond on the left side of A 1 , as drawn, is bound t wherein the bond on the right side of the A 2 moiety, as drawn, is bound to B or L 3 ; each Q is independently 1 to 3 rings selected from arylene, cycloalkylene, heteroarylene, and heteroeyclylene; each X is independently absent or 1 to 2 rings selected from arylene, cycloalkylene, heteroarylene, and heteroeyclylene; each X 1 is independently a heteroarylene or heteroeyclylene ring; each W is independently absent or 1 to 2 rings selected from arylene, cycloalkylene, heteroarylene, and heteroeyclylene; each W l is independently a heteroarylene or heteroeyclylene ring; each G is independently absent or a ring selected from arylene, cycloalkylene, heteroarylene, and heteroeyclylene; each G 1 and G 2 are independently heteroarylene or heteroeyclylene ring; each L 1 is independently selected from L 2 and L 3 are independently absent or are independently selected from each B is independently selected from _ heterocyciylene-— arylene — anj j

- | NR 3 -(C(R ;> ) 2 ) n -S(O) 2 -arylene-C(O)-, wherein the bond on the left side of B 1 , as drawn is bound to A 2 , L 3 , or L 1 ; and wherein the heteroarylene, heterocyclylene, and arylene are optionally substituted with alkyl, hydroxy alkyl, haloalkyl, alkoxy, halogen, or hydroxyl; each R is independently H or (C 1 -C 6 jalkyl; each R 4 is independently H, (C 1 -C 6 )alkyl, halogen, 5-12 membered heteroaryl, 5-12 membered heterocyclyl, (C 6 -Cioiaryl, wherein the heteroaryl, heterocyclyl, and aryl are optionally substituted with -N(R ) 2 , -OR 4 , halogen, (Ci-C 6 )alkyl, -(C -C 6 lalkylene- heteroaryl, -(C 1 -C 6 )alkylene-CN, -C(O)NR 3 -heteroaryl, or -C(O)NR 3 -heterocyclyl; each R 5 is independently H, (C 1 -Ccdalkyl, -C(G)0R 3 , or -N(R 3 ) 2 , wherein the alkyl of (C 1 -C 6 )alkyl is optionally substituted with -N 2 or -OR 3 ; each R 6 is independently H, (C 1 -C 6 jalkyL -C(O)GR 3 , or -N(R 3 ) 2 , wherein the alkyl of (C 1 -C jalkyl is optionally substituted with -N(R 3 ) 2 or -OR 3 ; each R·' is independently H, (C 1 -C 6 iaikyl, -C(O)0R 3 , or -N(R 5 ) 2 , wherein the alkyl of (C 1 -C 6 lalkyl is optionally substituted with --N or -OR ’; each R 8 is independently H, (C 1 -C 6 )alkyl, -C(O)0R 3 , or -N(R ) 2 , wherein the alkyl of (C 1 -C t Oalkyl is optionally substituted with -N(R 3 ) 2 or -OR 3 ; each Y is independently C(R 3 ) 2 or a bond; each n is independently an integer from one to 12; each o is independently an integer from zero to 30; each p is independently an integer from zero to 12; each q is independently an integer from zero to 30; and each r is independently an integer from one to 6.

[00306] Embodiment II-2. The compound of Embodiment II-1, wherein R 32 is =O.

[00307] Embodiment II-3. The compound of Embodiment II-1, wherein R 32 is -OR 3 .

[00308] Embodiment ii-4. The compound of any one of Embodiments II-1 to II-3, or a pharmaceutically acceptable salt or tautomer thereof, wherein the compound is represented by the structure of Formula (I -40b):

5UB5TITUTE SHEET (RULE 26)

[00309] Embodiment II-5. The compound of Embodiment II-4, wherein Z 1 is O.

[00310] Embodiment II- 6. The compound of Embodiment II -4, wherein Z s is S.

[00311] Embodiment II-7. The compound of any one of Embodiments P-4 to II- 6, wherein R 4ja Is -A 3 -L 3 -A 2 -B, wherein A 1 and A 2 are absent

[00312] Embodiment II-8. The compound of any one of Embodiments II-4 to II-6, wherein R 40a is -A 3 -L 3 -A 2 -B, wherein A 2 is absent.

[00313] Embodiment II-9 The compound of any one of Embodiments P-4 to II- 6, wherein R 40a is -A 3 -L 3 -A 2 -B, wherein A 3 is absent.

[00314] Embodiment II-10. The compound of any one of Embodiments II- 4 to II- 6, wherein R 4ja Is -A 1 -L 1 -A 2 -B.

[00315] Embodiment II-11. The compound of any one of Embodiments II-4 to ΪI-6, wherein R 40a is -A 3 -A 2 -B.

[00316] Embodiment II-12. The compound of any one of Embodiments II-4 to II- 6, wherein R 40a is -L 2 -A 3 -L 3 -A 2 -L 3 -B, wherein L 2 and A 3 are absent.

[00317] Embodiment 1I-13. The compound of any one of Embodiments II- 4 to II- 6, wherein R 40a is -L 2 - A 3 -L 1 - A 2 -L 3 -B , wherein L 2 is absent.

[00318] Embodiment 1I-14. The compound of any one of Embodiments II-4 to II-6, wherein R 40a is -L 2 -A 3 -L 1 -A 2 -L 3 -B, wherein V is absent. [00319] Embodiment 1I-15. The compound of any one of Embodiments II-4 to II- 6, wherein R 4ja is -O-(Ci-C 6 )alkyl or -O-(C 6 -Cio)aryl; wherein the aryl of -O-(C 6 -Cio)aryl is unsubstituted or substituted with 1-5 substituents selected from -NO and halogen.

[00320] Embodiment 1I-16. The compound of any one of Embodiments II-1 to II-3, or a pharmaceutically acceptable salt or tautomer thereof, wherein the compounds are represented by the structure of Formula (I-28b):

[00321] Embodiment 1I-17. The compound of Embodiment 1I-16, wherein Z 1 is O.

[00322] Embodiment 1I-18. The compound of Embodiment II-16, wherein Z l is 8.

[00323] Embodiment 1I-19. The compound of any one of Embodiments 1I-16 to II-

18, wherein R 28a is -A 1 -L 1 -A 2 -B, wherein A 1 and A 2 are absent.

[00324] Embodiment 11-20. The compound of any one of Embodiments II-16 to II- 18, wherein R 2Sa is -A*-ΐ A 2 -B, wherein A 2 is absent.

[00325] Embodiment 11-21. The compound of any one of Embodiments 1I-16 to II- 18, wherein R 28a is -A^ILA^B, wherein A 1 is absent.

[00326] Embodiment 11-22. The compound of any one of Embodiments 1I-16 to II- 18, wherein R 8a is -A'-L'-A^B.

[00327] Embodiment 11-23. The compound of any one of Embodiments II-16 to II- 18, wherein R 2Sa is -AAA B

[00328] Embodiment 11-24. The compound of any one of Embodiments 1I-16 to II- 18, wherein R 28a is wherein L 2 and A 1 are absent. [00329] Embodiment 11-25. The compound of any one of Embodiments 1I-16 to II- 18, wherein R z8a is --L 2 -A 1 -L 1 -A 2 -L J -B, wherein L 2 is absent.

[00330] Embodiment 11-26. The compound of any one of Embodiments II-16 to II- 18, wherein R z8a is -L 2 -A 1 -L 1 -A 2 -L 3 -B, wherein L 3 is absent.

[00331] Embodiment 11-27. The compound of any one of Embodiments II-16 to II- 18, wherein R 28a is -O-(C 1 -C 6 )alkyl or -O-(C 6 -C { o)aryl; wherein the aryl of -O-(C 6 -C { o)aryl is unsubstituted or substituted with 1-5 substituents selected from --NO 2 and halogen.

[00332] Embodiment 11-28. The compound of Embodiment II-1, or a pharmaceutically acceptable salt or tautomer thereof, wherein the compound is represented by the structure of Formula (I-32b): wherein R 32 is -O-C(=Z i )-R 32a .

[§§333] Embodiment 11-29. The compound of Embodiment 11-28, wherein Z 1 is O.

[§§334] Embodiment 11-30. The compound of Embodiment 11-28, wherein Z 1 is S.

[§§335] Embodiment II-31. The compound of any one of Embodiments 11-28 to II-

30, wherein R 32a is -A i -L i -A 2 -B, wherein A 1 and A 2 are absent.

[00336] Embodiment 11-32. The compound of any one of Embodiments 11-28 to II- 30, wherein R >2a is -A , -1/-A 2 ·-B, wherein A 2 is absent.

[00337] Embodiment 11-33. The compound of any one of Embodiments 11-28 to II- 30, wherein R 32a is -A 1 -L 1 -A 2 -B, wherein A 1 is absent.

[§§338] Embodiment 11-34. The compound of any one of Embodiments 11-28 to II- 30, wherein R 32a is -A i -L i -A 2 -B. [00339] Embodiment 11-35. The compound of any one of Embodiments 11-28 to II- 30, wherein R j2a is -Ad-A 2 -!!.

[00340] Embodiment 11-36. The compound of any one of Embodiments 11-28 to II- 30, wherein R >2a is -L 2 -A 1 -L 1 -A 2 -L 3 -B, wherein L 2 and A 1 are absent.

[00341] Embodiment 11-37. The compound of any one of Embodiments 11-28 to II- 30, wherein R 32a is — Z -A 1 -L 1 -A 2 -L 3 -B, wherein L 2 is absent.

[00342] Embodiment 11-38. The compound of any one of Embodiments 11-28 to II- 30, wherein R 32a is --L 2 -A I -L I -A 2 -L J -B, wherein L J is absent.

[00343] Embodiment 11-39. The compound of any one of Embodiments 11-28 to II- 30, wherein R >2a is -O-(C 1 -C 6 )aIkyl or -O-(C 6 -C 1 o)aryl; wherein the aryl of --O-fC 6 -Ciojaryl is unsubstituted or substituted with 1-5 substituents selected from -NO 2 and halogen.

[00344] Embodiment 11-40. The compound of any one of Embodiments II-1 to II- , , , , Embodiment 11-45. The compound of any one of Embodiments II-1 to II-

10, 1I-12 11-22, 11-24 to 11-35, and 11-36 to 11-39, wherein

[00350] Embodiment 11-46. The compound of any one of Embodiments II-1 to II-6,

1I-12 to 1I-18, 11-24 to 11-30, and 11-36 to 11-45, wherein L

Embodiment 11-47. The compound of any one of Embodiments II-1 to i 1-6.

1I-12 to 1I-18, 11-24 to 11-30, and 11-36 to 11-45, wherein

[00352] Embodiment 11-48. The compound of any one of Embodiments II-1 to II-7, II- 9, 1I-12, 1I-16 to 11-19, 11-21, 11-24, 11-28 to 11-31, 11-33, 11-36, and 11-39 to 11-45. wherein A 1 is absent.

[00353] Embodiment 11-49. The compound of any one of Embodiments II-1 to -6, II- 8, 11-10 to 11-1 1. 11-13 to 11-18, 11-20. 11-22 to 11-23, 11-25 to 11-30. 11-32, 11-34 to 11-35, and

11-37 to 11-45, wherein

[00354] Embodiment 11-50. The compound of any one of Embodiments II-1 to 11-6, II- 8, II-10 to 11-1 L 1I-13 to 11-18. 11-20, 11-22 to 11-23, 11-25 to 11-30, 11-32, 11-34 t o 11-35, and

11-37 to 11-45, wherein

[00355] Embodiment II-51. The compound of any one of Embodiments II-1 to 11-6,

11-8. II-10 to 11-11. 11-13 to 11-18. 11-20, 11-22 to 11-23, 11-25 to 11-30, 11-32, 11-34 to 11-35, and

11-37 to 11-45, wherein [00356] Embodiment 11-52. The compound of any one of Embodiments II-1 to II- 6, II- 8, 1I-10 to 1I-11, 1I-13 to 1I-18, 11-20, 11-22 to 11-23, 11-25 to 11-30, 11-32, 11-34 to 11-35, and

11-37 to 11-45, wherein

[00357] Embodiment 11-53. The compound of any one of Embodiments II-1 to II-6, II- 8, II-10 to II-11 , 1I-13 to 1I-18, 11-20, 11-22 to 11-23, 11-25 to 11-30, 11-32, 11-34 to 11-35, and

11-37 to 11-45, wherein

[00358] Embodiment 11-54. The compound of any one of Embodiments II-1 to II-6, II- 8, 1I-10 to II-11 , 1I-13 to 1I-18, 11-20, 11-22 to 0-23, 11-25 to 11-30, 11-32, ΪI-34 to 11-35, and

11-37 to IΪ-45, wherein

[00359] Embodiment 11-55. The compound of any one of Embodiments II-1 to II- 6,

II- 8, IMQ to 1I-11, P-13 to 1I-18, 11-20, 11-22 to 11-23, 11-25 to 11-30, 11-32, 11-34 to 11-35, and

11-37 to 11-45, wherein A 1 is.

[00360] Embodiment 11-56. The compound of any one of Embodiments II-1 to II-6, II- 8, 1I-10 to II-11 , 1I-13 to 1I-18, 11-20, 11-22 to 11-23, 11-25 to 11-30, 11-32, 11-34 to 11-35, and

11-37 to 11-45, wherein

[00361] Embodiment 11-57. The compound of any one of Embodiments II-1 to IT- 8 , 1I-15 to 11-20, 11-27 to ΪI-32, and 11-39 to 11-45, wherein A 2 is absent. Embodiment 11-58. The compound of any one of Embodiments II-1 to II- 6,

II-9 to 1I-18, 11-21 to 11-30, and 11-33 to 11-45, wherein

Embodiment 11-59. The compound of any one of Embodiments II-1 to II-6,

II- 9 to 1I-18, 11-21 to 11-30, and 11-33 to 11-45, wherein

Embodiment 11-60. The compound of any one of Embodiments II-1 to II- 6,

II-9 to 1I-18, 11-21 to 11-30, and 11-33 to 11-45, wherein

Embodiment 11-61. The compound of any one of Embodiments II-1 to 11-6,

ΪΪ-9 to 1I-18, 11-21 to 11-30, and 11-33 to ΪI-45, wherein

[00366] Embodiment 11-62. The compound of any one of Embodiments II-1 to II-6,

II- 9 to 1I-18, 11-21 to 11-30, and 11-33 to Ii-45, wherein A 2 is.

Embodiment 11-63. The compound of any one of Embodiments II-1 to II-6,

II- 9 to II-18, 11-21 to 11-30, and 11-33 to 11-45, wherein 8] Embodiment 11-64. The compound of any one of Embodiments II-1 to II- 6,

,

II-9 to II-18, 11-21 to 11-30, and 11-33 to 11-45, wherein A‘ is.

Embodiment 11-65. The compound of any one of Embodiments II-1 to II- 6,

II-9 to 11-18, 11-21 to 11-30, and 11-33 to 11-45, wherein [00370] Embodiment 11-66. The compound of any one of Embodiments II-1 to II-

65, wherein

[00371] Embodiment 11-67. The compound of any one of Embodiments II-1 to li-

65, wherein

[00372] Embodiment 11-68. The compound of any one of Embodiments II-1 to II-

65, wherein B 1 is ¾ NR 3 -(C(R 3 ) 2 )n-.

[00373] Embodiment 11-69. The compound of any one of Embodiments II-1 to II-

65, wherein

[00374] Embodiment 11-70. The compound of any one of Embodiments II-1 to II-

69, wherein R 4 is 5-12 membered heteroaryl, optionally substituted with -N(R 3 ) 2 , -OR 3 , halogen, (C 1 -C 6 jalkyl, -(C 1 -C 6 )alkylene-heteroaryl, -(C 1 -C 6 ialkylene-CN, or -C(O)NR 3 - heteroaryl.

[00375] Embodiment 11-71. The compound of any one of Embodiments II-1 to II-

70, or a pharmaceutically acceptable salt or tautomer thereof, wherein compound has the following formula:

Embodiment 11-72. A compound selected from the group consisting of: o or a pharmaceutically acceptable salt or tautomer thereof.

[00377] Embodiment 11-73. A compound selected from the group consisting of:

1 in _

or a pharmaceutically acceptable salt or tautomer thereof.

1 s s:

or a pharmaceutically acceptable salt or tautomer thereof.

[00379] Embodiment 11-75. A pharmaceutical composition comprising a compound of any one of Embodiments II-1 to 11-74, or a pharmaceutically acceptable salt thereof, and at least one of a pharmaceutically acceptable carrier, diluent, or excipient.

[00380] Embodiment 11-76. A method of treating a disease or disorder mediated by mTGR comprising administering to the subject suffering fro or susceptible to developing a disease or disorder mediated by mTOR a therapeutically effective amount of one or more compounds of any one of Embodiments II-1 to 11-74, or a pharmaceutically acceptable salt thereof

[00381] Embodiment 11-77. A method of preventing a disease or disorder mediated by m'TOR comprising administering to the subject suffering from or susceptible to developing a disease or disorder mediated by niTOR a therapeutically effective amount of one or more compounds of any one of Embodiments P-1 to 11-74, or a pharmaceutically acceptable salt thereof.

[00382] Embodiment 11-78. A method of reducing the risk of a disease or disorder mediated by mTOR comprising administering to the subject suffering from or susceptible to developing a disease or disorder mediated by mTOR a therapeutically effective amount of one or more compounds of any one of Embodiments P-1 to 11-74, or a pharmaceutically acceptable salt thereof.

[00383] Embodiment 11-79. The method of any one of Embodiments 11-76 to 11-78, wherein the disease is cancer or an immune-mediated disease.

[00384] Embodiment 11-80. The method of Embodiment 11-79, wherein the cancer is selected from brain and neurovascular tumors, head and neck cancers, breast cancer, lung cancer, mesothelioma, lymphoid cancer, stomach cancer, kidney cancer, renal carcinoma, liver cancer, ovarian cancer, ovary endometriosis, testicular cancer, gastrointestinal cancer, prostate cancer, glioblastoma, skin cancer, melanoma, neuro cancers, spleen cancers, pancreatic cancers, blood proliferative disorders, lymphoma, leukemia, endometrial cancer, cervical cancer, vulva cancer, prostate cancer, penile cancer, bone cancers, muscle cancers, soft tissue cancers, intestinal or rectal cancer, anal cancer, bladder cancer, bile duct cancer, ocular cancer, gastrointestinal stromal tumors, and neuro-endocrine tumors.

[00385] Embodiment 11-81. The method of Embodiment 11-79, wherein the immune- mediated disease is selected from resistance by transplantation of heart, kidney, liver, medulla ossium, skin, cornea, lung, pancreas, intestinum tenue, limb, muscle, nerves, duodenum, small-bowel, or pancreatic-islet-cell; graft-versus-host diseases brought about by medulla ossium transplantation; rheumatoid arthritis, systemic lupus erythematosus, Hashimoto's thyroiditis, multiple sclerosis, myasthenia gravis, type I diabetes, uveitis, allergic encephalomyelitis, and glomerulonephritis. [00386] Embodiment 11-82. A method of treating cancer comprising administering to the subject a therapeutically effective amount of one or more compounds of any one of Embodiments II-1 to 11-74, or a pharmaceutically acceptable salt thereof.

[00387] Embodiment 11-83. The method of Embodiment 11-82, wherein the cancer is selected from brain and neurovascular tumors, head and neck cancers, breast cancer, lung cancer, mesothelioma, lymphoid cancer, stomach cancer, kidney cancer, renal carcinoma, liver cancer, ovarian cancer, ovary endometriosis, testicular cancer, gastrointestinal cancer, prostate cancer, glioblastoma, skin cancer, melanoma, neuro cancers, spleen cancers, pancreatic cancers, blood proliferative disorders, lymphoma, leukemia, endometrial cancer, cervical cancer, vulva cancer, prostate cancer, penile cancer, bone cancers, muscle cancers, soft tissue cancers, intestinal or rectal cancer, anal cancer, bladder cancer, bile duct cancer, ocular cancer, gastrointestinal stromal tumors, and neuro-endocrine tumors.

[00388] Embodiment 11-84. A method of treating an immune-mediated disease comprising administering to the subject a therapeutically effective amount of one or more compounds of any one of Embodiments II-1 to 11-74, or a pharmaceutically acceptable salt thereof.

[00389] Embodiment 11-85. The method of Embodiment 11-84, wherein the immune- mediated disease is selected from resistance by transplantation of heart, kidney, liver, medulla ossium, skin, cornea, lung, pancreas, mtestinum tenue, limb, muscle, nerves, duodenum, small-bowel, or pancreatic-islet-cell; graft- versus-host diseases brought about by medulla ossium transplantation; rheumatoid arthritis, systemic lupus erythematosus, Hashimoto's thyroiditis, multiple sclerosis, myasthenia gravis, type I diabetes, uveitis, allergic encephalomyelitis, and glomerulonephritis.

[00390] Embodiment 11-86. A method of treating an age related condition comprising administering to the subject a therapeutically effective amount of one or more compounds of any one of Embodiments II-1 to 11-74, or a pharmaceutically acceptable salt thereof.

[00391] Embodiment 11-87. The method of Embodiment 11-86, wherein the age related condition is selected from sarcopenia, skin atrophy, muscle wasting, brain atrophy, atherosclerosis, arteriosclerosis, pulmonary emphysema, osteoporosis, osteoarthritis, high blood pressure, erectile dysfunction, dementia, Huntington’s disease, Alzheimer's disease, cataracts, age-related macular degeneration, prostate cancer, stroke, diminished life expectancy, impaired kidney function, and age-related hearing loss, aging-related mobility disability (e.g., frailty), cognitive decline, age-related dementia, memory impairment, tendon stiffness, heart dysfunction such as cardiac hypertrophy and systolic and diastolic dysfunction, immunosenescence, cancer, obesity, and diabetes.

[00392] Embodiment 11-88. A compound of any one of Embodiments II-1 to 11-74, or a pharmaceutically acceptable salt thereof, for use in treating, preventing, or reducing the risk of a disease or condition mediated by mTOR.

[00393] Embodiment 11-89. Use of a compound of any of Embodiments II-1 to II- 74, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating, preventing, or reducing the risk of a disease or disorder mediated by mTOR.

[00394] Embodiment 11-90. A compound of any one of Embodiments 1-74, or a pharmaceutically acceptable salt thereof, for use in treating cancer.

[00395] Embodiment 11-91. Use of a compound of any one of Embodiments II-1 to IΪ-74, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating cancer.

[00396] Embodiment 11-92. A compound of any one of Embodiments II-1 to 11-74, or a pharmaceutically acceptable salt thereof, for use in treating an immune-mediated disease.

[00397] Embodiment 11-93. Use of a compound of any one of Embodiments II-1 to 11-74, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating an immune-mediated disease.

[00398] Embodiment 11-94. A compound of any one of Embodiments II-1 to 11-74, or a pharmaceutically acceptable salt thereof, for use in treating an age related condition.

[00399] Embodiment 11-95. Use of a compound of any one of Embodiments II-1 to 11-74, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating an age related condition.

Examples

[00400] The disclosure is further illustrated by the following examples and synthesis examples, which are not to be construed as limiting this disclosure in scope or spirit to the specific procedures herein described. It is to be understood that the examples are provided to illustrate certain embodiments and that no limitation to the scope of the disclosure is intended thereby. It is to be further understood that resort may be had to various other embodiments, modifications, and equivalents thereof which may suggest themselves to those skilled in the art without departing from the spirit of the present disclosure and/or scope of the claims. [00401] Definitions used in the following examples and elsewhere herein are:

CH 2 CI2, DCM Methylene chloride, Dichloromethane

CH 3 CN, MeCN Acetonitrile

DIPEA Diisopropylethyl amine or Hunig’s base

DMA Dimethy l acetamide

DME Dimethoxyethane

DMF N,N-Dimethylformamide

EDCI 1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide

EtOAe Ethyl acetate h hour H 2 O Water

HCl Hydrochloric acid

HOBt Hydroxybenzotriazoie

HPLC High-performance liquid chromatography

LCMS Liq uid chroma tography-ma s s spectrometry

MeOH Methanol

MTBE Methyl tert-butyl ether

Na 2 S0 4 Sodium sulfate

PEG Poly eth y lene glycol

TBDMS / -butyldimethylsilyl

TEA Trifluoroacetic acid

THE T etrahydrofuran

TMS Tetramethylsilane

Series 1 bifunctional rapalogs

[00402] A general structure of Series 1 bifunctional rapalogs is shown in Scheme 1 below. For these types of bifunctional rapalogs, the linker may include variations where q = 0 to 30, such as q = 1 to 7, and r = I to 6. The linker amine can include substitutions, such as R = H and C1-C6 alkyl groups. The carbamate moiety, where Z 1 = O or S, can be attached to the rapalog at R’°or R S (Formula I and II), including variations found in Table 1 in the Examples Section. An mTOR active site inhibitor can attach to the linker via a primary or secondary amine, and may include variations found in Table 2 in the Examples Section.

Scheme 1. Series 1 bifunctional rapalogs.

Series 2 bifunctional rapalogs

[00403] A general structure of Series 2 bifunctional rapalogs is shown in Scheme 2 below. For these types of bifunctional rapalogs, the linker may include variations where q = 0 to 30, such as q = 1 to 7. The linker amine can include substitutions, such as R - H and C1-C6 alkyl groups. The pre-linker amine can include substitutions, such as R 2 = H, CT--C6 alkyl groups, and eycloalkyl including 4 to 8-rnembered rings. The carbamate moiety, where Z 1 - O or S, can be attached to the rapalog at R 40 or R 28 (Formula I and II), including variations found in Table 1 in the Examples Section. An mTOR active site inhibitor can attach to the linker via a primary or secondary amine, and may include variations found in Table 2 in the Examples Section.

Scheme 2. Series 2 bifunctional rapalogs. amine containing pre-linker

Series 3 bifunctional rapalogs

[00404] A general structure of Series 3 bifunctional rapalogs is shown in Scheme 3 below. For these types of bifunctional rapalogs, the linker may include variations where q = 0 to 30, such as q = 1 to 7. The linker amine can include substitutions, such as R = H and C1-C6 alkyl groups. The post-linker amine can include substitutions, such as R 2 = H, C1--C6 alkyl groups, and cycloalkyl including 4 to 8-membered rings. The carbamate moiety where Z 1 = O or S, can be attached to the rapalog at R 40 or R 28 (Formula I and II) including variations found in Table 1 in the Examples Section. An mTOR active site inhibitor can attach to the linker via a primary or secondary amine, and may include variations found in "fable 2 in the Examples Section. Scheme 3. Series 3 hifunctional rapalogs amine containing post-linker

Series 3 Bifunctionai rapalog

Series 4 bifunctional rapalogs

[00405] A general structure of Series 4 bifunctionai rapalogs is shown in Scheme 4 below'. For these types of bifunctionai rapalogs, the linker may include variations where q - 0 to 30, such as q = 1 to 7. The linker amine can include substitutions, such as R - H and C1---C6 alkyl groups. The pre- and post-linker amines can each include substitutions, such as R 2 = H, CT-C6 alkyl groups and cycloalkyl including 4 to 8-membered rings. The carbamate moiety, where Z 1 = O or S, can be attached to the rapalog at R 40 or R 8 (Formula I and II), including variations found in Table 1 in the Examples Section. An mTOR active site inhibitor can attach to the linker via a primary or secondary amine, and may include variations found in Table 2 in the Examples Section.

Scheme 4. Series 4 hifunctional rapalogs amine containing amine containin

Series 5 bifunctionai rapalogs

[00406] A general structure of Series 5 bifunctionai rapalogs is shown in Scheme 5 below'. For these types of bifunctionai rapalogs, the pre-linker amine can include substitutions, such as R 2 — H, C1---C6 alkyl groups, and cycloalkyl including 4 to 8-membered rings. The carbamate moiety, where Z 1 - O or S, can be attached to the rapalog at R 40 or R 28 (Formula I and II), including variations found in Table 1 in the Examples Section. An mTOR active site inhibitor can attach to the linker via a primary or secondary amine, and may include variations found in Table 2 in the Examples Section. Scheme 5. Series 5 bifunctional rapalogs amine containing

Series 6 bifunctional rapalogs

[00407] A general structure of Series 6 bifunctional rapalogs is shown in Scheme 6 below. For these types of bifunctional rapalogs, the linker may include variations where q = 0 to 30, such as q - 1 to 7. The linker amines can include substitutions, such as R - H and C1-C6 alkyl groups. The post-linker amine can include substitutions, such as R 2 = H, C1--C6 alkyl groups, and cycloalkyl including 4 to 8-mernbered rings. The carbamate moiety, where Z 1 = O or S, can be attached to the rapaiog at R 40 or R 28 (Formula I and ΪI), including variations found in Table 1 in the Examples Section. An mTGR active site inhibitor can attach to the linker via a primary or secondary amine, and may include variations found in Table 2 in the Examples Section.

Scheme 6. Series 6 bifunctional rapalogs. amine containing post-linker bitor

Series 7 bifunctional rapalogs

[00408] A general structure of Series 7 bifunctionai rapalogs is shown in Scheme 7 below. For these types of bifunctionai rapalogs, the linker may include variations where q - 0 to 30, such as q = 1 to 7. The linker amine can include substitutions, such as R = H and C1-C6 alkyl groups. The pre- and post-linker amines can each include substitutions, such as R 2 - H, CT-C6 alkyl groups, and cycloalkyl including 4 to 8-memhered rings. The carbamate moiety, where Z 1 = O or S, can be attached to the rapaiog at R 40 or R 28 (Formula I or II), including variations found in Table 1 in the Examples Section. An mTOR active site inhibitor can attach to the linker via a primary or secondary amine, and may include variations found in Table 2 in the Examples Section.

Scheme 7. Series 7 bifunctional rapalogs

Series 8 bifunctional rapalogs

[00409] A general structure of Series 8 bifunctional rapalogs is shown in Scheme 8 below. For these types of bifunctional rapalogs, the linker may include variations where q = 0 to 30, such as q = 1 to 7. The linker amine can include substitutions, such as R - H and C1-C6 alkyl groups. The post-linker amine can include substitutions, such as R = H, C1-C6 alkyl groups, and cycloalkyl including 4 to 8-membered rings. The carbamate moiety, where Z 1 = C) or S, can be attached to the rapalog at R 40 or R 28 (Formula I or II), including variations found in Table 1 in the Examples Section. An mTOR active site inhibitor can attach to the linker via a primary or secondary amine, and may include variations found in Table 2 in the Examples Section.

Scheme 8. Series 8 bifunctional rapalogs amine containing post-iinker amine containing post-linker

Table 1. Carbonate and tbiocarbonate containing rapalog monomers.

Table 2. Active Site inhibitor monomers.

Table 3. Active Site inhibitor monomers

Table 4 Amine containing pre- and post-linkers.





Preparation of Active Site Inhibitor Monomers

Monomer A, 5-(4-amino -1-(4-(amino methyl)benzyl)-1H-pyrazolo[3,4-d]pyrimidm-3- yl)beozo[d]oxazoI-2-am e triflaoroacetie acid salt

Step 1: Synthesis of tert-butyl 4-((4-amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1- yl)methy 1 sbenzylcarbamate

[00410] To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (3.8 g, 14.56 mmol, 1.0 equiv) in DMF (20 mL) was added NaH (582.27 mg, 14.56 mmol, 60 wt.%, 1.0 equiv) at 0 °C and the reaction solution was stirred at this temperature for 30 min, then tert -butyl 4- (bromomethyl)benzylcarbamate (4.59 g, 15.29 mmol, 1.05 equiv) was added to the reaction at 0 °C and the reaction solution was stirred at room temperature for 2 h. The solution was poured into HkO (80 mL) and the solid that precipitated out was filtered. The solid cake was washed with H 2 O (2 x 10 mL) and then dried under reduced pressure to give ierf-butyl 4-((4- amino-3-iodo-1H-pyrazoIo[3,4-d]pyrimidin-1-yl)metliyI)benzyl carbamate (5 g, 53% yield) as a yellow solid. LCMS (ESI) m/z: [M + Na] calcd for C 1 glTrJNeCb: 503.07; found 503.2.

Step 2: Synthesis of tert-butyl 4-((4-amino-3-(2-aminobenzo[d]oxazol-5-yI)-1H-pyrazolo[3,4- d]pyrimidm-1-yl)methyl)benzylcarbamate

[00411] To a bi-phasie suspension of /erl-butyl 4-((4-amino-3-iodo-1H-pyrazolo[3,4- d]pyrimidin-1-yl)methyl)benzylcarbamaie (5 g, 7.68 mmol, 1.0 equiv), 5-(4, 4,5,5- letramethyl-1,3,2-dioxaboro1an-2-y1)benzo[d]oxazo1-2-amme (2.40 g, 9.22 mmol, 1.2 equiv) and Pd(PPli3)4 (887.66 mg, 768.16 μmol, 0.1 equiv) in DME (100 mL) and HzO (50 mL) was added NazCO 3 (1.91 g, 23.04 mmol, 3.0 equiv) at room temperature under N 2 . The mixture was stirred at 110 °C for 3 h. The reaction mixture was cooled to room temperature and filtered, the filtrate was extracted by EtOAc (3 x 50 ml,)- The organic phases were combined and washed with brine (10 mL), dried over NaaSCfi, filtered, and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by silica gel chromatography (0→20% MeOB/EtOAc) to give teri-butyl 4-((4-amino-3-(2- aminobenzo[d]oxazol-5-yl)-1H-pyrazolo[3,4-d]pyrmiidin-1-yl)m ethyl)benzylcarbamate (4.5 g, 82% yield) as a yellow solid. LCMS (ESI) m/z: [M + H] calcd for (CslTeNsO 3 : 487.22; found 487.2.

Step 3: Synthesis of 5-(4-amino-1-(4-(aminomethyl)benzyl)-1H-pyrazolo[3,4-d] pyrimidin-3- yl)benzo[d]oxazol-2-amine

[00412] To a solution of tert-butyl 4-((4-amino-3-(2-aminobenzo[d]oxazol-5-yl)-1H- pyrazolo[3,4-d]pyrimidin-1-yl)methyl)benzylcarbamate (4.5 g, 6.29 mmol, 1.0 equiv) in DCM (50 mL) was added TEA (30.80 g, 270.12 mmol, 20 mL, 42.95 equiv) at 0 °C. The reaction solution was stirred at room temperature for 2 h. The reaction solution was concentrated under reduced pressure to give a residue, which was dissolved in 10 mL of MeCN, then poured into MTBE (100 mL). The solid that precipitated was then filtered and the solid cake was dried under reduced pressure to give 5-[4-amino-1-[[4- (aminomethyI)phenyl]methyl]pyrazo3o[3,4-d]pyrimidin- 3-yi]-1,3-benzoxazol-2-amine (2.22 g, 71% yield, TFA) as a yellow solid. LCMS (ESI) m/z: [M + H] calcd for CioHisNsO: 387.16; found 387.1.

Monomer B. 2-(4-amino-1-(4-amino butyl)-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-1H-mdol- 6-0I trifluoroacetic add salt.

Step 1: Synthesis of tert-butyl N-(4-{4-amino-3-[6-(benzyloxy)-1H-indol-2-yl]-1H- pyrazoio[3 ,4-d ]pyrimidin-1-yl } butyl)carbamate

[00413] To a mixture of lert-butyl (4-(4-amino-3-iodo-1H-pyrazoio[3,4-d]pyrimidin-1- yl)butyl)carbamate (300 mg, 694 mitioΐ, 1.0 equiv) and (6-(benzyloxy)-1-(i<?/t- butoxycarbonyl)-1H-indol-2-yl)boronic acid (763 mg, 2.08 mmol, 3.0 equiv) in DMF (2.6 mL), EtOH (525 mΐ,), and H 2 O (350 μL) were added Pd(OAc) 2 (15.5 mg, 69 mchoΐ, 0.1 equiv), triphenylphosphine (36.1 mg, 138 μmol, 0.2 equiv), and sodium carbonate (440 mg, 4.16 mmol, 6.0 equiv). The reaction was heated at 80 °C for 20 h, cooled to room temperature, and quenched with H 2 O (10 mL) and EtOAc (10 mL). The mixture was transferred to a separatory funnel and the aqueous phase was extracted with EtOAc (3 x 20 mL). The combined organic phase was washed with sat. aq. NaCl (1 x 20 mL), dried over NaaSO- , filtered, and concentrated under reduced pressure. The crude material was purified by silica gel chromatography (20→85% EtO Ac/heptane) to provide the product (201 mg, 46% yield) as an orange solid. LCMS (ESI) rn/'z: [M + H] calcd for C 2 9H 33 N 7 O 3 : 528.27; found 528.2.

Step 2: Synthesis of m/t-bntyl (4-(4-amino-3-(6-hydroxy-1H-indol-2-yl)-1H-pyrazolo[3,4- d]pyrimidin-1-yl)butyl)carbamate

[00414] To a solution of t -butyl N-(4- { 4-amino-3-[6-(benzyloxy)-1H-indol-2-yl]-1H- pyrazolo[3,4-d]pyrimidin-1-yl}butyl)carbamate (1.0 equiv) in EtOH is added Pd/C (10 mol%). The reaction is purged with H 2 and the reaction allowed to stir under an atmosphere of H 2 until consumption of starting material, as determined by LCMS. The reaction is then diluted with EtOAc filtered over Celite, and concentrated under reduced pressure. The resultant residue is purified by silica gel chromatography to afford the desired product.

Step 3: Synthesis of 2-(4-amino-1-(4-aminobutyl)-1H-pyrazolo[3,4-d]pyrimidin-3-yl )-1H- indol-6-ol

[00415] To a solution of feri-butyl (4-(4-amino-3-(6-hydroxy-1H-indol-2-yl)-1H- pyrazolo[3,4-d]pyrimidin-1-yl)butyl)carbarnaie (1.0 equiv) in anhydrous DCM is added TFA (50 equiv.) dropwise at 0 °C. The reaction is stirred at 0 °C and wanned to room temperature. Once the reaction is complete, as determined by LCMS, the reaction is concentrated under reduced pressure. The residue is triturated with MeCN, then dripped into MTBE over 10 min. The supernatant is removed and the precipitate is collected by filtration under Ni to give 2-(4- amino-1-(4-aminobutyl)- lH-pyraz,olo[ 3,4-d]py rimidin-3-y 1 j-1H-indol-6-ol .

Monomer C. 5-(4-amino-1-((1,2,3,4-tetrahydroisoquinolin-6-yl)methyI)-1H - pyrazolo[3,4-d]pyrimidin-3-yI)benzo[d]oxazol- 2-amine trill uoroacetic add salt.

Step 1: Synthesis of tert-butyl 6-((4-amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1- yl)methyl)-3,4-dihydroisoquinoIine-2(1H)-carboxylate

[00416] To a suspension of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (5 g, 19.16 mmol, 1.0 equiv) in DMF (50.0 mL) was added NaH (766.22 mg, 19.16 mmol, 60 wt.%, 1.0 equiv) at 4 C C. The mixture was stirred at 4 °C for 30 min. To the reaction mixture was added ie -butyl 6-(bromometliyl)-3,4-dihydroisoquinoline-2(1H)-carboxylate (6.87 g, 21.07 mmol, 1.1 equiv) in DMF (30 mL) at 4 °C. The mixture was stirred at room temperature for 2 h. The mixture was then cooled to 4 °C and KbO (400 mL) was added and the mixture was stirred for 30 min. The resulting precipitate was collected by filtration to give crude tert- butyl 6-((4-amino-3-iodo-IH-pyrazolo[3,4-d]pyrimidin-1-yl)methyl)- 3,4- dihydroisoquinoline-2(1H)-carboxylate (9.7 g, 76% yield) as a light yellow solid. The crude product was used for the next step directly. Step 2: Synthesis of ie/f-butyl 6-((4-amino-3-(2-aminobenzo[d]oxazol-5-yl)-1H- pyrazolo[3,4-d]pyrimidin-1-yl)methyl)-3,4-dihydroisoquinolin e-2(1H)-carboxyIate

[00417] To a bi-phasic suspension of tert-butyl 6-((4-amino-3-iodo-1H-pyrazolo[3 4- d]pyrimidin-1-yl)methyI)-3,4-dihydroisoquinoline-2(lH)-carbo xy]ate (9.7 g, 14.63 mmol, 1.0 equiv), 5-(4,4,5,5-tetramethyl-1,3 » 2-dioxaborolan-2-yl)benzo[d]oxazol-2-amme (4.57 g,

17.55 mmol, 1.2 equiv), and bfeCO 3 (7.75 g, 73.14 mmol, 5.0 equiv) in DME (120.0 mL) and H 2 O (60 mL) was added PdCPPhsL (1.69 g, 1.46 mmol, 0.1 equiv) at room temperature under N2. The mixture was stirred at 110 °C for 3 h. The reaction mixture was then cooled to room temperature and partitioned between EtOAc (100 mL) and H 2 O (100 mL). The aqueous layer was separated and extracted with EtOAc (2 x 60 ml,). The organic layers were combined, washed with brine (80 mL) and dried over anhydrous Na 2 S0 4 , filtered and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel chromatography (1→10Q% EtOAc/petroleum ether, then 20→50% MeOH/EtOAc) to afford tert-butyl 6-((4-amino-3-(2-aminobenzo[d]oxazol-5-yl)-1H-pyrazolo[3,4-d ]pyrimidin-1- yl)rnethyl)-3,4-dihydroisoquinoline-2(1H)-carboxylate (4.5 g, 58% ' yield) as a light yellow solid.

Step 3: Synthesis of 5-(4-amino-1-((1,2,3,4-tetrahydroisoquinolin-6-yl)methyl)-1H - pyrazolo[3,4-d]pyramidin-3-y])benzo[d]oxazol-2-amine

[00418] To neat TEA (32.5 mL, 438.97 mmol, 50.0 equiv) was added tert-butyl 6-((4- amino-3-(2-aminobenzo[d]oxazol-5-yl)-1H-pyrazolo[3,4-d]pyrim idin-1-yl)methyl)-3,4- dihydroisoquinoline-2(lH)-carboxylate (4.5 g, 8.78 mmol, 1.0 equiv) at room temperature. The mixture was stirred for 30 min and then concentrated under reduced pressure. The oily residue was triturated with MeCN (8 mL), then dripped into MTBE (350 mL) over 10 min. The supernatant was removed and then the precipitate was collected by filtration under N 2 to give 5-(4-amino-1-((1,2,3,4-tetrahydroisoquinolin-6-yl)methy])-1H -pyrazolo[3,4- d]pyrimidin-3 -yl)benzo [d]oxazol-2-amine (5.72 g over 100% yield, TEA) as a light pink solid. LCMS (ESI) m/z: [M + H] calcd for C 2 2H 2 0N8O: 413.18; found 413.2.

Monomer D, 2-(4-amino-1-(4-aminobutyl)-1H-pyrazolo[3,4-d]pyrimidin-3-yl )-1H-indol- 7-ol trifluoroacetic add salt.

Step 1: Synthesis of m/t-bntyl 2-(4-amino-1-(4-(( ri-biitoxycarbonyl)amino)butyl)-1H- pyrazolo[3,4-d]pvrimidin-3-yl)-7-methoxy-IH-indole-1-carboxy late

[00419] To a mixture of tert-butyl (4-(4-amino-3-iodo-1H-pyrazoIo[3,4-d]pyriniidin-1- yl)butyl)carbamate (1.0 equiv) and (1-(ieri-butoxycarbonyl)-7-methoxy-1H-indol-2- yl)boronic acid (3.0 equiv) in DME and H 2 O is added Pd< PPn R (0.1 equiv) and sodium carbonate (6.0 equiv). The reaction is heated at 80 °C until completion, as determined by LCMS and TLC analysis. The reaction is then quenched with H 2 O and EtOAc. The mixture is transferred to a separatory funnel and the aqueous phase is extracted with EtOAc. The organic phase is washed with sat. aq. NaCI, dried over Na 2 S0 4 , filtered, and concentrated under reduced pressure. The desired product is isolated after chromatography on s lica gel.

Step 2: Synthesis of m/t-bntyl 2-(4-aimno-1-(4-(( Jt-biitoxycarbonyl)amino)butyl)-1H- pyrazolo[3,4-d]pvrimidin-3-yl)-7-hydroxy-IH-indole-1-carboxy late

[00420] To a solution of ten-butyl 2-(4-amino-1-(4-((i -butoxycarbonyI)amino)butyl)- 1H-pyrazolo[3,4-d]pyrimidin-3-yl)-7-methoxy-1H-mdole-1-carbo xy late (1.0 equiv) in DCM at -10 °C is added BBr, (2.0 equiv). The reaction is allowed to stir until consumption of starting material, as determined by LCMS. The reaction is quenched by slow addition of sat. aq. NaHCO 3 , transferred to a separatory funnel and the mixture is extracted with DCM. The organic phase is washed with sat. aq. Nad, dried over Na SCL, filtered, and concentrated under reduced pressure. The desired product is isolated after chromatography on silica gel.

Step 3: Synthesis of 2-(4-amino-1-(4-ammobutyl)-1H-pyrazolo[3,4-d]pyrimidin-3-yl) -1H- indol-7-ol

[00421] To a solution of tert-butyl 2-(4-amino-1-(4-((fev -butoxycarbonyI)ammo)butyl)- 1H-pyrazolo[3,4-d]pyrimidin-3-yl)-7-hydroxy-1H-indole-i-carb oxylate (1.0 equiv) in DCM at 0 °C is added TEA dropwise. The reaction is stirred at 0 °C and warmed to room temperature. Once the reaction is complete, as determined by LCMS, the reaction is concentrated under reduced pressure. The residue is triturated with MeCN, then dripped into MTBE over 10 min. The supernatant is removed and the precipitate is collected by filtration under Ni to give 2-(4-amino-1-(4-aminobutyl)-1H-pyrazolo[3,4-d]pyrimidm-3-yl) -TH-indol-

7-ol.

Monomer E, 5-(4-amino-1-(piperidm-4-ylmethyI)-1H-pyrazoio[3,4-d]pyrimid m-3- yl)benz:o[(l]oxazoI-2-amme trifluoroacetic add salt.

Step i: Synthesis of tert-buty] 4-((4-amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1- yl)methyl)piperidine-1-carboxylate

[00422] To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (3 g, 11.49 mmol, 1.0 equiv) in DMA (30 mL) was added teri-butyl 4-(bromomethyl)piperidine-1-carboxylate (3.36 g, 12.07 mmol, 1.05 equiv) and K 2 CO 3 (4.77 g, 34.48 mmol, 3.0 equiv), then the reaction was stirred at 80 °C for 3 h. The reaction mixture was filtered to remove K 2 CO 3 and the filtrate was poured into H 2 O (200 mL). A solid precipitated was then filtered to give tert- butyl 4-((4-amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1-yl)methyl)p iperidine-1-carboxylate (3 g, 57% yield) as a light yellow solid. LCMS (ESI) rn/z [M + H] calcd for C 1 6H 2 3IN6O 2 : 459.10; found 459.1.

Step 2: Synthesis of / -butyl 4-((4-amino-3-(2-ammobenzo[d]oxazol-5-yl)-1H- pyrazoio[3,4-d]pyrimidin-1-yl)methyl)piperidine-1-carboxylat e

[00423] To a bi-phasic suspension of tert-butyl 4-((4-amino-3-iodo-1H-pyrazolo[3,4- d]pyrimidin-1-yljmethyljpiperidine-1-carboxylate (3 g, 6.55 mmol, 1.0 equiv) and 5-(4, 4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)benzo[d]oxazol-2-amine (2.04 g, 7.86 mmol, 1.2 equiv) and Na 2 C0 3 (3.47 g, 32.73 mmol, 5.0 equiv) in DME (60 mL) and H 2 O (30 mL) was added Pd(PPh3)4 (756.43 mg, 654.60 μmol, 0.1 equiv) at room temperature under N2. The mixture was stirred at 110 C C for 3 h and the two batches were combined together. The reaction mixture was cooled and partitioned between EtOAc (500 mL) and H 2 O (500 mL). The aqueous layer was separated and extracted with EtOAc (3 x 300 mL). All the organic layers were combined, washed with brine (20 mL), dried over anhydrous Na SO^ filtered, and the filtrate was concentrated under reduced pressure to give ieri-butyl 4-((4-amino-3-(2- ammobenzo[d]oxazol-5-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)me thyl)piperidine-1- carboxylate (4.5 g, 74% yield) as a yellow solid. LCMS (ESI) m/z: )M + H] calcd for C 2 3H 2 8N8O 3 : 465.24; found 465.2.

Step 3: Synthesis of 5-(4-amino-1-(piperidin-4 -ylmethyl)-1H-pyrazolo[3,4-d]pyrimidin-3- y l)benzo [d] o azol 2 -amine

[00424] A solution of len-hutyl 4-((4-amino-3-(2-aminobenzo[d]oxazol-5-yl)-1H- pyrazolo[3,4-d]pyrimidin-1-yl)methyl)piperidme-1-carboxylate (2.5 g, 5.38 mmol, 1.0 equiv) in TEA (25 mL) was stirred at room temperature for 30 min. The reaction solution was concentrated under reduced pressure to remove TFA. The residue was added to MTBE (400 mL) and a solid precipitated, which was then filtered to give 5-(4-amino-1-(piperidm-4- ylmethyl)-1H-pyrazolo[3 ,4-d]pyrimidin-3-yl)benzo[d]oxazol-2-amine (2.7 g, over 100 % yield, TFA) as a yellow solid. LCMS (ESI) m/z [M + H] calcd for C 1 gHhoNgO: 365.18; found 365.1.

Monomer F. 2-(4-amino-1-(4-aminohutyl)-1H-pyray.alo[3,4-d]pyrimidin-3-y l)-1H-indoI- 5-ol trifluoroacetic add salt.

Step 1: Synthesis of tert-butyl (4-(4-aimno-3-(5-((ieri-butyldimethylsilyl)oxy)-1H-indol-2- yl)-1H-pyrazolo[3,4-d]pyriinidin-1-yl)butyl)carbamate

[00425] To a solution of tert-huty\ (4-(4-amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1- yl)butyl)carbamate (1.0 g, 2.31 mmol, 1.0 equiv) in dioxane (10.5 mL) and H 2 O (3.5 mL) was added ( 1-(/ -butox carbony l)-5 -((ieri-butyldimethylsilyl)oxy)-1H-indol-2-yl)boronie acid (1.54 g, 2.78 mmol, 1.2 equiv), K3PO4 (1.47 g, 6.94 mmol, 3.0 equiv), Pd2(dba)3 (211.84 mg, 231.34 μmol, 0.1 equiv), and SPhos (189.95 rng, 462.69 mihoΐ, 0.2 equiv) at room temperature under N2. The sealed tube was heated at 150 °C for 20 min in a microwave. This was repeated for 9 additional batches. The 10 batches were combined and the reaction mixture was cooled and partitioned between EtOAc (60 mL) and I¾0 (80 mL). The aqueous layer was separated and extracted with EtOAc (2 x 50 mL). The organic layers were combined, washed with brine (60 mL) and dried over anhydrous Na 2 S0 4 . The suspension was filtered and the filtrate was concentrated under reduced pressure. The crude material was purified by silica gel chromatography (1→75% EtOAc/petroleum ether). The desired fractions were combined and evaporated under reduced pressure to give tert-butyl (4-(4- amino-3-(5-((/eri-butyldimethylsilyl)oxy)-1H-indol-2-yl)-1 -pyrazolo[3,4-d]pyrimidin-1- yl)butyl)carbamate (10 g, 60% yield) as a light yellow solid.

Step 2: Synthesis of tort-butyl (4-(4-amino-3-(5-hydroxy-1H-indol-2-yl)-1H-pyrazolo[3,4-d] pyrimidin-1-yl)but.yl)earbamate

[00426] To a mixture of to 7-hutyl (4-(4-amino-3-(5-((¾ri-butyldimethylsilyl)oxy)-1H- indol--2-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)butyl)carbamat e (10 g, 18.12 mmol, 1.0 equiv) in THF (100 mL) was added TBAR·3H 2 q (1 M, 54.37 mL, 3.0 equiv) in one portion at room temperature under N 2 . The mixture was stirred for 1 h and then H 2 O (100 ml,) was added to the reaction mixture. The layers were separated and the aqueous phase was extracted with EtOAc (2 x 80 mL). The combined organic phase was washed with brine (100 mL), dried with anhydrous Na SCL, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (l-→67% EtOAc/ petroleum ether) to afford torf-butyl (4-(4-amino-3-(5-hydroxy-1H-indol-2-yl)-1H-pyrazolo[3,4-d]py rimidin-1-yl)butyl)carbamate (7 g, 87% yield) as a light pink solid.

Step 3: Synthesis of 2-[4-amino-1-(4-aimnobutyl)pyrazolo[3,4-d]pyrimidin-3-yl]-1H -indol-5- ol

[00427] To TFA (50.0 mL, 675.26 mmol, 38.9 equiv) was added tert-butyl (4-(4-amino-3- (5-hydroxy-1H-indoi-2-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)b utyl)carbamate (7.6 g, 17.37 mmol, 1.0 equiv) at room temperature. The mixture was stirred for 40 min and was then concentrated under reduced pressure. The oily residue was triturated with MeCN (20 mL), then added dropwise into MTBE (300 mL) for 10 min. The supernatant was removed and then the precipitate was collected by filtration under N2 to give 2 -[4-amino-1-(4-- ammobutyl)pyrazolo[3,4-d]pyrimidin-3-yl]-1H-indol-5-ol (7.79 g, 91% yield, TFA) as light yellow solid. LC S (ESI) m/z: [M + H] calcd for C 1 7H J 9N7O: 338.17; found 338.2. Monomer G, 5-(4-amino -1-(azetidm-3-yIsBethy!)-1H-pyFazo!o[3,4-d]pyrimk!m-3- yl)benzo[d]oxazol-2-amine trifluoroacetic add salt.

Step I : Synthesis of teri-butyl 3-((4-amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1-yl) methyl)azetidine-1-carboxylate

[0(1428] To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (4 g, 15.32 mmol, 1.0 equiv), ierf-butyl 3-(hydroxymethyl)azetidine-1-carboxylate (3.01 g, 16.09 mmol, 1.05 equiv) and PPI13 (6.03 g, 22.99 mmol, 1.5 equiv) in THF (80 mL) cooled to 0 °C was added DIAD (4.47 mL, 22.99 mmol, 1.5 equiv), dropwise. After the addition was complete, the reaction was stirred at room temperature for 14 h. The reaction was poured into H 2 O (200 mL) and then extracted with EtOAc (3 x 50 mL). The organic layers were combined and washed with brine (2 x 50 mL). The organic phase was dried over NaaSCL, filtered, the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by silica gel chromatography (Q--->100% EtOAc/petroleum ether) to give ieri-butyl 3-((4- amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1-yl)methyl) azetidine-1-carboxylate (4.2 g, 64% yield) as a white solid. LCMS (ESI) m/z [M + H] calcd for C HisINeCL: 431.07; found 431.0.

Step 2: Synthesis of tert-butyl 3-((4-amino-3-(2-aminobenzo[d]oxazol-5-yl)-1H-pyrazolo [3 ,4-d]pyrimidin-1-yl)methyl)azetidine-1-carboxylate

[00429] To a bi-phasic suspension of tert-butyl 3-((4-amino-3-iodo-1H-pyrazolo[3,4- d]pyrimidin-1-yl) methyl)azetidine-1-carboxylate (4 g, 9.30 mmol, 1.0 equiv), 5-(4, 4,5,5- tetramethyl-1,3,2 -dioxaborolan-2-yl)benzo[d]oxazol-2-amine (2.90 g, 11.16 mmol 1.2 equiv) and Na CO 3 (4.93 g, 46.49 mmol, 5.0 equiv) in DME (100 L) and H 2 O (50 mL) was added 1M(R1¾) 4 ( 1.07 g, 929.71 μmol, 0.1 equiv) at room temperature under N2. The mixture was stirred at 110 °C for 3 h. The reaction mixture was then cooled to room temperature and filtered, and the filtrate was extracted by EtOAc (3 x 50 mL). The organic layers were combined and washed with brine (10 mL), dried over Na. ' SG:. filtered and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by silica gel chromatography (0→ 2Q% MeOH/EtOAc) to give tert-bniyl 3-((4-amino-3-(2- aminobenzo[d]oxazol-5-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)m ethyl)azetidine-1- carboxylate (3.5 g, 80% yield) as a yellow solid. LCMS (ESI) m/z: [M + H] calcd for C 2 1H 2 4N8O 3 : 437.20; found 437.2.

Step 3: Synthesis of 5-(4-amino-1-(azetidin-3-ylmethyl)-1H-pyrazolo[3,4-d]pyrimid in- 3- yl)benzo[d]oxazol-2-amine

[00430] To a solution of ierf-butyl 3-((4-arnino-3-(2-aimnobenzo[d]oxazol-5-yl)-TH- pyrazolo[3,4-d] pyrimidin-1-yl)methyl)azetidine-1-carboxylate (3.29 g, 6.87 mmol 1.0 equiv) in DCM (20 mL) was added TFA (7.50 mL, 101.30 mmol, 14.7 equiv) at 0 °C. The reaction was warmed to room temperature and stirred for 2 h. The reaction solution was concentrated under reduced pressure to give a residue. The residue was dissolved in MeCN (6 mL) and then poured into MTBE (80 mL). A solid precipitated, which was filtered and the solid cake was dried under reduced pressure to give 5-[4- amino-1-(azetidin-3- ylmethyl)pyrazolo[3,4-d]pyrimidin-3-yl]-i,3-benzoxazol-2-ami ne (4.34 g, over 100% yield, TFA) as a yellow solid. LCMS (ESI) m/z [M + H] calcd for C 1 eHieNsO: 337.15; found 337.1.

Monomer H, 5-(4-aimno-1-(4-aminobutyl)-1H-pyrazolo[3,4-d]pyrinndm-3-yl) benzo[d]- oxazoI-2-amme trifluoroacetic add salt.

[00431] This monomer was synthesized following the procedures outlined in Nature 2015, 534 , 272-276 which is incorporated by reference in its entirety.

Monomer I. 5-(4-amino -1-(pyrrolidm-3-ylmethyl)-1H-pyrazolo[3,4-d]pyrimidin-3- yl)benzo[d]oxazol-2-amine trifluoroacetic add salt.

Step 1: Synthesis of tert-butyl 3-((4-ammo-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1-yl) methyl)pyrrolidine-1-carhoxylate

[00432] A suspension of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (4.5 g, 17.24 mmol, 1.0 equiv), tert-butyl 3-(bromomethyl)pyrrolidine-1-carboxylate (4.78 g, 18.10 mmol, 1.05 equiv) and K 2 CO 3 (7.15 g, 51.72 mmol, 3.0 equiv) in DMA (40 mL) was heated to 85 °C. The reaction was stirred at 85 °C for 3 h, at which point the solution was cooled to room temperature. Then, H 2 O (80 mL) was added to the reaction, and a solid precipitated out. The mixture was filtered, and the solid cake was washed with H 2 O (2 x 40 mL), and then dried under reduced pressure to give fe t-hutyl 3-((4-ammo-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1- yl) methyl)pyrrolidine-1-carboxylate (6 g, 78% yield) as a yellow solid. LCMS (ESI) m/z:

[M + H] calcd for C 1 5H 2 1IN6O 2 : 445.08; found 445.1.

Step 2: Synthesis of / -butyl 3-[[4-amino-3-(2-amino-1,3-benzoxazol-5-yl)pyrazolo[3,4-d] pyrimidin 1-y 1] me thy l]pyrrolidine-1- carboxylate

[00433] To a bi-phasic suspension of I -butyl 3-((4-amino-3-iodo-1H-pyrazoIo[3,4- d] pyrimidin-1-yl) methyl)pyrrolidine-1-carboxylate (4 g, 9.00 mmol, 1.0 equiv), 5-(4, 4,5,5- tetramethyl-1,3,2- dioxaborolan-2-yl)benzo[d]oxazol-2-amine (2.81 g, 10.80 mmol, 1.2 equiv) and NaiCO 3 (4.77 g, 45.02 mmol, 5.0 equiv) in DME (120 mL) and H 2 O (60 mL) was added Pd(PPh . )4 (1.04 g, 900.35 μmol, 0.1 equiv) at room temperature under N 2 . The mixture was stirred at 110 °C for 3 h. The reaction mixture was cooled to room temperature and filtered and the filtrate was extracted with EtOAc (3 x 50 mL). The organic phases were combined and washed with brine (50 mL), dried over Na SCL, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel chromatography (0→20% MeOH/EtOAc) to give tert-butyl 3-((4-amino-3-(2- aminobenzo[d]oxazol-5-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl) methyl)pyrrolidine-1- carboxylate (3 g, 64% yield) as a yellow' solid. LCMS (ESI) m/z: [M + H] calcd for C 2 2H 2 0N8O 3 : 451.21, found 451.2. Step 3: Synthesis of 5-(4-amino-1-(pyrrolidin-3-ylmethyl)-1H-pyrazolo[3,4-d]pyrim idin- 3- yl)benzo[d]oxazol-2-amine

[00434] To a solution of tert-butyl 3-((4-amino-3-(2-aminobenzo[d]oxazol-5-yl)-1H- pyrazolo[3,4-d]pyrimidin-1-yl)methyl)pyrrolidine-1-carboxyla te (3 g, 6.66 mmol, 1.0 equiv) in DCM (40 mL ) was added TEA (20 mL) at 0 °C, dropwise. The reaction mixture was warmed to roo temperature and stirred for 2 h. The reaction solution was then concentrated under reduced pressure to give a residue. The residue was dissolved in MeCN (4 mL), then poured into MTBE (100 mL), and a solid precipitated out. The solid was filtered and the cake was dried under reduced pressure to give 5-(4-amino-1-(pyrrolidin-3-ylmethyl)-1H- pyrazolo[3,4-d]pyrimidin-3-yl)benzo[d]oxazol-2-amine (4.00 g, over 100% yield, TEA) as a yellow solid. LCMS (ESI) m/z: [M + H] calcd for CxrHigNgO: 351.17; found 351.2.

Monomer J. 1-(4-amino butyl)-3-(7-methoxy-1H-mdol-2-yl)-1H-pyrazolo[3,4- d]pyrimidin-4-aminetrifluoroacetic acid salt.

Step 1: Synthesis of ierf-butyl 2-(4-amino-1-(4-((fe/?-butoxycarbonyl)amino)butyl)-1H- pyrazolo[3,4-d]pyrimidin-3-yl)-7-methoxy-1H-mdole-1-carboxyl ate

[00435] To a mixture of tert-hutyl (4-(4-amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1- yl)butyi)carbamate (1.0 equiv) and (1-(/er/-butoxycarbonyl)-7-methoxy-1H-indol-2- yl)boronic acid (3.0 equiv) in DME and H 2 O is added Pd(PPhs)4 (0.1 equiv) and sodium carbonate (6.0 equiv). The reaction is heated at 80 °C until completion, as determined by LCMS and TLC analysis. The reaction is then quenched with H 2 O and EtOAc. The mixture is transferred to a separator )'· funnel and the aqueous phase is extracted with EtOAc. The organic phase is washed with sat. aq. NaCl, dried over Na 2 S0 4 , filtered, and concentrated under reduced pressure. The desired product is isolated after chromatography on silica gel.

Step 2: Synthesis of 1-(4-aminobutyl)-3-(7-methoxy-1H-indol-2-yl)-1H-pyrazolo[3,4 - d]pyrimidin-4- amine

1 m s [00436] To a solution of tert-hutyl 2-(4-amino-1-(4-((ieri-butoxycarbonyl)amino)butyl)- 1H-pyrazolo[3,4-d]pyrimidin-3-yI)-7-hydroxy-1H-indole-1-carb oxylate (1.0 equiv) in DCM at 0 °C is added TFA dropwise. The reaction is stirred at 0 °C and warmed to room temperature. Once the reaction is complete, as determined by LCMS, the reaction is concentrated under reduced pressure. The residue is triturated with MeCN, then dripped into MTBE over 10 min. The supernatant is removed and the precipitate is collected by filtration under Ni to give 1-(4-aminobutyl)-3-(7-metboxy-1H-indol-2-yl)-1H-pyrazolo[3,4 - d]pyrimidin-4-amine.

Monomer K, Synthesis of 1-(4-aminobutyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine triflnoroacetie acid salt.

Step 1: Synthesis of tert-butyl (4-(4-amino-1H-pyrazolo[3,4-d]pyrimidixi-1- yl)butyl)carbamate

[00437] To a mixture of iert-butyl (4-(4-amino-3-iodo-1 i-t-pyrazolo[3,4-d]pyrimidin-1- yl)butyl)carbamate (300 mg, 694 μmol, 1.0 equiv) in MeOH (14 mL) at 0 °C was added zinc dust (226 mg, 3.46 mmol, 5.0 equiv). Sat. aq. NH4CI (14 mL) was added to the reaction mixture and the reaction was warmed to room temperature and stirred for 18 h. The reaction was quenched by EtOAc (40 mL) and H 2 O (10 mL) and the mixture was transferred to a separatory funnel. The aqueous phase was extracted with EtOAc (3 x 20 mL) and the combined organic phases were washed with sat. aq. NaHCO 3 (15 mL), dried over Na SCL, filtered, and concentrated under reduced pressure to provide the product (210 mg, 99% yield) as a light yellow solid that was used without further purification. LCMS (ESI) m/z: [M + H] calcd for C 1 4H 2 2N6O 2 : 307.19; found 307.1.

Step 2: Synthesis of 1-(4-aminobuty]>iH-pyrazofo[3,4-d]pyrimidin-4-amine

[00438] To a solution of tert-buty! (4-(4-amino-1H-pyra olo[3,4-d]pyrimidin-1- yl)butyl)carhamate (210 mg, 691 mihoΐ) in DCM (3.5 mL) at 0 °C was added TFA (3.5 mL), dropwise. After 3 h, the reaction was warmed to room temperature and concentrated under reduced pressure to provide the trifluoroacetale salt of the product (220 mg, 99% yield) as a

1 brown oil, which was used without further purification. LCMS (ESI) m/z: [M + H] calcd for C 9 HI 4 N 6 : 207.13; found 207.1.

Monomer L. 1-[4-(piperazin-1-yl)-3-(trifluoromethyl)phenyl]-9-(quinoIm- 3-yl)-1H,2H- benzofh] 1 ,6-iiaphthyridiii-2-one

[§§439] The preparation of this monomer has been previously reported in the literature.

See the following references: i) Liu, Qingsong; Chang, Jae Won; Wang, Xinhua; Kang, Seong A.; Thoreen, Carson C ; Markliard, Andrew; Hur, Wooyoung; Zhang, Jianming; Sim, Taebo; Sabalini, David M.; et al From Journal of Medicinal Chemistry (2010), 53(19), 7146-7155. ii) Gray, Nathanael; Chang, Jae Won; Zhang, Jianming; Thoreen, Carson C.; Kang, Seong Woo Anthony; Sabalini, David M ; Liu, Qingsong From PCT Int. Appl. (2010), WO 2010044885A2 which are incorporated by reference in their entirety.

Monomer M, 5-(1-(4-aminobutyl)-4-(dimethylamino)-1H-pyrazolo[3,4-d]pyri midin-3- I ) benzojfd ] oxazoL 2- arn i ne trifluoroacetic acid salt.

Step I : Synthesis of 3-iodo-1-trityl-1H-pyrazolo[3,4-d]pyrimidin-4-amine

[§§440] A suspension of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (10.5 g, 40.23 mmol, 1.0 equiv) in DMF (170.0 mL) was treated with CscCO 3 (19.7 g, 60.34 mmol, 1.5 equiv) and [chloro(diphenyl)methyl]henzene (13.5 g, 48.27 mmol, 1.2 equiv) at room temperature. The reaction mixture was stirred at 70 °C for 4 h under a nitrogen atmosphere. The reaction mixture was added to H 2 O (1200 mL). The precipitate was filtered and washed with H 2 O. The residue was purified by silica gel chromatography (0→60% EtOAc/ petroleum ether) to afford 3-iodo-1-triiyl-1H-pyrazolo[ 3, 4-d]pyrimidin-4-amine (15.40 g, 73.5% yield) as a white solid.

Step 2: Synthesis of 3-iodo-A%V-dimethyl-1-trityl-1H-pyrazolo[3,4-d]pyrimidin-4-a mine

[00441] To a suspension of Nall (2.98 g, 74.50 mmol, 60 wt.%, 2.5 equiv) in DMF (150 mL) was added the solution of 3-iodo-1-trityl-1H-pyrazoIo[3,4-d]pyrimidin-4-amine (15.0 g, 29.80 mmol, 1.0 equiv) in DMF (50 mL) at 0 °C. The mixture was stirred at 0 °C for 10 min. To the reaction mixture was then added iodomethane (16.92 g, 119.20 mmol, 7.42 mL, 4.0 equiv) at 0 °C. The mixture was stirred at room temperature for 2 h, at which point H 2 O (1400 mL) was added at 0 °C. The mixture was stirred for an additional 10 min at 0 °C. The resulting precipitate was collected by filtration to give crude product, which was purified by- silica gel chromatography (1%→25% EtOAc/petroleum ether) twice to afford 3-iodo -N,N- dimethyl-1-trityl-1H-pyrazolo[3,4-d]pyrimidin-4-amine (9.0 g, 89% yield) as a white solid.

Step 3: Synthesis of 3-iodo-Y,A-dimethyl-1H-pyrazolo[3,4-d]pyrimidm-4-amine

[00442] To a cooled solution of TFA (19.1 mL, 258.1 mmol, 15.0 equiv) in DCM (100.0 mL) was added 3-iodo-iV,N-dimethyl-1-trityl-1H-pyrazolo[3,4-d]pyrimidin-4- amine (9.10 g, 17.12 mmol, 1.0 equiv) at 4 °C. The mixture was stirred at room temperature for 1 h. The residue was poured into H 2 O (100 mL) and the aqueous phase was extracted with DCM (2 x 50 mL). To the aqueous phase was then added a saturated aqueous solution of NaHCO 3 until the solution was pH 8. The resulting precipitate was collected by filtration to give 3-iodo- AOV-dimethyl-1H-pyrazolo[3,4-d]pyrimidin-4-amine (3.40 g, 68.7% yield) as a white solid.

Step 4: Synthesis of tert-bulyl (4-(4-(dimethylamino)-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1- yl)butyl)carbamate

[00443] To a suspension of 3-iodo-A,A-dimethyl-1H-pyrazolo[3,4-d]pyrimidin-4-amine (1.7 g, 5.88 mmol, 1.0 equiv) in DMF (20 mL) was added NaH (247 mg, 6.17 mmol, 60 wt.%, 1.05 equiv) at 4 °C. The mixture was stirred at 4 °C for 30 min. To the reaction mixture was then added / -butyl N-(4-bromobutyl)carbamate (2.22 g, 8 82 mmol, 1.81 mL,

1.5 equiv) in DMF (10 mL) at 4 °C. The mixture was stirred at room temperature for 2 h. To the mixture was then added H 2 O (100 mL) at 4 °C. The mixture was stirred for an additional 30 min at 4 °C and the resulting precipitate was collected by filtration to give crude product. The residue was purified by silica gel chromatography (0→75% EtOAc/petroleum ether) to afford iert-hulyl(4-(4-(dirnethylamino)-3-iodo-1H-pyrazolo[3,4-d]py rimidin-1- yl)butyl)carbamate (2.0 g, 56% yield) as a white solid.

Step 5: Synthesis of tert-butyl (4-(3-(2-aminobenzo[d]oxazol-5-yl)-4-(dimethylamino)-1H- pyrazolo[3,4-d]pyrimidin-1-yl)butyl)carbarnate

[00444] To a bi -phasic suspension of ieri-butyl (4-(4-(dimethylamino)-3-iodo-1H- pyrazolo[3,4-d]pyrimidin-1-yl)butyl)earbamate (4.0 g, 8.69 mmol, 1.0 equiv), 5-(4, 4,5,5- tetrametiiyl-1,3 » 2-dioxaborolan-2-yl)benzo[d]oxazol-2-amine (3.4 g, 13.03 mmol, 1.5 equiv), and NaiiCO 3 (4.6 g, 43.45 mmol, 5.0 equiv) in DME (80.0 mL) and H 2 O (40.0 mL) was added Pd(PPli3)4 (1.0 g, 868.98 μmol, 0.1 equiv) at room temperature under N2. The mixture was stirred at 110 °C for 3 h. The reaction mixture was then cooled and partitioned between EtOAc (300 mL) and I¾0 (600 mL). The aqueous layer was separated and extracted with EtOAc (2 x 100 mL). The organic layers were combined, washed with brine (2 x 60 mL) and dried over anhydrous Na^SCfr, filtered, and concentrated under reduced pressure. The crude material was purified by silica gel column chromatography (50% EtO Ac/hexanes followed by 20% MeOH/EtOAc). The desired fractions were combined and concentrated under reduced pressure to give te -butyl (4-(3-(2-aminobenzo[d]oxazol-5-yl)-4-(dimethylamino)- 1H-pyrazolo[3,4-d]pyramidin-1-yl)butyl)carbamate (3.2 g, 78.9% yield) as a light brown solid.

Step 6: Synthesis of 5-(1-(4-aminobutyl)-4-(dimethylamino)-1H-pyrazolo[3,4-d]pyri midin-3- yl)benzo[d]oxazol-2-amine

[00445] To TEA (20.82 mL, 281.27 mmol, 36.5 equiv) was added /erf-butyl (4-(3-(2- aminobenzo[d]oxazol-5-yl)-4-(dimethylamino)-1H-pyrazolo[3,4- d]pyrimidin-1- yl)butyl)carbamate (3.6 g, 7.72 mmol, 1.0 equiv) at room temperature. The mixture was stirred for 30 min, at which point the mixture was concentrated under reduced pressure. The oily residue was triturated with MeCN (8 mL) and MTBE (60 mL) for 10 min. The supernatant was removed and then the precipitate was collected by filtration under N2 to give 5-(1-(4-aminobutyl)-4-(dimethylamino)-1H-pyrazolo[3,4-d]pyri midin-3-yl)benzo[d]oxazol- 2-amine (4.0 g, crude, TEA) as a light brown solid.

[00446] To 1 M NaOH (107.2 mL, 14.7 equiv) was added 5-(1-(4-aminobutyl)-4- (dimethylamino)-1H-pyrazolo[3,4-d]pyrimidin-3-yl)benzo[d]oxa zol-2-amine (3.5 g, crude, TEA) at roo temperature. The mixture was stirred for 10 min and then the aqueous phase

1 c nr was extracted with DCM (3 x 50 mL ). The combined organic phase was washed with brine (50 ml.), dried with anhydrous NaaSO- , filtered and concentrated under reduced pressure. TFA (539.37 μL, 7.28 mmol, 1.0 equiv) was added and concentrated under reduced pressure. MeCN (10 mL ) was then added, followed by MTBE (150 mL). The resulting precipitate was collected by filtration to give 5-(T(4-aminobutyl)-4-(dimethylamino)-1H-pyrazolo[3,4- d]pyrimidin-3-yl)benzo[d]oxazol-2-amine (1.3 g, 36.6 ( ¾ yield, TFA) as a light brown product. LCMS (ESI) m/z: [M + H] calcd for € ΐ8 H 22 N 8 0: 367.19; found 367.1.

Monomer N, 6-(4-amino -1-(4-aminobutyl)-1H-pyrazolo[3,4-d]pyrimidm-3-yl)benzo- [d]isoxazoi-3-amine triflnoroacetie acid salt.

Step 1: Synthesis of tert-butyl (6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)benzo[d]isoxazol-3-yI)carbamate

[00447] To a solution of teri-butyl (6-bromobenzo[d]isoxazol-3-yl)carbamate (1.0 equiv) in dioxane is added Pd(PPh3)4 (0.1 equiv), sodium carbonate (6.0 equiv), and bis(pinacolato)diboron (3.0 equiv). The reaction mixture is stirred and heated until completion, as determined by LCMS and TLC analysis. The reaction is cooled to room temperature, quenched with sat. aq. NaHCO 3 , and the mixture transferred to a separatory funnel. The aqueous phase is extracted with EtOAc and the organic phase is washed with sat. aq. NaCl, dried over NaaSO- , filtered, and concentrated under reduced pressure. The desired product is isolated after purification by silica gel chromatography.

Step 2: Synthesis of tert-bu l (4-(4-amino-3-(3-((l ·- hutoxyearhonyl)ammo)benzo[d]isoxazol-0-yl)-iH-pyrazolo[3,4-d ]pyrimidin-1- yl)butyl)carbamate

[00448] To a mixture of I -huiy (4-(4-amino-3-iodo-1H-pyrazoIo[3,4-d]pyrimidin-1- yl)butyl)carbamate (1.0 equiv) and tert-hutyl (6-(4,4,5,5-tetramethyl-1,3,2-dioxaboroIan-2- yl)benzo[d]isoxazol-3-yl)carbamate (3.0 equiv) in DME and H 2 O is added Pd(PPh3)4 (0.1 equiv) and sodium carbonate (6.0 equiv). The reaction is heated at 80 °C until completion, as determined by LCMS and TLC analysis. The reaction is then quenched with H 2 O and EtOAc. The mixture is transferred to a separatory funnel and the aqueous phase is extracted with EtOAc. The organic phase is washed with sat. aq. NaCl, dried over NarSO^ filtered, and concentrated under reduced pressure. The desired product is isolated after chromatography on silica gel.

Step 3: Synthesis of 6-(4-amino-1-(4-ammobutyl)-1H-pyrazolo[3,4-d]pyrimiclin-3-yl )benzo- [d]isoxazol-3-amine

[§§449] To a solution of tert-butyl (4-(4-amino-3-(3-(0¾ri- butoxycarbonyl)amino)benzo[d]isoxazol-6-yi)-1H-pyrazolo[3,4- d]pyrimidin-1- yl)butyl)carbamate (1.0 equiv) in DCM at 0 °C is added TFA, dropwise. The reaction is stirred at 0 °C and warmed to room temperature. Once the reaction is complete, as determined by LCMS, the reaction is concentrated under reduced pressure. The residue is triturated with MeCN, then added dropwise into MTBE over 10 min. The supernatant is removed and the precipitate is collected by filtration under N2 to give 6-(4-amino-1-(4- ammohutyl)-iH-pyrazolo[3,4-d]pyrimidm-3-yl)benzo-[d]isoxazol -3-amine.

Monomer O, 4-(5-(4-morphoImo-1-(I-(pyridm-3-ylmethyl)piperidm-4-yl)-1H- pyrazolo[3,4-d]pyrimidin-6-yl)-1H-indol-1-yl)butan-1-aniine triflnoroacetie acid salt.

[§§450] The synthesis of this monomer proceeds by alkylation of WAY-600 (CAS# 1062159-35-6) with len-hiityl (4-bromobutyl)carbamate under basic conditions followed by Boc-deproteetion using TFA to produce the TFA salt.

[00451 ] Reference for preparation of WAY-600: Discovery of Potent and Selective Inhibitors of the Mammalian Target of Rapamycin (mTOR) Kinase: Nowak, P.; Cole D.C.; Brooijmans, N.; Bursavich, M.G.; Curran, K.J.; Eilingboe, J.W.; Gibbons, J.J.; Hollander L; Hu, Y.; Kaplan, J.; Malwitz, D.J.; Toral-Barza, L.; Verheijen, J.C.; Zask, A.; Zhang, W.-G.; Yu, K. 2009; Journal of Medicinal Chemistry Volume 52, Issue 22, 7081-89, which is incorporated by reference in its entirety.

Monomer P, 2-(4-(8-(6-(aminomefhyl)quinolm-3-yl)-3-methyl-2-oxo-2,3-dih ydro-1H- imidazo[4,5-c]quinolin-1-y l)pheny l)-2-methylpropanenitrile triflu oroacetic add salt.

[00452] The synthesis of this monomer proceeds first by synthesis of the Suzuki reaction coupling partner (3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolane)quinolin-6-yl)-l V-boc- methanamine starting from methyl 3-bromoquinoline-6-carboxylate. Reduction of the methyl ester with lithium aluminum hydride followed by Mitsunobu reaction with phthalimide and hydrazine cleavage provides the benzylie amine. Protection of the benzylic amine with di- tert-butyl dicarbonate followed by a Miyaura borylation reaction provides (3 -(4, 4,5,5- tetramethyl-1 ,3 ,2-dioxaborolane)qumolm-0-yl)-A--boc--meihanamine .

[00453] An 8 N AG reaction of 2-(4-aminophenyl)-2-methylpropanemtrile with 6-hromo-4- chloro-3-nitroquinoline provides the substituted amino-nitro-pyridine. Reduction of the nitro group with Raney-Ni under a hydrogen atmosphere followed by cyclization with trichloromethy] chloroformate provides the aryl-substituted urea. Substitution of the free N-H of the urea with methyl iodide mediated by tetrabutylammonium bromide and sodium hydroxide followed by Suzuki coupling of (3-(4,4,5,5-tetramethyl-1,3,2- dioxaborolane)quinolin-6-yl)-/V-boc-methanamine and then Boc-deprotection using TFA produces the TFA salt.

[00454] Reference for preparation of 2-[4-(8-bromo-3-methyl-2-oxo-2,3-dihydro-imidazo [4,5 -c]quino3in-1 -yl)-phenyl] -2-methyl-propionitrile: Vannucchi, A.M.; Bogani, C.; Bartalucci, N. 2016. JAK PI3K/mTOR combination therapy. US9358229. Novartis Pharma AG, Incyte Corporation, which is incorporated by reference in its entirety .

Monomer , 8-(6-methoxypyridio-3-yl)-3-meihyl-1 [4-(piperazm-1-yl) 3 (trinuoromethyl)phenyI]-1H,2H,3H-linldazo[4,5-c]quinolin-2-o ne

[00455] This monomer is a commercially available chemical known as BGT226(CAS# 1245537-68-1). At the time this application was prepared, it was available for purchase from several vendors as the free amine.

Monomer R. 3-(4-amino -1-(4-amino butyl)-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-N-(4,5- dihydrothiazol-2-yl)henzamide triflu oroacetic acid salt

Step 1: Synthesis of tert-butyl (4-(4-amino-3-(3-((4,5-dihydrothiazoI-2- yl)carbamoyl)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)butyl) carbamate

[00456] To a solution of (3-((4,5-dihydrothiazol-2-yl)carbamoyl)phenyl)boronic acid (500 mg, 1.15 mmol, 1.0 equiv) and /erz-butyl (4-(4-amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin- 1-yl)butyl)carbamate (575 mg, 2.30 mmol, 2.0 equiv) in dioxane (19.1 mL), EtOH (3.8 mL), and FLO (2.3 mL) was added Pd(PP¾3)4 (265 mg, 230 μmol, 0.2 equiv) and sodium carbonate (730 mg, 6.89 mmol, 6.0 equiv). The reaction mixture was sonicated until formation of a clear, yellow solution, which w'as subsequently heated at 80 °C for 14 h. The reaction was then diluted with sat. aq. NaCl (30 niL) and the mixture transferred to a separatory funnel.

The aqueous phase was extracted with DCM (3 x 25 mL). The combined organic phases were dried over Na SCC, filtered, and concentrated under reduced pressure. The desired product was isolated as a yellow ' solid (324 mg, 53% yield) after silica gel chromatography (0→ 15% MeOH/DCM). LCMS (ESI) m/z: [M + H] calcd for C 2 4H30N8O 3 S: 511.22; found 511.2.

Step 2: Synthesis of 3-(4-amim> 1-(4-aminobutyl)-1H-pyrazolo[3,4-d]pyrimidin-3-yl )-N-(4,5- dihydrothiazol-2-yl)benzamide

[00457] To a solution of ieri-butyl (4-(4-amino-3-(3-((4,5-dihydrothiazol-2- yl)carbamoyl)phenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yi)butyl) carbamate (324 mg, 614 μmol) in DCM (4.1 mL) at 0 °C was added TFA (1.5 mL), dropwise. After 1 h, the reaction was warmed to room temperature and concentrated under reduced pressure to provide the trill uoroacetate salt of the product as a yel low solid (320 mg, 99% ' yield). Used without further purification. LCMS (ESI) m/z : [M + H] calcd for C 19 H 22 N 8 OS: 411.16; found 411.1

Monomer S. 2-(5-(4-morpholino-1-(1-(pyridin-3-ylmeihyl)piperidin-4-yl)- 1H- pyrazolo[3,4-d]pyrimidm-6-yl)-1H-mdol-3-yl)ethan-1-amine.

[00458] The synthesis of this monomer proceeds by condensation of 2,4,6- trichloropyrimidine-5-earbaldehyde with 3-((4-hydrazineylpiperidin-1-y])methyl)pyridine hydrochloride. Reaction of the product with morpholine followed by a Suzuki reaction with boronic ester gives the Boc-protected amine. Final deprotection with TFA gives the monomer. This synthesis route follows closely to the reported preparation of highly related structures in the following references: i) Nowak, Pawel; Cole, Derek C.; Brooijmans, Natasja; Curran, Kevin j.; Ellingboe, John W.; Gibbons, James J.; Hollander, Irwin; Hu, Yong Bo; Kaplan, Joshua; Malwitz, David J.; et al From Journal of Medicinal Chemistry (2009),

52(22), 7081-7089. ii) Zask, Arie; Nowak, Pawel Wojciech; Yerheijen, Jeroen; Curran,

Kevin J.; Kaplan, Joshua; Malwitz, David; Bursa vich, Matthew Gregory; Cole, Derek Cecil; Ayral-Kaloustian, Semiramis; Yu, Ker; et al From PCT Ini. Appl. (2008), WO 2008115974 A2 20080925, which are incorporated by reference in their entirety.

Monomer T, 1-(4-aminobutyI)-3-iodo-1H-pyrazoIo[3,4-d]pyrimidm-4-amine trifluoroacetic add salt.

[00459] To a mixture of ierf-butyl (4-(4-amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1- yl)butyl)carbamate (496 mg, 1.14 mmol, 1.0 equiv) in DCM (5.7 mL) at 0 °C was added TFA (1.5 mL) dropwise. The reaction was allowed to stir at 0 °C for 1 h, at which time the reaction was concentrated under reduced pressure to provide a yellow' solid (505 mg, 99% yield) w'hich was taken on without further purification. LCMS (ESI) rn/'z: [M + H] calcd for C9H13IN & : 333.02; found 332.9.

Monomer II. 5-(4-amino -I-(4-(methylamino )butyl)-1H-pyrazolo[3,4-d]pyriimdiii-3- yl)benzo[d]oxazol-2-amine trifluoroacetic add salt.

Step 1: Synthesis of teri-butyl (4-hydroxybutyl)(methyl)carbamate

[00460] To a solution of 4-(methylaniino)butan-1-ol (0.5 g, 4.85 mmol, 104.2 mL, 1.0 equiv) in DCM (10 mL) at room temperature was added B0C 2 O (1.06 g, 4.85 mmol, 1.11 mL, 1.0 equiv). The mixture was stirred for 3 h at room temperature and then the mixture was concentrated under reduced pressure at 30 °C. The residue was purified by silica gel chromatography (100/1 to 3/1 petroleum ether/EtOAc) to afford /erf-butyl (4- hydroxybutyl)(methyl)carbamate (0.9 g, 91.4% yield) as a colorless oil.

Step 2: Synthesis of tert-butyl (4-bromobutyl)(methyl)carbamate

[00461] To a solution of reri-butyl (4-hydroxybutyl)(methyl)carbamate (0.9 g, 4.43 mmol, 1.0 equiv) in THF (20 mL) at room temperature was added PPhs (2.21 g, 8.41 mmol, 1.9 equiv) and CBn; (2.79 g, 8.41 mmol, 1.9 equiv). The mixture was stirred for 1 h and then the reaction mixture was filtered and concentrated. The residue was purified by silica gel chromatography (1/0 to 4/1 petroleum ether/EtOAc) to afford ieri-butyl (4- hromobutyl)(methyl) carbamate (1.1 g, 93.3% yield) as a colorless oil.

Step 3: Synthesis of ieri-bvAyl (4-(4-amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1-yl) butyl) (methyl)carbamaie

[00462] To a suspension of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (0.9 g, 3.45 mmol, 1.0 equiv) in DMF (10 mL) at 4 °C was added NaH (137.92 mg, 3.45 mmol, 60 wt.%, 1.0 equiv). The mixture was stirred at 4 °C for 30 min and then a solution of tert-butyl (4- bromobutyl)(methyl)carbamate (1.01 g, 3.79 mmol, 25.92 mL, 1.1 equiv) in DMF (3 mL) was added. The mixture was stirred at room temperature for 3 h, at which point FLO (100 mL) was added. The aqueous phase was extracted with EtOAc (3 x 30 mL) and the combined organic phases were washed with brine (20 mL), dried with anhydrous Na 2 S0 4 , filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (1/0 to 0/1 petroleum ether/EtOAc) to afford fert-butyl (4-(4-amino-3-iodo-1H-pyrazolo[3,4- d]pyrimidin-1-yl)butyl) (methyl) carbamate (1.2 g, 78% yield) as a white solid. LCM8 (ESI) m/z [M + H] calcd for iViF JMXL: 447.10; found 447.1.

Step 4: Synthesis of tert-butyl (4-(4-amino-3-(2-aminobenzo[d]oxazol-5-yl)-1H- pyrazolo[3,4- d] pyrimidin-1-yl)butyl)(methyl)carbamate

[90463] To a bi-phasic suspension of fe t-hutyl (4-(4-amino-3-iodo-1H-pyrazolo[3,4-d] pyrimidin-1-yl)butyl)(methyl)carbamate (1.2 g, 2.69 mmol, 1.0 equiv), 5-(4, 4,5,5- tetramethyl-1,3,2- dioxaborolan-2-yl)benzo[d]oxazol-2-amine (1.19 g, 3.23 mmol, 1.2 equiv), and Na CO 3 (1.42 g, 13.44 mmol, 5.0 equiv) in DME (20 mL) and FbO (10 mL) at room temperature w ' as added Pd(PPli3)4 (310.71 mg, 268.89 μmol, 0.1 equiv) under Nc. The mixture was stirred at 110 °C for 3 h and then the reaction mixture was cooled and partitioned between EtOAc (20 mL) and H 2 O (15 mL). The aqueous layer was separated and extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (2 x 20 mL), dried over anhydrous NaaSCE, filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (1/0 to 4/1 EtOAc/MeOH) to give icri-butyl (4-(4-amino-3-(2- aminobenzo[d]oxazol-5-yl)-1H-pyrazolo [3,4-d]pyrimidin-1 - yl)butyi)(methyl) carbamate (0.78 g, 62.5% yield) as an orange solid.

Step 5: Synthesis of 5--(4--amino-1--(4--(methylamino)butyl)-1H--pyrazolo[3,4--d] pyrimidin-3- y 1) benzo [d] oxazol-2-amine

100464] A solution of ieri-butyl(4-(4-ajmino-3-(2-ammobenzo[d]oxazol-5-yl)-1H- pyrazoio[3,4-d]pyrimidin-1-yl)butyl)(methyl)carbamate (0.78 g, 1.72 mmol, 1.0 equiv) in TEA (5 mL) at room temperature was stirred for 30 min. The solution was concentrated under reduced pressure and the oily residue was triturated with MeCN (1 mL) and then added to MTBE (100 ml.)- The supernatant was removed and then the precipitate was collected by filtration under Nr to give 5-(4-amino-1-(4-(methylamino) butyl)-1H-pyrazolo[3,4- d]pyrimidin-3-yl)benzo[d]oxazol -2-amine bis-trifliiorosiilfonate (0.959 g, 93% yield) as an orange solid. LCMS (ESI) rn/z [M + H] calcd for CnH 2 oNsO: 353.18; found 353.1.

Monomer V. 1-(4-(4-(5-(aminnmethyl)pyrImIdin-2-yl)pIperazio-1-yl)-3-

(iriiluoromethyI)plienyl)-8-i6-iiielhoxypyridiii-3-yl)-3- methyI-1,3-diliydro-2H- miidazo[4,5-c]qn oHn-2-one.

Step 1: Synthesis of tert-butyl N-ieri-butoxycarbonyl-N-[(2-cliloiOpyrimidin-5-yl)methyl] carbamate

[00465] To a solution of tert-huty\ N-ier/-bntoxycarbonylcarbamate (7.33 g, 33.74 mmol, 1.0 equiv) in DMF (80 mL) was added NaH (1.62 g, 40.49 mmol, 60 wt.%, 1.2 equiv) at 0 °C. r fhe mixture was stirred at 0 °C for 30 min and then 5-(bromomethyl)-2-chloro- pyrimidine (7 g 33.74 mmol, 1 equiv) was added. The reaction mixture was stirred at room temperature for 1.5 h and then the mixture was poured into sat. NH4CI (300 mL) and stirred for 5 min. The aqueous phase was extracted with EtOAc (3 x 80 mL) and the combined organic phases were washed with brine (50 mL), dried over anhydrous Na SOi, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (20:1 to 1:1 petroleum etlier/EtO Ac) to afford terf-butyl N-½ri-butoxycarbonyl-N-[(2-chloro pyrimidin-5-yl)methyl]carbamate (7.0 g, 60.3% yield) as a white solid. LCMS (ESI) rn/z: [M + H] calcd for C 1 5H 2 2CIN3O4: 344.14; found 344.2.

Step 2: Synthesis of tert-butyl N-teri-butoxycarbonyl-N-[[2-[4-[4-[8-(6-methoxy-3-pyridyl)-

3-methyl-2-oxo-imidazo[4,5-c]quinolin-1-yl]-2-(trifluorom ethyl)phenyl]piperazin-1- yl]pyrimidin-5-yl]methyl]carbamate

[00466] To a solution of 8-(6-methoxy-3-pyridyl)-3-methyl-1-[4-piperazin-1-yl-3- (trifluoromethyl)phenyl]imidazo[4,5-c]quinolin-2-one (0.4 g, 748.32 μmol, 1.0 equiv) in MeCN (7 mL) was added reri-butyl N-ieri-butoxycarbonyl-N-[(2-chloropyrimidin-5- yl)methyl] carbamate (514.55 mg, 1.50 mmol, 2.0 equiv) and K 2 CO 3 (413.69 mg, 2.99 mmol, 4 equiv) at room temperature. The reaction mixture was stirred at 80 °C for 14 h and then the mixture w'as cooled to room temperature, filtered and concentrated under reduced pressure. The residue was purified by washing with M’TBE (5 mL) to give tert-butyl N -tert- butoxycarbonyl-N-[[2-[4-[4-[8-(6-methoxy-3-pyridyl)-3-methyl -2-oxo-imidazo[4,5- c]quinolin-1-yl]-2-(trifluoromethyl)phenyl]piperazin-1-yl]py rimidin-5-yl]meihyl]carbamate (0.57 g, 90.5% yield) as a light yellow solid. LCMS (ESI) m/z: [M + H] calcd for C 4 3H46F3N9O6: 842.36; found 842.7

Step 3: Synthesis of 1-[4-[4-[5-(aminomethyl)pyrimidin-2-yl]piperazin-1-yl]-3- (tdfluoromethyl) phenyl]-8-(6-methoxy-3-pyridyl)-3-methy]-imidazo[4,5-c]quino lin-2-one

[00467] A solution of ten-butyl N-ii?ri-butoxycarbonyl-N-[[2-[4-[4-[8-(6-methoxy-3- pyridyl)-3-methyl-2-oxo-imidazo[4,5-c]quinolin-1-yl]-2-(trif luoromethyl)phenyl]piperazin-1- yl]pyrirmdin-5-yl]methyl]carbamate (095 g, 1.13 mmol, 1 equiv) in TEA (10 mL) was stirred at room temperature for 1 h, at which point the solvent was concentrated. The residue was dissolved in MeCN (10 mL) and then the solution was added to MTBE (150 mL), dropwise. The precipitate was collected to give 1-[4-[4-[5-(aminomethyl)pyrimidin-2-yl]piperazin-1- yl]-3-(trifluoromethyl)phenyl]-8-(6-methoxy-3-pyridyl)-3-met hyl-imidazo[4,5-c]quinolin-2- one trifluoromethanesulfonate (0.7 /8 g, 84.8% yield) as a yellow solid. LCMS (ESI) m/z : [M + H] ealcd for C 3 3H 3 0F3N9O 2 : 642.26; found 642.4

Monomer W. I-(4-amino butyI)-3-(1H-pyrroloI2,3-b]pyridin-5-yI)pyrazolo(3,4- d]pyriimdin-4-aimne.

Step 1: Synthesis of tert-butyl N-[4-[4-amino-3-(1H-indol-5-yl)pyrazolo[3,4-d]pyrimidin-1- y 1] b u tyl] carbama te

[00468] To a bi-phasic suspension of ierf-butyl N-[4-(4-amino-3-iodo-pyrazolo[3,4- d]pyrimidin-1-yl)butyl]carbamate (8 g, 18.51 mmol, 1 equiv), 5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-1H-pyrrolo[2,3-b]pyridine (5.42 g, 22.21 mmol, 1.2 equiv) and NaaCO 3 (9.81 g, 92.54 mmol, 5 equiv) in diglyme (160 mL) and H 2 O (80 mL) was added Pd(PPh . 3)4 (2.14 g, 1.85 mmol, 0.1 equiv) at room temperature under N2. The mixture was stirred at 110 °C for 3 h. The reaction mixture was cooled to room temperature, filtered and the filtrate was partitioned between EtOAc (500 mL) and H 2 O (500 mL). The aqueous layer was separated and extracted with EtOAc (3 x 300 mL). The organic layers were combined, washed with brine (20 mL) and dried over anhydrous NaaSOr, then filtered and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel chromatography (1/0 to 0/1 petroleum ether/EtOAc then 4/1 EtOAc/MeOH) to give teri-butyl N-[4-[4-amino- 3-(1H-indol-5-yl)pyrazolo[3,4-d]pyrimidin-1-yl]butyl]carbama te (6.6 g, 84.6% yield) as a yellow solid. LCMS (ESI) m/z [M + H] ealcd for C 2 2H 2 7N7O 2 : 422.22; found 423.3.

Step 2: Synthesis of 1-(4-aminobutyl)-3-(1H-pyn O lo[2,3-b]pyridin-5-yl)pyrazolo[3,4- d]pyriniidm-4-amine

[00469] To /f.T/-butyl N-[4-[4-amino-3-(1H-indol-5-yl)pyrazolo[3,4-d]pyrimidin-1- yl]butyl]carbamate (6.6 g, 15.66 mmol, 1 equiv) was added TEA (66 mL), which was then stirred at room temperature for 30 min. The reaction solution was concentrated under reduced pressure to remove TEA and then MTBE (400 mL) was added to the residue. The suspension was stirred for 15 min, at which point the yellow' solid was filtered, and the solid cake dried under reduced pressure to give 1-(4-aminobutyl)-3-(1H-pyrrolo[2,3-b]pyridin-5- yl)pyrazolo[3,4-d]pyrimidin-4-amine (10.2 g, 97.1% yield) as a yellow' solid. LCMS (ESI) rn/z [M + H] calcd for C 1 eHisNs: 323.17; found 323.1.

Monomer X. 2-(4-amino-1-((1,2,3 5 4-tetrahydroisoquinolm-6-yI)methyl)- 1H- pyrazolo[3,4-d]pyrimidm-3-yl)-1H-indol-5-ol 2,2,2-trifluoroacetate.

Step 1: Synthesis of tert-butyl 6-((4-amino-3-(5-((ieri-butyldimethyIsilyl)oxy)-1H-indol-2- yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)methyI)-3,4-dihydroisoq uinoline-2(1H)-carboxylate

[00470] To a solution of tert-butyl 6-((4-amino-3-iodo-1H-pyrazoio[3,4-d]pyrimidin- 1- yl)methyl)-3,4-dihydroisoquinoline-2(lH)-carboxylate (1 g, 1.97 mmol, 1.0 equiv) in dioxane (10.5 mL) and H 2 O (3.5 mL) was added (1-(ieri-butoxycarbonyl)-5-((ieri- butyldimethylsilyl)oxy)-1H-indol-2- yljboronic acid (1.16 g, 2.96 mmol, 1.5 equiv), K3PO4 (1.26 g, 5.92 mmol, 3.0 equiv), Pd2(dba)3 (180.85 mg, 197.50 (tmol, 0.1 equiv), and SPhos (162.16 mg, 394.99 μmol, 0.2 equiv) at room temperature under N 2 . The sealed tube was heated at 150 °C for 20 min under microwave. The reaction mixture was then cooled and 6 separate batches were combined together. The reaction mixture was partitioned between EtOAc (100 mL) and H 2 O (100 mL). The aqueous layer was separated and extracted with EtOAc (3 x 80 mL). The organic layers were combined, washed with brine (100 mL) and dried over anhydrous Na2S()4. The solution was filtered and the filtrate was concentrated under reduced pressure. The crude material was purified by silica gel column chromatography (100/1 to 1/4 petroleum ether/EtOAc) to give lerl-butyl 6-((4-amino-3-(5- ((ieri-butyldimethylsilyl)oxy)-1H-indol-2-yl)-1H-pyrazolo [3,4-d]pyrimidin-1-yl)methyl)- 3,4-dihydroisoquinoline-2(1H)-carboxyIate (6.17 g, 829% yield) as a light yellow' solid.

Step 2: Synthesis of /eri-butyl 6-((4-amino-3-(5-hydroxy-1H-mdol-2-yl)-1H-pyrazolo[3,4- d]pyrimidin-1-yl)methyl)-3,4-dihydroisoquinoline-2(1H)-carbo xylate

[00471] To a mixture of fe t-hutyl 6-((4-amino-3-(5-((½ri-butyldimethylsilyl)oxy)-1H- indol-2-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)methyl)-3,4-dih ydroisoquinoline-2(lH)- carboxylate (6.17 g, 9.86 mmol, 1.0 equiv) in THF (100 mL ) was added tetrabutylammonium fluoride trihydrate (1 M, 10.84 mL, 1.1 equiv) in one portion at 0 °C under N 2 . The mixture was stirred at 0 °C for 1 h and was then added to H 2 O (100 mL). The aqueous phase was extracted with EtOAc (3 x 80 mL) and the combined organic phase was washed with brine (2 x 80 mL), dried with anhydrous Na SCL, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (1/1 to 0/1 petroleum ether/EtOAc) to afford fe t-butyl 6-((4-amino-3-(5-hydroxy-1H-indol-2-yl)-1H-pyrazolo[3,4-d]py rimidm-1- yl)methyl)-3,4-dihydroisoquinoline-2(1H)-carboxylate (4 g, 79.3% yield) as a light pink solid LCMS (ESI) m/z: [M + H] calcd for C 2 8H 2 9N7O 3 : 512.24; found 512.3

Step 3: Synthesis of 2-(4-amino-1-((l ,2,3,4-tetrahydroisoquinolin-6-yl)methyl)- 1H- pyrazolo[3,4-d]pyrimidin-3-yl)-1H-indol-5-ol 2,2,2-trifluoroacetate

[00472] To a solution of tert-buiyl 6-((4-amino-3-(5-hydroxy-1H-indol-2-yl)-1H-pyrazolo [3,4-d]pyrimidin-1-yl)methyl)-3,4-dihydroisoquinoline-2(l H)-carboxylate (4.5 g, 8.80 mmol, 1.0 equiv) in MeOH (50 mL) was added HCl in MeOH (4 M, 50 mL, 22.7 equiv) at room temperature. The mixture was stirred at room temperature overnight and was then concentrated under reduced pressure. To the crude product was added EtOAc (100 mL) and the resulting precipitate was collected by filtration under N2 to give 2-(4-amino-1-(( 1 ,2,3 ,4- tetrahydiOisoquinoiin-6-yl)methyl)-1H-pyrazoio[3,4-d]pyrimid in-3-yl)-1H-indol-5-oi 2,2,2- trifluoroacetate (4.1 g, 85.0% yield, 3HCl) as a light yellow' solid. LCMS (ESI) m/z [M + H] calcd for C 2 3H 2 1N7O: 412.19; found 412.1.

Monomer Y. 3-(1H-pyrrolo[2,3-b]pyridin-5-yl)-1-((1,2,3,4-tefrahydroisoq ui nolin-6- yl)methyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine 2,2,2-trifluoroacetate.

Step 1: Synthesis of /eri-butyi 6-(bromometbyl)-3,4-dibydroisoquinoline-2(1H)- carboxylate

[00473] A solution of NBS (34.07 g, 191 39 mmol 4 equiv) in THF (200 mL) was added in portions to a solution of tert-hutyl 6-(hydroxymethyl)-3,4-dihydroisoquinoline-2(lH)- carboxylate (12.6 g, 47.85 mmol, 1.0 equiv) and triphenylphosphine (37.65 g, 143.55 mmol, 3.0 equiv) in THF (200 mL) at 0 °C. After the addition was complete, the mixture was stirred for 1 h at room temperature. EtOAc (150 mL) was added and the mixture was washed with H 2 O (200 mL) and brine (150 mL), dried over anhydrous Na2SOi and concentrated under reduced pressure. The residue was purified by silica gel chromatograph (100/1 to 10/1 petroleum ether/EtOAc) to afford tert-butyl 6-(bromomethyI)-3,4-dihydroisoquinoline-2(1H)- carboxylate (8.56 g, 54.8% yield) as a light yellow solid.

Step 2: Synthesis of /eri-butyl 6-((4-amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1-yl) methyl)-3,4-dihydroisoquinoline-2(1H)-carboxylate

[00474] To a suspension of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (9.5 g, 3640 mmol, 1.0 equiv) in DMF (110 mL) was added NaH (1.46 g, 36.40 mmol, 60 wt.%, 1.0 equiv) at 0 °C. The mixture was stirred at 0 °C for 30 min at which point a solution of tert- butyl 6-(bromomethyl)-3,4-dihydroisoquinoiine-2(1H)-carboxylate (12.47 g, 38.22 mmol, 1.05 equiv) in DMF (40 mL) was added at 0 °C. The mixture was stirred at room temperature for 1 h and then H 2 O (1000 L) was added at 0 °C. The mixture stirred at 0 °C for 30 min and then the resulting precipitate was collected by filtration to give ieri-butyl 6-((4-amino-3- iodo-1H-pyrazolo[3,4-d] pyrimidin-1-yl)methyl)-3,4-dihydroisoquinoline-2(1H)-carboxy late (17.8 g, 76.3% yield) as a light yellow solid, which was used the next step directly. LCMS (ESI) m/z [M + H] calcd for CioMNeC .: 507.10; found 507.1.

Step 3: Synthesis of t -butyl 6-((4-amino-3-(1H-pyrrolo[2,3-b]pyridin-5-yl)-1H- pyrazolo[3 ,4-d]pyrimidin-· 1-yl)methyl) -3 ,4-dihydiOisoquinoline-2( 1H)-carboxylate

[00475] To a bi-phasic suspension of tert-butyl 6-((4-amino-3-iodo-1H-pyrazolo [3,4-d] pyrimidin-1-yl)methyl)-3,4-dihydroisoquinoline-2(1H)-carboxy late (6.5 g, 10.14 mmol, 1.0 equiv), 5-(4,4,5,5-teframethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrrolo [2 3 -b] pyridine (2.97 g, 12.16 mmol, 1.2 equiv), and NaeCO 3 (5.37 g, 50.68 mmol, 5.0 equiv) in diglyme (100 ml ) and H 2 O (50 mL) was added Pd(PPh3)4 (1.17 g, 1.01 mmol, 0.1 equiv) at room temperature under N2. The mixture was stirred at 110 °C for 3 h. The reaction mixture was then cooled and partitioned between EtOAc (100 mL) and H 2 O (100 mL). The aqueous layer was separated and extracted with EtOAc (2 x 100 mL). The combined organic phase was washed with brine (100 mL), dried with anhydrous Na SCL, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (0/1 to 1/4 MeOH/EtOAc) to afford tert-butyl 6-((4-aniino-3-(lH-pyrrolo[2,3-h]pyridin-5-yl)-TH-pyrazolo[3 ,4-d]pyramid in-1-yl) methyl)-3,4-dihydroisoquinoline-2(1H)-carboxylate (3.77 g, 72.1% yield) as a light yellow solid LCMS (ESI) m/z [M + H] calcd for CerlLsNsC b: 497.24; found 497.3.

Step 4: Synthesis of 3-(1H-pyrrolo[2,3-b]pyridin-5-yl)-1-((1,2,3,4-tetrahydroiso quinolin-6- yl)methyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine 2,2,2-trifluoroacetaie

[00476] tert- Butyl 6-((4-amino-3-(1H-pyrrolo[2,3-b]pyridin-5-yl)-1H-pyrazolo[3, 4-d] pyrimidin-1-yl)methyl)-3,4-dihydroisoquinoline-2(1H)-carboxy late (3.77 g, 7.59 mmol, 1.0 equiv) was added to TFA (85.36 mL, 1.15 mol, 151.8 equiv) at room temperature. The reaction mixture was stirred for 1 h. It was then concentrated under reduced pressure and the oily residue was triturated with MeCN (3 mL), then dripped into MTBE (200 mL) for 5 min. The supernatant was removed and then the precipitate was collected by filtration under N2 to give the product, which was dissolved in MeCN (20 mL), and finally concentrated under reduced pressure to give 3-(1H-pyrrolo[2, 3-b]pyridin-5-yl)-1-(( 1,2,3, 4-tetrahydroisoquinolin- 6-yl)metiiyl)-1H-pyrazolo[3,4-d]pyrimidin-4- amine 2,2,2-trifluoroacetate (4.84 g, 85.0% yield, 3TFA) as a light yellow solid. LCMS (ESI) m/z : [M + H] calcd for C 2 2H 2 0N8: 397.19; found 397.2. Monomer Z. (4-((2-aminoethyl)suIfonyl)-3-fluoro-2-methylphenyI)(7- (6-aminopyridin- 3-yl)-2,3-dihydrobenzo|f]|T,4]oxazepiii-4(5H)-yl)methanone 2,2,2-trifluoroacetate.

Step 1: Synthesis of methyl 3,4-difluoro-2-methylbenzoate

[00477] To a solution of 3,4-difluoro-2-methylbenzoic acid (2 g, 11.62 mmol, 1.0 equiv) in DMF (20 mL) was added K 2 CO 3 (4.82g, 34.86 mmol, 3.0 equiv) and iodomethane (3.26 mL, 52.29 mmol, 4.5 equiv) at room temperature. The mixture was stirred at room temperature for 3 h. The solution of methyl 3,4-diiluoro-2-methylhenzoate in DMF (20 mL) was used directly in the next step.

Step 2: Synthesis of methyl 4-((2-((ieri-butoxycarbonyl)amino)ethyl)thio)-3- fLuoro-2- methylbenzoate

[00478] To a solution of methyl 3,4-difluoro-2-methylbenzoate (2.16 g, 11.28 mmol, 1.0 equiv) in DMF (20 mL) was added rerf-butyl (2-mercaptoethyl)carbamate (2.0 g, 11.28 mmol, 1 equiv) and K 2 CO 3 (3.12 g, 22.56 mmol, 2.0 equiv) at room temperature. The reaction was stirred at 110 °C for 12 h, at which point the mixture was added to H 2 O (50 mL). The aqueous solution was then extracted with EtOAc (3 x 30 mL) and the organic phase was combined and concentrated under reduced pressure. The residue was purified by silica gel chromatography (I/O to 3/l petroleum ether/EtOAc) to afford methyl 4-((2-((i¾r/- butoxycarbonyl)amino)ethyI)thio)-3-fluoro-2-methylbenzoate (3.0 g, 76% yield) as light yellow solid.

Step 3: Synthesis of methyl 4-((2-(( eri-butoxycarbonyl)amino)ethyl)sulfonyl)-3- fluoro-2- methylbenzoate

[00479] To a solution of methyl 4-((2-((ie/r-butoxycarbonyl)amino)ethyl)thio)-3-fluoro-2- rnelhylbenzoate (3.3 g, 9.6 i mmol, l.O equiv), NaOH (2 M, 4.80 mL, l.O equiv), and NaHCO 3 (2.42 g, 28.83 mmol, 3.0 equiv) in acetone (30 mL) was added potassium peroxymonosulfate (12.35 g, 20.08 mmol, 2.1 equiv). The mixture was stirred for 12 h at room temperature and then the mixture was acidified to pH 5 by addition of IN HCl. The aqueous layer was extracted with EtOAc (3 x 30 mL) and the combined organic phase was washed with brine (20 mL), dried with anhydrous Na2SC>4, filtered and concentrated under reduced pressure. The residue w'as purified by silica gel chromatography (1/0 to 3/1 petroleum ether/EtOAc) to afford methyl 4-((2-((ieri-butoxycarbonyl)amino)ethyl)sulfonyl)- 3-fluoro-2-methylbenzoate (2.1 g, 58.2% yield) as a yellow solid. LCMS (ESI) m/z [M-56 + H] calcd for C 1 & H’iFNOeS: 320.12; found 320.1

Step 4: Synthesis of 4-((2-((i<eri-butoxycarbonyl)amino)ethyl)sulfonyl)-3-fluo ro- 2- methylbenzoic acid

[00480] To a solution of methyl 4-((2-((teri-butoxycarbonyl)amino)ethyl)sulfonyl)-3- iluoro-2-methylbenzoate (2.1 g, 5.59 mmol, 1.0 equiv) in THE (20 mL), MeOH (10 mL) and ¾G (10 mL) w ' as added LiOH^EbO (704.16 mg, 16.78 mmol, 3.0 equiv) at room temperature. The reaction mixture was stirred at 40 °C for 4 h. The mixture was then concentrated under reduced pressure to remove THE and MeOH. The aqueous phase was neutralized with 0.5N HCl and was then extracted with EtO Ac (5 x 20 mL). The combined organic phase was washed with brine (2 x 20 mL), dried with anhydrous Na 2 S0 4 , filtered and concentrated under reduced pressure to give A-((2-((tert- butoxycarbonyl)amino)ethyl)su3fonyl)-3-fliioiO-2-methylbenzo ic acid (2.01 g, 97.1% yield) as a white solid. LCMS (ESI) m/z [M-100 + H] calcd for C 15 H 20 FNO 0 S: 262.11; found 262.1.

Step 5: Synthesis of (4-(i<?rf-butoxycarbonyl)-2,3,4,5-tetrahydrobenzo[f][1,4] oxazepin-7- yllboronic acid [00481] To a solution of tert-hutyl 7-bromo-2,3-dihydrobenzo[f][1,4]oxazepine-4(5H)- carboxylate (4 g, 12.19 mmol, 1.0 equiv) in THF (80 mL) at -60 °C was added B(OiPr)3 (4.58 g, 24.38 mmol, 5.60 mL, 2.0 equiv) followed by dropwise addition of n-BuLi (2.5 M, 12.19 mL, 2.5 equiv) in «-hexane. The reaction was stirred at -65 °C for 1 li. The reaction mixture was quenched with IN HCl (12.25 mL) and allowed to warm to room temperature. The reaction mixture was extracted with EtOAc (3 x 30 mL), dried over anhydrous NaiSCL, filtered and concentrated under reduced pressure to give (4-(/e/7-butoxycarbonyl)-2, 3,4,5- tetrahydrobenzo[f][1,4]oxazepin-7-yl)boronic acid (3.5 g, crude) as light yellow oil, which was used to the next step directly. LCMS (ESI) m/z: [M-100 + H] calcd for C 1 4H 2 0BNO5: 194.15; found 194.2.

Step 6: Sythesis of re/t-butyl 7-(6-aminopyridm-3-yl)-2,3-diliydrobenzo[f][1,4] oxazepine- 4(5H)-carboxylate

[00482] To a solution of (4-(ier/-butoxycarbonyl)-2, 3,4,5- tetrahydrobenzo[f][1,4]oxazepin- 7-yl)boronic acid (4.2 g, 14.33 mmol, 1.0 equiv) in H 2 O (20 mL) and dioxane (60 mL) was added 5-bromopyridin-2-amine (2.48 g, 14.33 mmol, 1.0 equiv), Pd(dppf)Cl2*DCM (1.17 g, 1.43 mmol, 0.1 equiv) and EtaN (4.35 g, 42.99 mmol,

5.98 mL, 3.0 equiv) at room temperature. The mixture w'as stirred at 85 °C for 12 h. The mixture was then cooled to room temperature and the residue was poured into H 2 O (15 mL). The aqueous phase was extracted with EtOAc (3 x 40 mL) and the combined organic phase was washed with brine (2 x 40 mL), dried with anhydrous NaaSO- , filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (1/0 to 1/8 petroleum ether/EtO Ac) to afford /erf-butyl 7-(6-ammopyridin-3-yl)-2,3- dihydrobenzo[f][1,4]oxazepine-4(5H)-carhoxylate (3.3 g, 65.0% yield) as light yellow solid. LCMS (ESI) m/z: [M + H] calcd for C 1 9H 2 3N3O 3 : 342.18; found 342.2.

Step 7: Synthesis of 5-(2,3,4,5-tetrahydrobenzo[f][1,4]oxazepin-7-yl)pyridin-2-am ine

[00483] To a solution of tert-butyl 7-(6-aminopyridin-3-yl)-2,3-dihydrobenzo[f][1,4] oxazepine-4(5H)-carboxylate (3.3 g, 9.67 mmol, 1.0 equiv) in THF (40 mL) was added HCl in EtOAc (4 M, 100 mL, 41.38 equiv) at room temperature. The mixture was stirred for 3 h. The reaction mixture was filtered and the filter cake was washed with EtOAc (3 x 15 mL) and then dried under reduced pressure to give 5-(2,3,4,5-tetrahydrobenzo [f] [ 1 ,4]oxazepin-7- yl)pyridin-2-amine (3 g, 95.1% yield, 2HCl) as a light yellow solid. Step 8: Synthesis of ierf-butyl (2-((4-(7-(6-aminopyridin-3-yl)-2, 3,4,5- tetrahydrobenzo[f ] [ 1 ,4]oxazepine-4-carbonyl)-2-iluoro-3- meth y Iphen y 3) su l fony l)etb y 1 )carb amate

[00484] To a solution of 4-((2-((/e/t-butoxycarbonyl)amino)ethyl)sulfonyl)-3-fluoro-2 - methylbenzoic acid (690.08 mg, 1.91 mmol, 1.0 equiv) in DMF (10 mL) was added FlATU (1.09 g, 2.86 mmol, 1.5 equiv) and DIPEA (1.66 mL, 9.55 mmol, 5 equiv). The reaction was stirred at room temperature for 30 min and then 5-(2,3,4,5-tetrahydrobenzo[f][1,4]oxazepin- 7 -yl)pyridin-2-amine (0.6 g, 1.91 mmol, 1.0 equiv, 2HCl) was added. The mixture was stirred for 2 h, at which point H 2 O (40 mL) was added. The mixture was stirred for 5 min and the resulting precipitate was collected b filtration to give the crude product. The residue was purified by silica gel chromatography (1/0 to 10/1 EtOAc/MeOH) to afford /erf-butyl (2-((4- (7-(6-aminopyridin-3-yl)-2,3,4,5-tetrahydrobenzo[f][1,4] oxazepine- 4-carhonyl)-2-iluoro-3- methylphenyl)sulfonyl)ethyl)carbamate (0.538 g, 47.4% yield) as a light yellow solid. LCMS (ESI) m/z [M + H] calcd for C 2 9H 3 3FN4O6S: 585.22; found 585.3.

Step 9: Synthesis of (4-((2-aminoethyl)sulfonyl)-3-fluoro-2-methylphenyl)(7-(6- aminopyridin-3-yl)-2,3-dihydrobenzo[f] [ 1 ,4]oxazepin-4(5H)-yl)methanone 2,2,2- triiluoroacetate

[00485] A solution teri-butyl (2-((4-(7-(6-aminopyridin-3-yl)-2, 3,4,5- tetrahydrobenzoff] [ 1 ,4] oxazepine- 4-carbonyl)-2-fluoro-3- methylphenyl)sulfonyl)ethyl)carbamate (0.538 g, 920.20 μmol, 1.0 equiv) in TFA ( 10.35 L, 139.74 mmol, 151.85 equiv) was stirred at room temperature for 2 h. The solution was then concentrated under reduced pressure. The oily residue was triturated with MeCN (1 mL) and then dripped into MTBE (30 mL) for 10 min. The supernatant was removed and then the precipitate was collected by filtration under N:> to give (4-((2-aminoethyl)sulfonyl)-3-fluoro- 2-methylphenyl)(7-(6-aminopyridin-3-yl)-2,3-dihydrobenzo[f][ 1,4]oxazepin-4(5H)- yl)methanone 2,2,2-trifluoroacetate (0.50 g, 87.4% yield) as light brown solid. LCMS (ESI) m/z [M + t] calcd for C 2 4H 2 5FN4O4S: 485.17; found 485.1. Monomer A A. 5-(4-amino-1-(6-(piperazm-1-yl)pyrimidm-4-yl)-1H-pyrazolo[3, 4- d]pyrimidm-3-yl)benzo|d]oxazol-2-amhie trifluoroacetic add salt.

Step 1 : Synthesis of 1-(6-chloropyrixnidin-4-yl)-3-iodo-1H-pyrazolo[3,4-d]pyrimid in-4-amine

[00486] To a suspension of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (5 g, 19.16 mmol, 1.0 equiv) in DMF (60 mL) was added Nall (804.53 rng, 20.11 mmol, 60 wt.%, 1.05 equiv) at 0 °C. The mixture was stirred at 0 °C for 30 min. To the reaction mixture was then added 4,6-dichloropyrimidine (3.42 g, 22.99 mmol, 1.2 equiv) at 0 °C. The mixture was stirred at room temperature for 2.5 h, at which point the reaction mixture was added to H 2 O (600 mL). The suspension was then filtered to give the product (7.1 g, 99.2% yield) as yellow solid. LCMS (ESI) rn/z [M + H] calcd for C9H5CIIN7: 373.94; found 373.9.

Step 2: Synthesis of /eri-butyl 4-(6-(4-amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1- yl)pyrimidin-4-yl)piperazine-1-carboxylate

[00487] To a solution of 1-(6-chloropyrimidin-4-yl)-3-iodo-1H-pyrazolo[3,4-d]pyrimidi n- 4-amine (5 g, 13.39 mmol, 1.0 equiv) and n?rf-hutyl piperazine·· 1-carboxylate (2.99 g, 16.06 mmol, 1.2 equiv) in DMF (50 mL) w'as added K 2 CO 3 (3.70 g, 26.77 mmol, 2.0 equiv). The reaction mixture was stirred at 100 °C for 4 h, at which point it was added to H 2 O (500 mL). The suspension was then filtered to give the product (6.2 g, 88.5% yield) as yellow' solid. LCMS (ESI) rn/z: [M + H] calcd for C 1 SH 2 2IN9O 2 : 524.09; found 524.2.

Step 3: Synthesis of tert-butyl 4-(6-(4-amino-3-(2-aminobenzo[d]oxazol-5-yl)-1H- pyrazolo [3 ,4-d] pyrimidin-1- yl)pyrimidin-4-yl)piperazine-1-carboxylate [00488] To a bi-phasic suspension of 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)benzo[d]oxazol-2-amine (3.08 g, 11.85 mmol, 1.0 equiv), teri-butyl 4-(6-(4-amino-3-iodo- 1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyrimidin-4-yl)piperazine-1 -carboxylate (6.2 g, 11.85 mmol, 1.0 equiv) and Na2CO 3 (6.28 g, 59.24 mmol, 5.0 equiv) in H 2 O (100 mL ) and DME (200 mL) was added Pd(PPli3)4 (1.37 g, 1.18 mmol, 0.1 equi v) at room temperature under N 2 . The mixture was stirred at 110 °C for 24 h and then the mixture was filtered to give a solid cake. The solid was added to dioxane (20 mL) and stirred at 110 °C for 60 min, then filtered to give the product (3.5 g, 55.8% yield) as brown solid. LCMS (ESI) m/'z: [M + H] calcd for C 2 5H 2 7N11O 3 : 530.24; found 530.3.

Step 4: Synthesis of 5-(4-amino-1-(6-(piperazin-1-yl)pyrimidin-4-yl)-1H-pyrazolo[ 3,4- d]pyrimidin-3-yl)benzo[d]oxazol-2-amine trifluoroacetic acid salt

[00489] A solution of tert-hutyl 4-(6-(4-amino-3-(2-aminobenzo[d |oxazol-5-yl)-iH- pyrazolo[3,4-d]pyrimidin-1-y])pyrimidin-4-yl)piperazine-1-ca rboxylate (3.5 g, 6.61 mmol,

1.0 equiv) in TFA (35 mL) was stirred at room temperature for 1 h. The reaction solution was concentrated under reduced pressure and the resulting crude material was dissolved in MeCN (20 mL) and added dropwise to MTBE (500 mL). The resulting solid was then filtered to give the product (5.5 g, 91.9% yield) as brown solid. LCMS (ESI) m/z: [M + H] calcd for C 20 H 19 N 11 O: 430.19; found 430.1.

Monomer AB. 8-(6-methoxypyridin-3-yl)-3-methyl-1-(4-(4-(5, 6,7,8- tetrahydropyrido[4,3-d]pyrimidm-2-yl)piperazin-1-yl)-3-(trif luoroinethyl)phenyI)-1H- imidazo[4,5-c]qumolm-2(3H)-one trifluoroacetic add salt.

Step I : Synthesis of teri-butyl 2-(4-(4-(8-(6-methoxypyridin-3-yl)-3-methyl-2-oxo-2,3- dihydro-1H-irnidazo[4,5-c]quinolin-1-yl)-2-(trifluoromethyl) phenyl)piperazin-1-yl)-7,8- dihydropyrido[4,3-d]pyrimidine-6(5H)-carboxylate

100490] To a mixture of 8-(6-methoxypyridin-3-yl)-3-methyl-1-(4-(piperazin-1-yl)-3- (trifluoromethyl)phenyl)-1H-imidazo[4,5-c]quinolin-2(3H)-one (0.3 g, 561.24 μmol, 1.0 equiv) and ierf-butyl 2-chloro-7,8-dihydropyrido[4,3-d]pyrimidine-6(5H)-carboxylat e (151.38 mg, 561.24 mihoΐ, 1.0 equiv) in DMF (5 mL) was added K 2 CO 3 (193.92 mg, 1.40 mmol, 2.5 equiv). The mixture was stirred at 100 °C for 14 h, at which point H 2 O (20 mL) was added. The aqueous layer was extracted with EtOAc (3 x 40 mL) and the combined organic layers were concentrated under reduced pressure. The crude material was purified by column chromatography (30/1 to 15/1 DCM/MeOH) to give the product (0.30 g, 69.6% yield) as a light-yellow solid. LCMS (ESI) m/z: [M + H] calcd for C 4 0H40F3N9O4: 768.33; found 768.5.

Step 2: Synthesis of 8-(6-methoxypyridin-3-yl)-3-metiiyl-1-(4-(4-(5, 6,7,8- tetrabydropyrido[4,3-d]pyrknidin-2-yl)piperazin-1-yl)-3-(tri fluoromethyl)phenyl)-1H- imidazo[4,5-c]quinolin-2(3H)-one

[00491] A solution of iert-butyl 2-(4-(4-(8-(6-methoxypyridin-3-yl)-3-methyl-2-oxo-2,3- dihydro-1H-imidazo[4,5-c]quinolin-1-yl)-2-(trifluorornethyl) phenyl)piperazin-1-yl)-7,8- dihydropyrido[4,3-d]pyrimidine-6(5H)-carboxylate (0.8 g, 1.04 mmol, 1.0 equiv) in TFA (8 mL) was stirred at room temperature for 2 h. The solvent was concentrated and the residue was dissolved in MeCN (5 mL), then the solution was added dropwise to MTBE (150 mL). The precipitate was filtered and the solid was dried under reduced pressure to give the product (600 mg, 70.6% yield) as a yellow solid. LCMS (ESI) m/z : [M + H] calcd for C 3 5H 3 2F3N9O 2 : 668.27; found 668.3.

Monomer AC. 5-(4-amino-1-(piperidin-4-ylmethyl)-1H-pyrazolo[3,4-d]pyrimi dm-3- yl)benzo[d]oxazol-2-amine trifluoroacetic add salt. Step 1: Synthesis of ten-butyl 4-((metliylsulfonyl)oxy)piperidine-1-carboxylate

[00492] To a solution of tert-\miy\ 4-hydroxypiperidine-1-carboxy late (4 g 19.87 mmol, 1.0 equiv) and EtaN (3.87 mL , 27.82 mmol, 1.4 equiv) in DCM (40 mL) was added MsCl (2.15 mL, 27.82 mmol, 1.4 equiv) at 0 °C. Then the reaction mixture was stirred at room temperature for 1 h. HbO (50 mL) was added and the aqueous phase was extracted with DCM (3 x 50 mL). The combined organic phase was washed with brine, dried with anhydrous NaiSO , filtered and concentrated under reduced pressure to give the product (5.62 g, 101% crude yield) as yellow solid which was used directly in the next step.

Step 2: Synthesis of tert-butyl 4-(4-amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1- yl)piperidine-1-carboxyl ate

[00493] To a suspension of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (5 g, 19.16 mmol, 1.0 equiv) and ien-butyl 4-((methylsulfonyl)oxy)piperidine-1-carboxylate (5.62 g, 20.11 mmol, 1.05 equiv) in DMF (100 mL) was added K 2 CO 3 (5.29 g, 38.31 mmol, 2.0 equiv). The mixture was stirred at 80 °C for 12 h. The reaction mixture was then added to H 2 O (400 mL) at 0 °C. The resulting precipitate was filtered to give the product (5.0 g, 58.8% yield) as yellow solid. LCMS (ESI) rn/z: [M + H] calcd for CisEbilNeC : 445.09; found 445.1.

Step 3: Synthesis of ten-butyl 4-(4-amino-3-(2-aminobenzo[d]oxazol-5-yl)-1H-pyrazolo[3,4- d]pyrimidin-1-yl)piperidine-1-carboxy late

[00494] To a suspension of ten-butyl 4-(4-amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1- yl)piperidine-1-carboxy late (5 g, 11.25 mmol, 1.0 equiv), 5-(4,4,5,5-tetramethyI-L3,2- dioxaboroIan-2-yI)benzo[d]oxazoI-2-amine (3.51 g, 13.51 mmol, 1.2 equiv) and Na2CC>3 (5.96 g, 56.27 mmol, 5.0 equiv) in H 2 O (50 mL) and DME (100 mL) was added Pd(PPhi) 4 (1.30 g, 1.13 mmol, 0.1 equiv) at room temperature under N 2 . The mixture was stirred at 110 °C for 3 h. The reaction mixture was then cooled to room temperature and filtered. The filtrate was partitioned between EtOAc (100 L) and I¾0 (100 mL) and then the aqueous layer was separated and extracted with EtOAc (3 x 100 mL). The combined organic layer was washed with brine (20 mL) and dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. The residue was triturated with EtOAc (30 mL) and filtered to give the product (3.6 g, 71% yield) as yellow solid. LCMS (ESI) ni/z: [M + H] calcd for C 2 2H 2 6N8O 3 : 451.22; found 451.3. Step 4: Synthesis of 5-(4-amino-1-(piperidin-4-yl)-1H-pyrazolo[3,4-d]pyrimidin-3- yl)benzo[d]oxazol-2-amine trifiuoroacetic acid salt

[00495] A solution of tert-hutyl 4-(4-amino-3-(2-aminobenzo[d]oxazol-5-yl)-1H- pyrazoio[3,4-d]pyrimidin-1-yl)piperidine-1-carboxylate (1.4 g, 3.11 mmol, 1.0 equiv) in TEA (10 mL) was stirred at room temperature for 30 min. The reaction solution w'as concentrated under reduced pressure and the crude solid was dissolved in MeCN (20 mL). The solution was added dropwise to MTBE (100 mL) and the resulting solid was filtered to give the product (1.6 g, 85.8% yield) as yellow solid. LCMS (ESI) m/z: [M + H] calcd for CnHisNsO 3 : 351.17; found 351.1.

Monomer AD, 1-(piperidm-4-yI)-3-(1H-pyrrolo[2,3-b]pyridin-S-yl)-1H-pyraz olo[3,4- d]pyrimidin-4-amine trifluoroacetic add salt.

Step 1: Synthesis of ten-butyl 4-(4-amino-3-(1H-pyrrolo[2,3-b]pyridin-5-yl)-1H- pyrazolo[ ,4-d]pyrimidin-1-yl)piperidine-1-carhoxylate

[00496] To a suspension of 5-(4,4,5-trimethyl-1,3,2-dioxaborolan-2-yl)-1H-pynOlo[2,3- h]pyridine (857.12 mg, 3.51 mmol, 1.2 equiv), fe/t-hutyl 4-(4-amino-3-iodo-1H- pyrazolo[3,4-d]pyrimidin-1-yl)piperidine-1-carboxylate (1.3 g, 2.93 mmol, 1.0 equiv) and NaiCO 3 (1.55 g, 14.63 mmol, 5.0 equiv) in DME (20 mL) and H 2 O (10 mL ) was added PdiPPlivk (338.13 mg, 292.62 μmol, 0.1 equiv) at room temperature under N2. The mixture was stirred at 110 °C for 3 li. The reaction mixture was then cooled to room temperature and filtered. The filtrate was partitioned between EtOAc (50 mL) and H 2 O (50 mL) and the aqueous layer was separated and extracted with EtOAc (3 x 50 mL). The combined organic layer were washed with brine, dried over anhydrous Na .' SO:. filtered, and concentrated under reduced pressure. The residue was triturated with EtOAc (10 mL), filtered, the solid cake was dried under reduced pressure to give the product (1.0 g, 78.7% yield) as yellow' solid. Step 2: Synthesis of 1-(piperidin-4-yI)-3-(1H-pynOlo[2,3-b]pyridin-5-yl)-1H-pyraz oio[3,4- d]pyrimidin-4-amine trifiuoroacetic acid salt

[00497] A solution of tert-hutyl 4-(4-ammo-3-(1H-pyrrolo[2,3-b]pyridin-5-yl)-1H- pyrazoio[3,4-d]pyrimidin-1-yl)piperidine-1-carboxylate (1.5 g, 3.45 mmol, 1.0 equiv) in TFA (10 ml.,) was stirred at room temperature for 30 min. The reaction solution was concentrated under reduced pressure and the crude residue was dissolved in MeCN (20 mL). The solution was added dropwise to MTBE (100 mL) and the resulting solid was filtered to give the product (1.19 g, 74.2% yield) as light yellow solid. LCMS (ESI) rn/z: [M + H] calcd for CnHigNs: 335.18; found 335.1.

Monomer AE. (4-((2-aminoethyl)sulfbnyl)-2-inethylphenyl)(7-(6-aiiimopyri dm-3-yl)-2,3- dihydrohenzo[f][1,4]oxazepin-4(5H)-yI)methanone.

Me O HS ' V ^~ NHBOC

Me O

CH 3 I, K,C0 3 K2C ° f^ On 3

DMF MF 110 c

Step 1: Synthesis of methyl 4-fluoro-2-methylbenzoate

[00498] To a solution of 4-fluoro-2-methylbenzoic acid (86 g, 557.94 mmol, 1.0 equiv) in DMF (900 mL) was added K 2 CO 3 (231.33 g, 1.67 mol, 3.0 equiv) and iodomethane (79.19 g, 557.94 mmol, 34.73 mL, 1.0 equiv). The mixture was stirred at room temperature for 1 h.

The solution of methyl 4-fluoro-2-meihyibenzoate in DMF (900 mL) was used directly in the next step.

Step 2: Synthesis of methyl 4-((2-((ieri-butoxycarbonyl)amino)ethyr)thio)-2-methylbenzoa te [00499] To a solution of methyl 4-fluoro-2-methylbenzoate (93.8 g, 557.94 mmol, 1.0 equiv) in DMF (900 rnL) was added ZerZ-butyl (2-mercaptoethyl)carbamate (98.91 g, 557.97 mmol, 1.0 equiv) and K 2 CO 3 (154.23 g, 1.12 mol, 2.0 equiv). The reaction was stirred at 110 °C for 12 h, at which point the mixture was cooled to room temperature and added to H 2 O (1000 niL). The aqueous layer was then extracted with EtOAc (3 x 600 ml.) and the combined organic layers were washed with brine, dried, and concentrated under reduced pressure. Purification by silica gel chromatography (0→ 25% EtOAc/petroleum ether) afforded the desired product as a colorless oil (144 g 79% yield).

Step 3: Synthesis of methyl 4-((2-((ieri-butoxycarbonyl)amino)ethyl)sulfonyl)-2- methylbenzoate

[00500] To two separate batches containing a solution of methyl 4-((2 -({tert- butoxycarbonyl)amino)ethyl)thio)-2-methylbenzoate (72 g, 221.25 mmol, 1.0 equiv), NaOH (2 M, 110.6 rnL, 1.0 equiv), and NaHCOs (55.76 g, 663.75 mmol, .3.0 equiv) in acetone (750 rnL) was added potassium peroxymonosulfate (284.28 g, 462.41 mmol, 2.1 equiv). The mixture was stirred for 12 h at room temperature, at which point the two batches were combined and then the mixture was acidified to pH 5 by addition of IN HCl. The aqueous layer was extracted with EtOAc (3 x 1500 ml.) and the combined organic phases were washed with brine (2 x 500 mL), dried, and concentrated under reduced pressure. Purification by silica gel chromatography (Q-->25% EtOAc/petroleum ether) afforded the desired product as a white solid (120 g, 76% yield).

Step 4: Synthesis of 4-((2-((ieri-butoxycarbonyl)amino)ethyI)sulfonyl)-2-methylbe nzoic acid

[00501] To a solution of methyl 4-((2-((ieri-butoxycarbonyl)amino)ethyl)sulfonyl)-2- methylbenzoate (35 g, 97.92 mmol, 1.0 equiv) in THE (200 mL), MeOH (100 rnL) and H 2 O (100 mL) was added LiOH ® H 2 O (12.33 g 293.77 mmol, 3.0 equiv) at room temperature. The reaction mixture was stirred at 40 °C for 1 h. The mixture was then concentrated under reduced pressure to remove THE and MeOH. The aqueous phase was neutralized with 0.5N HCl and the resulting precipitate was isolated by filtration. The solid cake was washed with H 2 O (3 x 20 mL) to afford the desired product as a white solid (25 g, 74% yield).

Step 5: Synthesis of tert-butyl (2-((4-(7-(6-aminopyridin-3-yl)-2,3,4,5- tetrahydrobenzo[f][1,4]oxazepine-4-carbonyl)-3-methylphenyl) sulfonyl)ethyl)cai¾amate

[00502] To a solution of 4-((2-((ieri-butoxycarbonyl)amino)ethyl)sulfonyl)-2- methylbenzoic acid (9.7 g, 28.25 mmol, 1.0 equiv) and 5-(2,3,4,5- tetra ydrobenzo[f][1,4]oxazepin-7-yl)pyridin- 2-amine (8.88 g, 28.25 mmol, 1.0 equiv, 2HCl) in DMF (120 mL) was added HATH (16.11 g, 42.37 mmol, 1.5 equiv) and DIPEA (18.25 g, 141.24 mmol, 24.60 mL, 5.0 equiv). The reaction was stirred at room temperature for 1 h, at which point the reaction mixture was poured into H 2 O (1000 mL) The mixture was stirred for 5 mi and the resulting precipitate was collected by filtration to give the crude product. The crude product was triturated with EtOAc (100 mL), filtered, and the solid cake was dried under reduced pressure to afford the desired product as a white solid (14 g, 87% yield).

Step 6: Synthesis of (4-((2-aminoethyl)sulfonyl)-2-methylphenyl)(7-(6-aminopyridi n-3-yl)- 2,3-diliydrobenzo[f][1,4]oxazepin-4(5H)-yl)methanone

[00503] A solution /urt-butyl (2-((4-(7-(6-aminopyridin-3-yl)-2,3,4,5- tetraliydrobenzo[f] [ 1 ,4] oxazepine -4- carbonyl) -3 -methylphenyl)sulfonyl)ethyl)carbamate ( 19 g, 33.53 mmol, 1.0 equiv) in TF.A (100 mL) was stirred at room temperature for 30 min. The solution was then concentrated under reduced pressure. The residue was triturated with MeCN (30 mL) and then dripped into MTBE (600 mL) and stirred for 20 min. The suspension was filtered and the resulting solid was dissolved in MeCN (30 mL) and concentrated under reduced pressure to afford the desired product as a light yellow solid (24 g, TEA salt). LCMS (ESI) m/z: [M + H] calcd for C 2 4H 2 6N4O4S :467.18; found 467.1.

Monomer AF. 5-(4-amino-1-((5,6,7,8-tetrahydropyrido[4,3-d]pyrimidm-2-yl) methyl)- 1H-pyrazolo[3,4-d]pyrimidm-3-yl)benzo[d]oxazol-2-amine.

Step i: Synthesis of (Z)-te rt-butyl 3-((dimethylamino)met!iylene)-4-oxopiperidine-1- carboxylate

[00504] A solution of tert-butyl 4-oxopiperidine-1-carboxylate (15 g, 75.28 mmol, 1.0 equiv) and 1,1-dimethoxy-/V,iV-dimethylmethanamine (11.00 mL, 82.81 mmol, 1.1 equiv) in DMF (105 mL) was stirred at 95 C for 12 h. The reaction mixture was then concentrated under reduced pressure and the resulting residue was dissolved in EtOAc (30 ml,) and washed with brine (3 x 30 mL). The aqueous phase was extracted with EtOAc (50 mL), and the combined organic phases were dried and concentrated under reduced pressure to afford the desired product as a yellow solid (10.1 g, 53% yield).

Step 2: Synthesis of ten-butyl 2-(hydroxymethyl)-7,8-dihydropyrido[4,3-d]pyrimidine- 6(5H)-carboxylate

[00505] To a solution of NaOEt (1.98 g, 29.10 m ol, 1.0 equiv) in EtOH (70 mL) was added (Z)-/m-huiyl 3-((dimethylamino)methylene)-4-oxopiperidine-1-carboxylate (7.4 g, 29.10 mmol, 1.0 equiv) and 2-hydroxyacetimidamide hydrochloride (3.54 g, 32.01 mmol, 1.1 equiv) The reaction mixture was heated to 90 °C for 12 h, at which point the mixture was cooled to room temperature and concentrated under reduced pressure. The residue was partitioned with EtOAc (40 mL) and washed with sat. NaHC(>3 (40 mL). The aqueous phase was extracted with EtOAc (3 x 20 mL) and the combined organic phases were washed with brine (2 x 50 mL), dried, and concentrated under reduced pressure. Purification b silica gel chromatography (25% EtOAc/petroleum ether) afforded the desired product as a yellow solid (7.24 g 94% yield).

Step 3: Synthesis of tert-butyl 2-(bromomethyl)-7,8-dihydropyrido[4,3-d]pyrimidine-6(5H)- carboxylate

[00506] To a solution of t -butyl 2-(hydroxymetbyl)-7,8-dihydropyrido[4,3- d]pyrimidine-6(5H)-carboxylate (6.24 g, 23.52 mmol, 1.0 equiv) and PPI13 (12.34 g, 47.04 mmol, 2.0 equiv) in DCM (140 mL) was added CBI4 (14.82 g, 44.69 mmol, 1.9 equiv). The mixture was stirred at room temperature for 3 h, at which point mixture was concentrated under reduced pressure. The residue was partitioned between EtOAc (20 mL) and H 2 O (20 mL), the aqueous phase was extracted with EtOAc (3 x 20 mL). The combined organic phases were washed with brine (2 x 50 mL), dried, and concentrated under reduced pressure. Purification by silica gel chromatography (14% EtOAc/petroleum ether) afforded the desired product as a yellow solid (3.6 g, 47% yield).

Step 4: Synthesis of tert-bu l 2-((4-amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1- yl)methyl)-7,8-dihydropyrido[4,3-d]pyrimidine-6(5H)-carboxyl ate

[00507] To a solution of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (1.59 g, 6.09 mmol, 1.0 equiv) in DMF (15 mL) was added NaH (243.73 mg, 6.09 mmol, 60 wt.%, 1.0 equiv) at 0 °C. The suspension was stirred for 30 min and then ler/-huiyl 2-(bromomethyl)-7,8- dihydropyrido[4,3-d]pyrirmdine-6(5H)-carboxylate (2.2 g, 6.70 mmol, 1.1 equiv) was added. The reaction mixture was warmed to room temperature and stirred for 3 h. The mixture was poured into HeO at 0 °C and the precipitate was collected by filtration to afford the desired product as a brown solid (2.5 g, 66% yield).

Step 5: Synthesis of tert-butyl 2-((4-amino-3-(2-aminobenzo[d]oxazol-5-yl)-1H- pyrazolo[3,4-d]pyrimidin-i-yl)metliyl)-7,8-dihydropyrido[4,3 -d]pyrimidine-6(5H)- carhoxylate

[00508] To a solution of feri-butyl 2-((4-amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1- yl)methyl)-7,8-dihydropyrido[4,3-d ]pyrirmdme-6(5H)-carboxylate (4.55 g, 8.95 mmol, 1.0 equiv), 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzo[d]oxazo l-2-amine (2.79 g, 10.74 mmol, 1.2 equiv) and NaaCOi (4.74 g, 44.76 mmol, 5.0 equiv) in dioxane (70 mL) and H 2 O (35 mL) was added Pd(PPlt3)4 (1.03 g, 895.11 μmol, 0.1 equiv). The reaction mixture was heated to 110 °C for 3 h, at which point the mixture was cooled to room temperature and poured into H 2 O at 0 °C. The precipitate was filtered, and the solid cake was dried under reduced pressure. The crude product was washed with EtOAc (50 mL) to afford the desired product as light yellow' solid (3.14 g, 68% yield).

Step 6: Synthesis of 5-(4-amino-1-((5,6,7, 8-tetrahydropyrido[4, 3-d]pyrimidin-2-yl)methyl)- 1H-pyrazolo [3 ,4-d]pyrimidin-3 -yl)benzo [d] oxazol-2-amine

[00509] A solution of /art-butyl 2-((4-amino-3-(2-aminobenzo[d]oxazoI-5-yl)-1H- pyrazolo[3,4-d]pyrimidin-1-yl)methyl)-7,8-dihydropyrido[4,3- d]pyrimidine-6(5H)- carhoxylate (3.14 g, 6.10 mmol, 1.0 equiv) in TFA (20 mL) was stirred at room temperature for 30 min. The mixture was concentrated under reduced pressure and the resulting residue was added dissolved in MeCN (7 mL) and added to MTBE (700 mL). The precipitate was collected by filtration to afford the desired product as a brown solid (4.25 g, 92% yield, 3 TEA). LCMS (ESI) m/z: [M + H] calcd for ChoHigNioO: 415.18; found 415.1. i r '> -> Monomer AG. 5-(4-amino-1-((2-((2-aminoethyI)sulfonyl)-1,2,3,4-tetrahydro isoquinolin- 6-yl)methyl)-1H-pyrazolo|3,4-d]pyrimidm-3-yl)benzo[d]oxazol- 2-amhie.

Step 1 : Synthesis of iV-Boc taurine tetrabutylammonium salt

[00510] To a solution of 2-aminoethanesulfonic acid (10.00 mL, 79.91 mmol, 1.0 equiv) in THF (60 mL) and aqueous NaOH (2 M, 40 mL, 1.0 equiv) was added B0C 2 O (18.31 g, 83.90 mmol, 1.05 equiv). The mixture was stirred at room temperature for 15 h, at which point the mixture was extracted with EtOAc (10 mL). The aqueous phase was diluted with H 2 O (450 mL), treated with LiOH ® H 2 O (3.35 g, 79.83 mmol, 1.0 equiv) and HBVUNHSCL (27.13 g 79.90 mmol, 1.0 equiv) and stirred for 30 min. This mixture was extracted with DCM (3 x 80 mL), and the combined organic phases were dried and concentrated under reduced pressure to afford the desired product as a colorless oil (34.26 g, 91% yield).

Step 2: Synthesis of tert-buty\ (2-(chlorosulfonyl)etliyl)carbamate

[00511] To a solution of N-Boc taurine tetrabutylammonium salt (4.7 g, 10.05 mmol, 1.0 equiv) in DCM (42 mL) was added DMF (77.32 μL, 1.00 mmol, 0.1 equiv) followed by a solution of triphosgene (0.5 M, 8.04 mL, 0.4 equiv) in DCM at 0 °C. The mixture was warmed to room temperature and stirred for 30 min. The solution of tert-butyl (2- (chiorosulfonyI)ethyl)carbamate (2.45 g, crude) in DCM was used directly in the next step.

Step 3: Synthesis of tert-butyl (2-((6-((4-amino-3-(2-aminobenzo[d]oxazol-5-yl)-1H- pyrazolo[3,4-d]pyrimidin-1-yl)methyl)-3,4-dihydroisoquinolin -2(1H)- yl)sulfonyl)ethyl)carbamate i r ' > , t [00512] To a solution of 5-(4- amino-1- (( 1 ,2,3 ,4-tetrahydroisoquinolin-6-y l)methyl)-1H- pyrazolo[3,4-d]pyrimidin-3-yl)benzo[d]oxazol-2-amine (6.04 g, 9.44 mmol, 1.0 equiv, 2TFA) in DMF (40 mL) was added 1¾N (7.88 mL, 56.63 mmol, 6.0 equiv). A solution of tert-\mty\ (2-(chlorosulfonyl)ethyl)carbamate in DCM (42 mL) at 0 °C was added. The mixture was warmed to room temperature and stirred 16 h. The reaction mixture was concentrated under reduced pressure to remove DCM and the resulting solution was purified by reverse phase chromatography (15-->45% MeCN/EbO) to afford the desired product as a white solid (5.8 g, 83% yield, TFA). LCMS (ESI) m/z: [M + H] calcd for C 2 9H 3 3N9O5S: 620.24; found 620.3.

Step 4: Synthesis of 5-(4-amino-1-((2-((2-aminoethy l)sulfonyl)-1 ,2,3,4- tetraliydroisoquinolin-6-y limethyl)-1H-pyrazolo [3 ,4-d]pyrimidin-3 -yl)benzo [d] oxazol-2- amine

[00513] A solution of tert-butyi (2-((6-((4-aniino-3-(2-aniinohenzo[d]oxazoI-5-yl)-1H- pyrazolo[3,4-d]pyrimidm-1-yl)methyl)-3,4-dihydroisoquinolm-2 (IH)- yl)sulfonyl)ethyl)carbamate (5.8 g, 9.36 mmol, 1.0 equiv) in TFA (48 mL) was stirred at room temperature for 0.5 h, at which point the reaction mixture was concentrated under reduced pressure. The crude product dissolved in MeCN (30 mL) and was added dropwise into MTBE (200 mL). The mixture was stirred for 5 min and filtered, the filter cake was dried under reduced pressure to afford the desired product as a yellow solid (3.6 g, 62% yield, 2.2TFA). LCMS (ESI) m/z: [M + H] calcd for C 2 4H 2 5N9O 3 S: 520.19; found 520.1.

Monomer AH. feri-butyl ((5-((4-amino-3-(2-amino benzo[d]oxazol-5-yl)-1H- pyrazoio[3,4-d]pyrimidin-1-yl)methyl)pyriniidin-2-yl)methyl) carbamaie.

Step 1: Synthesis of (2-(((/i ? r/-butoxycarbonyl)amino)methyl)pyrimidin-5-yl)methyl methanes u Ifon a te

[00514] To a solution of tert-butyl ((5-(hydroxymethyl)pyrirnidin-2-yl)methyl)carbamate (42 g, 17.55 mmol, 1.0 equiv) in DCM (42 mL ) at 0 °C was added EtsN (7.33 mL, 52.66 mmol, 3.0 equiv) followed by MsCl (2.41 g, 21.06 mmol, 1.63 mL, 1.2 equiv). The mixture was stirred at 0 °C for 10 min, and then H 2 O (15 mL) was added. The reaction mixture was extracted with DCM (5 10 mL) and the combined organic phases were washed with brine (5 mL), dried, filtered, and concentrated under reduced pressure to afford the desired product (5 5 g, 98.7% yield) as a colorless solid.

Step 2: Synthesis of teri-butyl ((5-((4-amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1- yl)methyl)pyrimidin-2-yl)methyl)carbamate

[00515] To a solution of (2-(((ieri-butoxycarbonyl)amino)methyl)pyrimidin-5-yl)methyl ethanesulfonate (5.47 g, 17.24 mmol, 1.2 equiv) and 3-iodo-1H-pyrazolo[3,4-d] pyrimidin- 4- amine (3.75 g, 14.37 mmol, 1.0 equiv) in DMF (55 mL) at room temperature was added K 2 CO 3 (5.96 g, 43.10 mmol, 3 equiv). The mixture was slimed at 80 °C for 5 h, at which point H 2 O (100 mL) and brine (20 mL) were poured into the reaction mixture. The solution was extracted with EtOAc (10 x 30 mL) and the combined organic phases were dried, fi ltered, and concentrated under reduced pressure. Purification by silica gel chromatography (Q→ 30% EtOAc/MeOH) afforded the desired product (2 g, 28.9% yield) as a yellow' solid. r '7 Step 3: Synthesis of ie/f-butyl ((5-((4-amino-3-(2-aminobenzo[d]oxazol-5-yl)-1H- pyrazolo[3,4-d]pyrimidin-1-yl)methyl)pyrimidin-2-yl)metbyl)e arbamate

[00516] To a solution of tert-buiyl ((5-((4-amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1- yl)methyl)pyrimidin-2-yl)methyl)carbamate (2 g, 4 15 mmol, 1.0 equiv), 5-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3-benzoxazol-2-amine (1 13 g, 4.35 mmol, 1.05 equiv) and NaaCOa (688.39 mg, 8.29 mmol 2.0 equiv) in dioxane (20 mL ) and H 2 O (10 mL) was added Pd(PPli3)4 (479.21 mg, 414.70 mhioΐ, 0.1 equiv). The mixture was stirred at 110 °C for 1 b, at which time the mixture was cooled to room temperature, filtered, and the solid cake washed with MeOH (3 x 10 mL). The filtrate was concentrated under reduced pressure to remove MeOH and then added dropwise into H 2 O (50 mL). The resulting suspension was filtered, and the filter cake was washed with H 2 O (3 x 10 mL). The solid cake w'as stirred in MeOH (20 mL) for 30 min. The resulting suspension was filtered, and the filter calve washed with MeOH (3 x 8 mL). The filter cake was dried under reduced pressure to afford the desired product (1.03 g, 48.9% yield) as a white solid. LCM8 (ESI) n / 'z [M + H] caicd for C 2 3H 2 4N10O 3 : 489.21; found 489.2.

Step 4: Synthesis of 5-(4-amino-1-{[2-(aminomethyl)pyrimidin-5-yl]methyl}-1H- pyrazolo[3,4-d]pyrimidin-3-yl)-1,3-benzoxazol-2-amine

[00517] To tert-butyl ((5-((4-amino-3-(2-aminobenzo[d]oxazol-5-yl)-TH-pyrazolo[3,4 - d]pyrimidin-1-yl)methyl)pyrimidin-2-yl)methyl)carbamate (100 mg, 0.205 mmol, 1.0 equiv) was added con. HC 3 (850 μL, 10.2 mmol, 50 equiv). The reaction was stirred for 1 h and was then poured into acetone (3 mL) The resulting precipitate was filtered, washed with acetone, and dried under reduced pressure to afford the desired product (80 mg, 92% yield) as a brown solid. LCMS (ESI) m/z: [M + H] caicd for CisHieNioO: 389.16; found 389.0.

Monomer AI. 5-(4-(dimeihylamino)-1-((1,2,3,4-tetrahydroisoquinolin-6-yl) methyl)-1H- pyrazolo[3,4-d]pyrimidm-3-yl)benzo[d]oxazol-2-ainme trifluoroacetic add salt.

Step 1: Synthesis of m/t-butyl 6-((4-(dimethylamino)-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-1- yl)methyl)-3,4-dihydroisoquinoline-2(1H)-carboxylate [00518] To a solution of 3-iodo-/V,iV-dimetliyI-1H-pyrazolo[3,4-d]pyrimidin-4-amine (3.6 g, 12.45 mmol, 1.0 equiv) in DMF (36 mL ) at 0 °C was added Nall (523.00 mg, 13.08 mmol, 60 wt.%, 1.05 equiv). The mixture was stirred at 0 °C for 30 min. To the reaction mixture was then added a solution of fe/ -butyl 6-(bromomethyl)-3,4-dihydroisoquinoline-2(1H)- carboxylate (4.47 g, 13.70 mol, 1.1 equiv) in DMF (18 mSL) at 0 °C. The mixture was stirred at room temperature for 2 h. The reaction mixture was then added to cold H 2 O (200 mL) and slimed for 30 min. The resulting precipitate was collected by filtration to afford the desired product (6 g, 71.9% yield) as a white solid.

Step 2: Synthesis of tert-butyl 6-((3-(2-aminobenzo[d]oxazol-5-yl)-4-(dimethylamino)-1H- pyrazolo[3,4-d]pyrimidin-1-yl)methyl)-3,4-dibydroisoquinolin e-2(1H)-carboxylate

[00519] To a solution of tert-butyl 6-((4-(dimethylamino)-3-iodo-1H-pyrazolo[3,4- d]pyrimidin-1-yl)methyl)-3,4-dihydroisoquinolme-2(lH)-carbox ylate (2 g, 2.96 mmol, 1.0 equiv) and 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolajn-2-yl)benzo[d]oxaz ol-2-amine (922.81 mg, 3.55 mmol, 1.2 equiv) in dioxane (24 mL) and H 2 O (12 mL) was added NaiCCb (1.57 g, 14.78 mmol, 5.0 equiv) and Pd(PPli3)4 (341.66 mg, 295.66 μmol, 0.1 equiv). The mixture was stirred at 110 °C for 12 h. The reaction mixture was then poured into cold H 2 O (200 mL) and stirred for 30 min. The resulting precipitate was collected by filtration. Purification by silica gel chromatography (5— 100% petroleum etlier/EtOAe) afforded the desired product (1.2 g, 72.3% yield) as a yellow' solid.

Step 3: Synthesis of 5-(4-(dimethylamino)-1-((1,2,3, 4-tetrahydroisoquinolin-6-yl)methyl)- 1H-pyrazolo [3 ,4-d]pyrimidin-3 -yl) benzo [d] oxazol-2- amine

[00520] A solution of tert-butyl 6-((3-(2-aminobenzo[d]oxazol-5-yl)-4-(dimethylamino)- lH-pyrazolo[3,4-d]pyrimidin-1-yl)methyl)-3,4-dihydroisoquino line-2(1H)-carboxylate (1.7 g, 3.14 mmol, 1.0 equiv) in TFA (10 mL) w'as stirred at room temperature for 30 min. The reaction mixture was then concentrated under reduced pressure. The residue was added to MeCN (10 mL) and the solution was added dropwise into M’TBE (200 mL). The resulting solid was dissolved in MeCN (30 mL) and the solution was concentrated under reduced pressure to afford the desired product (1.67 g, 92.9% yield,) as a yellow' solid. LCMS (ESI) m/z: [M + H] calcd for C 2 4H 2 4N8O: 441.22; found 441.2.

1 o Monomer AJ. 4-amino-5-(2-aminobenzo[d]oxazol-5-yl)-5H-pyrimido[5,4-b]mdo le-7- carboxylic add.

[00521] This monomer can be prepared from 7-methyl-5H-pyrimido[5,4-b]indol-4-ol by benzylic oxidation to the carboxylic acid, conversion to the ethyl ester, followed by O- ethylation with triethyloxonium tetrafluoroboroate. Palladium -mediated aiylation followed by ester hydrolysis and final ammonia-olysis provides the monomer.

Monomer AK. 4-amino-5-(2-aminobenzo[d]oxazo-5-yl)-5H-pyrimido[5,4-b]indo le-8- carboxylic add. [00522] This monomer can be prepared following a similar route as that to prepare the previous monomer, but using the isomeric starting material from 8-methyl-5H-pyrimido[5,4- b]indol-4-ol. Benzylic oxidation to the carboxylic acid, conversion to the ethyl ester, followed by O-ethylation with triethyloxonium tetrafluoroboroate and palladium-mediated arylation, followed by ester hydrolysis and final ammonia-olysis provides the monomer.

Monomer AL. 3-(2,4-bis((S) -3-methylmorphoiino)-4a,8a-dihydropyrido[2,3- d]pyrimidin-7-yl)benzoic add.

Step 1: Synthesis of (35')-4-[7-chioro-2-[(35)-3-methylmorphoiin-4-yl]pyrido[2,3- d] pyrimidin-4-yl] 3-methyl-morpholine

[00523] To a solution of 2,4,7 -trichloropyrido[2,3-d]pyrimidine (4.0 g, 17.06 mmol, 1.0 equiv) in DMA (10 mL) was added (35)-3 -methylmorpholine (4.31 g, 42.65 mmol, 2.5 equiv) and DIPEA (5.51 g, 42.65 mmol, 7.43 mL, 2.5 equiv). The reaction solution was heated to 70 °C for 48 h. The reaction suspension was cooled to room temperature, poured into cold H 2 O (50 mL) to precipitate out a solid. The solid was filtered and the filter cake was rinsed with H 2 O, and dried under reduced pressure to give the crude product, which was purified by column chromatography on silica gel (0-->l00% petroleum ether/EtOAc) to give (35)-4-[7- chloro-2-[(3S)-3-methylmorphoIin-4-yI]pyrido[2,3-d] pyrimidin-4-yI] 3 -methy -morpholine (3.5 g, 56.4% yield) as a yellow solid. LCMS (ESI) m/z: [M + H] calcd for C 1 7H 2 2CIN5O 2 : 364.15; found 364.2

Step 2: Synthesis of 3-[2,4-bis[(3S)-3-methylmorpholin-4-yl]pyrido[2,3-d]pyrimidi n-7- yl]benzoic acid

[00524] To a solution of (3S)-4-[7-chloro-2-[(3S)-3-methylmorpholin-4-yl]pyrido[2,3- d]pyrimidin-4-yl] -3 -methyl -morpholine (2 g, 5.50 mmol, 1.0 equiv) and 3-boronobenzoic acid (1.09 g, 6.60 mmol, 1.2 equiv) in 1,4-dioxane (40 mL) was added a solution of K 2 CO 3 (911.65 mg, 6.60 mmol, 1.2 equiv) in H 2 O (4 mL), followed by PdiPPlisM (317.60 mg,

274.85 μmol, 0.05 equiv). The solution was degassed for 10 min and refilled with N 2 , then the reaction mixture w ? as heated to 100 °C under N for 5 h. The reaction w ? as cooled to room temperature and filtered. The filtrate was acidified by HCl (2N) to pH 3, and the aqueous layer was washed with EtOAc (3 x 20 ml.,). The aqueous phase was concentrated under reduced pressure to give a residue, which was purified by column chromatography on silica gel (50% 1Q0% petroleum ether/EtO Ac) to give 3-[2,4-bis[(3S)-3-methylmorpholin-4- yl]pyrido)2,3-d]pyrimidin-7-yl]benzoic acid hydrochloride (2.5 g, 89.9% yield) as a yellow solid. LCMS (ESI) m/z: [M + H] calcd for C 2 4H 2 7N5O4: 450.21; found 450.2.

[00525] Reference for preparation of this monomer: Menear, K.; Smith, G.C.M.; Malagu, K.; Duggan, H.M.E.; Martin, Leroux, F.G.M. 2012. Pyrido-, pyrazo- and pyrimido- pyrimidine derivatives as mTOR inhibitors. USB 101602. Kudos Pharmaceuticals, Ltd, which is incorporated by reference in its entirety.

Monomer AM. (lr,4r)-4-[4-amino-5-(7-methoxy-1H-indol-2-yI)imidazo[4,3- f ] [ 1 ,2,4] triazin-7-yl ] cyclohexane-1-carboxylic add

[00526] This monomer, also known as OS 1-027 (CAS# = 936890-98-1), is a commercially available compound. At the time this application was prepared, it was available for purchase from several vendors.

Monomer AN, 2-(4-(4-(8-(6-methoxypyridm-3-yl)-3-methyl-2-oxo-2,3-dihydro -1H- imidazo[4,5-c]qumolm-1-yI)-2-(trifluoromethyl)phenyl)piperaz in-1-yl)pyrimidine-5- carboxylic add.

[00527] Preparation of this monomer proceeds by reaction of BGT226 with methyl 2- chloropyrimidine-5-carboxylate, followed by ester hydrolysis, to give the titled Monomer. Monomer AO. 4-amino-5-{1H-pyrrolo[2,3-b]pyridin-5-yl}-5H-pyrimido[5,4-b] indole-8- carboxylic add.

[00528] This monomer can be prepared from 7-methyl-5H-pyrimido[5,4-b]indol-4-ol by benzylic oxidation to the carboxylic acid, conversion to the ethyl ester followed by O- etJiylation with triethyloxonium tetrailuoroboroate. Palladium-mediated arylation followed by ester hydrolysis and final ammonia-olysis provides the monomer.

Preparation of pre- and post-linkers

Boi!dmg block A. teri-l t l N-|(ieri-butoxy)carbonyl]-N-{[2-(piperazm-1-yl)pyriniidm-

5-yl]methyl}carbamate.

Step 1: Synthesis of 5-(bromomethyl)-2-chloropyrimidine [00529] To a solution of 2-chloro-5-methylpyrimidine (92 g, 715.62 mmol 1.0 equiv) in CC (1000 rnL) was added NBS (178.31 g, 1.00 mol, 1.4 equiv) and benzoyl peroxide (3.47 g, 14.31 mmol, 0.02 equiv). The mixture was stirred at 76 °C for 18 h. The reaction mixture was then cooled to room temperature and concentrated under reduced pressure. The reaction mixture was filtered and the solid cake was washed with DCM (150 mL). The resulting solution was concentrated under reduced pressure to give the crude product. The residue was purified by silica gel chromatography (1/0 to 0/1 petroleum ether/EtOAc) to give the product (70.8 g, 47.7% crude yield) as yellow oil, which was used directly for the next step. LCMS (ESI) m/z [M + H] ealed for C5H4B1CIN 2 : 206.93; found 206.9.

Step 2: Synthesis of tert -butyl N-/er/-hutoxycarbonyl-N-((2-piperazin-1-ylpyrimidin-5- yl)methyl)earbamate

[00530] To a solution of tert-buiyl N-Zert-butoxycarbonylcarbamate (36.89 g, 169.79 mmol, 0.74 equiv) in DMF (750 mL) was added NaH (6.88 g, 172.09 mmol, 60 wt.%, 0.75 equiv) at 0 °C. The mixture was stirred at 0 °C for 30 min. Then, 5-(bromomethyl)-2-chloro- pyrimidine (47.6 g, 229.45 mmol, 1.0 equiv) was added at 0 °C. The reaction mixture was stirred at room temperature for 15.5 h. The mixture was then poured into H 2 O (1600 mL) and the aqueous phase was extracted with EtOAc (3 x 300 mL). The combined organic phase was washed with brine (2 x 200 mL), dried with anhydrous Na SCL, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (1/0 to 0/1 petroleum ether/EtOAc) to give the product (70 g, crude) as a yellow solid, which was used to next step directly.

Step 3: Synthesis of / -butyl N-½ri-butoxycarbonyl-N-[(2-piperazin-1-ylpyrimidin-5- y l)me thy 1] carbamate

[00531] To a solution of 1-benzylpiperazine (30.44 g, 122.16 mmol, 1.0 equiv, 2HCl) in MeCN (550 rnL) was added tert-butyl N-i -butoxycarbonyl-N-((2-chloropyrimidin-5- yl)methyl)carbamate (42 g, 122.16 mmol, 1.0 equiv) and K 2 CO 3 (84.42 g, 610.81 mmol, 5.0 equiv). The mixture was stirred at 80 °C for 61 h. The reaction mixture was then diluted with EtOAc (150 mL) and the mixture was filtered. The resulting solution was concentrated under reduced pressure to give the crude product. The residue was purified by silica gel chromatography (1/0 to 0/1 petroleum ether/EtOAc) to give the product (45 g, 74% yield) as a white solid. Step 4: Synthesis of ierf-butyl N-ieri-butoxycarbonyl-N-[(2-piperazin-1-ylpyrimidin-5- yl)melhyl]carbamaie

[00532] To a solution of tert-butyl N-[[2-(4-benzylpiperazin-1-yl)pyrimidin-5-yl]methyl]- N-ierf-butoxycarbonyl-carbamate (24 g, 49.63 mmol, 1.0 equiv) in MeOH (600 mL) was added Pd/C (24 g, 47.56 mmol, 10 wt.%, 1.0 equiv) under argon. The mixture was degassed under reduced pressure and purged with ¾ three times. The mixture was stirred under ¾ (50 psi) at 50 °C for 19 h. The reaction mixture was cooled to room temperature, filtered, and the filter cake was washed with MeOH (500 mL). The resulting solution was concentrated under reduced pressure. The residue w'as purified by silica gel chromatography (1/0 to 0/1 EtOAc/MeOH) to give the product (25.5 g, 68% yield) as a white solid.

Building block B. 2-(4-(5-(((terf-butoxycarbonyl)amino )metfayl)pyrimidin-2- yl)piperazm-I- yl)pyrimidme-5-carboxylic add.

Step 1: Synthesis of ethyl 2-(4-(5-((bis(teri-butoxycarbonyl)amino)methyl)pyrimidin-2- ylSpiperazin-1-yl)pyrimidine-5-carboxylate

[00533] To a solution of ethyl 2-chloropyrimidine-5-carboxylate (2.37 g, 12.71 mmol, 1.0 equiv) and teri-butyl N-teri-butoxycarbonyl-N-((2-piperazin-1-ylpyrimidin-5- y3)methyl)carbamate (5 g, 12.71 mmol, 1.0 equiv) in MeCN (80 mL) was added K 2 CO 3 (5.27 g, 38.12 mmol, 3.0 equiv). The mixture was stirred at 80 °C for 16 h. The reaction mixture was then poured into H 2 O (200 mL) and the suspension was filtered. The filtrate was washed with H 2 O (80 mL) and dried under reduced pressure to give the product (6.1 g, 87% yield) as a white solid.

Step 2: Synthesis of 2-(4-(5-(((f<?ri-butoxycarbonyl)amino)methyl)pyrimidin-2- yI)piperazin-

1-yljpyriinidine-S-carboxylic acid

[00534] To a solution of ethyl 2-(4-(5-((bis(½ri-butoxycarbonyl)ammo)methyr)pyrimidin-

2-yl)piperazin-1-yI)pyrimidine-5-carboxylate (5 g, 9.20 mmol, 1.0 equiv) in H 2 O (50 mL ), EtOH (15 mL) and THF (50 mL) was added Li OH H a) (1.54 g, 36.79 mmol, 4.0 equiv). The reaction mixture was stirred at 55 C C for 16 h. The mixture was then concentrated to remove THE and EtOH and then the mixture was diluted with H 2 O (55 mL) and was acidified (pH=3) with aqueous HCl (1 N). The mixture was filtered and the filter cake was washed with H 2 O (36 L). The filter cake was dried under reduced pressure to give the product (2.7 g, 69.3%) as a white solid. LCMS (ESI) m/z [M + H] calcd for C 1 9H 2 5N7O4: 416.21; found 416.1.

Building block C. iert~\miy\ 2-(piperazin-1-yI)-7,8-dihydropyrido[4,3-d]pyrinndine- 6(5H)-carboxylate.

Step 1: Synthesis of teri-butyl 2-(4-benzylpiperazin-1-yl)-7,8-dihydropyrido[4,3- d]pyrimidine-6(5H)-carboxylate

[00535] To a solutuion of tert-butyl 2-chloro-7,8-dihydiOpyrido[4,3-d]pyrimidine-6(5H)- carboxylate (15 g, 55.61 mmol, 1.0 equiv) in MeCN (150 rnL) was added 1-benzylpiperazine (11.76 g, 66.73 mmol, 1.2 equiv) and K 2 CO 3 (46.12 g, 333.67 mmol, 6.0 equiv). The mixture was stirred at 80 °C for 27 h. The reaction mixture was diluted with EtOAc (200 rnL), filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (1/0 to 0/1 petroleum ether/EtOAc) to give the product (20.2 g, 80% yield) as a white solid. LCMS (ESI) m/z: [M + H] calcd for C 2 3H 3 1N5O 2 : 410.26; found 410.1.

Step 2: Synthesis of tert-butyl 2-(piperazin-1-yl)-7,8-dihydropyrido[4,3-d]pyrimidine-6(5H)- carboxylate

[00536] To a solution of t -butyl 2-(4-benzylpiperazin-1-yl)-7,8-dibydropyrido[4,3- d]pyrimidine-6(5H)-carboxylate (8 g, 19.53 mmol, 1.0 equiv) in MeOH (200 mL) was added Pd/C (8 g, 19.53 mmol, 10 wt.%, 1.0 equiv) under argon. The mixture was degassed and purged with ¾ three times. The mixture was stirred under ¾ (50 psi) at 50 °C for 19 h. The reaction mixture was cooled to room temperature, filtered through a pad of Celiie and the filter calve was washed with MeOH (150 mL). The resulting solution was concentrated under reduced pressure. The crude product was washed with petroleum ether (60 mL) to give the product (9.25 g, 72% yield) as a white solid. LCMS (ESI) m/z [M + H] calcd for C 1 6H 2 5N5O 2 : 320.21; found 320.2. Building block D. 2-(4-(6-(teri-butoxycarbonyl)-5,6,7,8-tetrahydropyrido[4,3- dlpyrimidin- 2-yl)piperazin-1-yl)pyrimidme-5-carboxylic add.

Step 1: Synthesis of tert-bxxtyl 2-(4-(5-(ethoxycarbonyl)pyrimidin-2-yl)piperazin-1-yl)-7,8 - dihydropyrido[4,3-d]pyrimidine-6(5H)-carboxylate

[00537] To a solution of ethyl 2-chloropyrimidine-5-carboxylate (4.09 g, 21.92 mmol, 1.0 equiv) in dioxane (80 mL) was added tert-butyl 2-(piperazin-1-yl)-7,8- dihydropyrido[4,3- d]pyrimidine-6(5H)-carboxylate (7 g, 21.92 mmol, 1.0 equiv) and £¾N (9.15 mL, 65.75 mmol, 3.0 equiv). The mixture was stirred at 90 °C for 64 h. The solution was poured into H 2 O (200 mL) and then the mixture was filtered and the filter cake was washed with H 2 O (100 mL) followed by petroleum ether (60 mL). The filter cake was dried under reduced pressure to give the product (10.1 g, 92% yield) as a brown solid. LCMS (ESI) rn/z: [M + H] calcd for C 2 3H 3 1N7O4: 470.25; found 470.4.

Step 2: Synthesis of 2-(4-(6-(ieri-butoxycarbonyl)-5,6,7,8-tetrahydropyrido[4,3- d]pyrimidin-- 2-yl)piperazin-1-yl)pyrimidine-5-carboxylic acid

[00538] To a solution of teri-butyl 2-(4-(5-(ethoxycarbonyl)pyrimidin-2-yl)piperazin-1- yl)-7,8-dihydropyrido[4,3-d]pyrimidine-6(5H)-carboxylate (6.0 g, 12.78 mmol, 1.0 equiv) in THE (40 mL), EtOH (20 mL) and H 2 O (40 mL) was added LiOlMbO (1.07 g, 25.56 mmol, 2.0 equiv). The reaction mixture was stirred at 35 °C for 15 h. The mixture was then concentrated under reduced pressure to remove THE and EtOH. The mixture was then diluted with H 2 O (500 mL) and was adjusted to pH 3 with aqueous HCl (1 N). The mixture was filtered and the filter cake was washed with H 2 O (80 mL) followed by petroleum ether (80 mL). The filter cake was dried under reduced pressure to give the product (3.8 g, 65% yield) as a white solid. LCMS (ESI) m/z: [M + H] calcd for C 2 1H 2 7N7O 4 : 442.22; found 442.3. Building block E. ieri-buty! meihyl((2-(piperazin-1-yI)pyrimidin-5- yl)methyl)carbamate.

Step 1: Synthesis of (2-chloropyrimidin-5-yl)methanamine

[00539] To a solution of tert-butyl N-/er/-butoxycarbonyl-N-((2-cMoropyrimidin-5- yl)methyl)carbamate (28 g, 81 44 mmol, 1.0 equiv) in EtOAc (30 mL) was added HCl in EtOAc (260 mL). The reaction mixture was stirred at room temperature for 5 h. The reaction mixture was filtered and the filter cake was washed with EtOAc (100 mL). The solid cake was dried under reduced pressure to give the product (14.3 g, 96.6% yield, HCl) as a white solid.

Step 2: Synthesis of tert-buiyl ((2-chloropyrimidin-5-yl)methyl)carbamate

[00540] To a solution of (2-chloropyrimidin-5-yI)methanamine (13 g, 7221 mmol, 1.0 equiv, HCl) in DCM (130 mL) was added DIPEA (20.41 mL, 144.42 mmol, 1.8 equiv) and B0C 2 O (1659 mL, 72.21 mmol, 1.0 equiv), then the mixture was stirred at room temperature for 3 h. The reaction mixture was added to H 2 O (100 mL) and then the aqueous layer was separated and extracted with DCM (2 x 100 mL). Then combined organic phase was washed with sat. NH4CI (2 x 200 mL) and brine (2 x 200 mL), dried with anhydrous NaeSO-n filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (1/0 to 1/1 petroleum ether/EtOAc) to give the product (12 g, 68.2% yield) as a white solid.

Step 3: Synthesis of tert-butyl ((2-chloropyrimidin-5-yl)methyl)(methyl)carbamate

[00541] To a solution of feri-butyl ((2-chloropyrimidin-5-yl)methyl)carbamate (11 g,

45.14 mmol, 1.0 equiv) and Mel (14.05 mL, 225.70 mmol, 5.0 equiv) in THE (150 mL) was added Nall (1.99 g, 49.65 mmol, 60 wt.%, 1.1 equiv) at 0 °C. The mixture was stirred at 0 °C for 3 h and then the reaction was quenched with H 2 O (100 mL). The aqueous phase was extracted with EtOAc (3 x 150 mL) and the combined organic phase was washed with brine (50 mL), dried with anhydrous NaiSO , filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (1/0 to 3/1 petroleum ether/EtOAc) to give the product (9 g, 77.4% yield) as a white solid.

Step 4: Synthesis of t -butyl ((2-(4-benzylpiperazin-1-yl)pyrimidin-5- yl)methyl)(methyl)carbamate

[00542] To a solution of mri-butyi ((2-chloropyrimidin-5-yl)methyl)(methyl)carbamate (9 g, 34.92 mmol, 1.0 equiv) in MeCN (90 mL) was added 1- benzyl piperazine (8.70 g, 34.92 mmol, 1.0 equiv, 2HCl), and K 2 CO 3 (24.13 g, 174.61 mmol, 5.0 equiv). The reaction mixture was stirred at 80 °C for 20 h. The mixture was then filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (1/0 to 1/1 petroleum ether/EtOAc) to give the product (12 g, 86.4% yield) as a yellow oil.

Step 5: Synthesis of /eri-butyi methyl((2-(piperazin-1-yl)pyrimidin-5-yl)methyl)carbamate

[00543] To a solution of tert-butyl ((2-(4-benzylpiperazin-1-yl)pyrimidin-5- yl)methyl)(methyl)carbamate (12 g, 30.19 mmol, 1.0 equiv) in MeOH (120 mL) was added Pd/C (2 g, 10 wt.%). The suspension was degassed and purged with I¾ and then the mixture was stirred under ¾ (15 psi) at room temperature for 3 h. The reaction mixture was filtered through Celite and the filtrate was concentrated under reduced pressure. The residue as purified by silica gel chromatography 1/0 to 1/1 petroleum ether/EtOAc) to give semi-pure material (9 g) as a yellow' oil. Petroleum ether was added to the residue and the solution was stirred at -60 °C until solid appeared. The suspension was filtered and the filtrate was concentrated under reduced pressure to give the product (4.07 g, 55.6% yield) as a yellow oil. LCMS (ESI) m/z: [M + H] calcd for C 15 H 25 N5O 2 : 308.21 ; found 308.1.

Building block G. 2-(4-(5-(((ieri-butoxycarbonyl)(methyl)amino)methyl) pyrimidin-2- yl)piperazm-I-yl)pyrimidine-5-carboxylic add.

Step 1: Synthesis of ethyl 2-(4-(5-(((ferf-butoxycarbonyl)(methyl)amino)methyl) pyrimidin- 2-yl)piperazin-1-yl)pyrimidine-5-carboxylate

[00544] To a mixture of tert-butyl methyl((2-(piperazin-1-yl)pyrimidin-5- yl)methyl)carbamate (4.3 g, 13.99 mmol, 1.0 equiv) and ethyl 2-chloropyrimidine-5- carboxylate (2.87 g, 15.39 mmol, 1.1 equiv) in MeCN (20 mL) was added K 2 CO 3 (3.87 g, 27.98 mmol, 2.0 equiv). The mixture was stirred at 80 °C for 12 h. The reaction mixture then cooled to room temperature and was filtered. The filtrate was concentrated under reduced pressure and the crude product w'as purified by silica gel chromatography (1/0 to 1/1 petroleum ether/EtOAc) to give the product (4.7 g, 713% yield) as a white solid.

Step 2: Synthesis of 2-(4-(5-(((it t-butoxycarbonyl)(meihyl)ammo)methyl) pyrimidin-2- yl)piperazin-1-yl)pyrimidine-5-carboxylic acid

[00545] To a solution of ethyl 2-(4-(5-(((/¾r/- butoxycarbonyl)(methyl)amino)methyl)pyrimidin-2-yl)piperazin -1-yl)pyrimidine-5- carboxylate (6 g, 13.11 mmol, 1.0 equiv) in THF (100 mL), EtOH (30 mL), and H 2 O (30 mL) was added LiOH « H 2 O (1.10 g, 26.23 mmol, 2.0 equiv). The mixture was stirred at room temperature for 16 h. The mixture was then concentrated under reduced pressure to remove THF and EtOH and then neutralized by the addition of IN HCl. The resulting precipitate was collected by filtration to give the product (5.11 g, 90.1% yield) as a white solid. LCMS (ESI) m/z: [M + H] calcd for C 20 H 2 7N7O4: 430.22; found 430.2.

Building block G. feri-butyl N-f<?rf-butoxycarbonyl-N-((2-(2-((ieri- butyl(diphenyl)silyl)oxymethyl)piperazin-1-yl)pyriinidin-5-y l)methyl)carbamate.

Step 1: Synthesis of tert-butyl N-((2-(4-benzyl-2-(hydroxymethyl)piperazin-1-yl)pyrimidin- 5-yl)methyl)-N-/er/-butoxycarbonyl-carbamate

[00546] To a solution of tert-butyl N-/er/-butoxycarbonyl-N-((2-chloropyrimidin-5- yl)methyl)carbamate (18.33 g, 53.32 mmol, 1.1 equiv) and (4-benzylpiperazin-2-yl)methanol (10 g, 48.48 mmol, 1.0 equiv) in DMF (100 mL) was added K 2 CO 3 (13.40 g, 96.95 mmol,

2.0 equiv). The mixture was stirred at 100 °C for 12 h. The reaction mixture was then cooled to room temperature and H 2 O (100 mL) was added. The aqueous layer was extracted with EtOAc (2 x 150 mL) and the combined organic layer was washed with brine (20 mL), dried with NaaS0 4 , filtered and the filtrate was concentrated under reduced pressure to give the product (7.3 g, 29.3% yield) as a yellow' oil. LCMS (ESI) m/z: [M + H] calcd for C 2 7H 3 9N5O5: 514,31; found 514.5

Step 2: Synthesis of tert-bulyl N-((2-(4-benzyl-2-((terf- biUyl(diphenyl)silyI)oxymethyl)piperazin-1-yl)pyrimidin-5-yl )methyi)-N-ii?ri- butoxycarbonyl-carbamate

[00547] To a solution of tert-butyl N-((2-(4-benzyl-2-(hydroxymethyI)piperazin-1- yl)pyrimidin-5-y1)meihyl)-N-i -butoxycarbonyl-carbamate (2.3 g, 4.48 mmol, 1.0 equiv) in DCM (30 mL) %'as added imidazole (609.69 mg, 8.96 mmol, 2.0 equiv) and TBDPSC 1 (1.73 mL, 6.72 mmol, 1.5 equiv). The reaction mixture was stirred at room temperature for 2 h. The mixture was then washed with H 2 O (lOOmL) and the aqueous phase extracted with EtOAc (2 x 60 mL). The combined organic phase w'as washed with brine (20 mL), dried with Na SOi, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (20/1 to 3/1 petroleum ether/ElOAe) to give the product (4 g, 59.4% yield) as a yellow' oil. LCMS (ESI) m/z [M + H] calcd for C 4 3H57N5O5S1: 752.42; found 752.4.

Step 3: Synthesis of /eri-butyl N-terributoxycarbonyl-N-((2-(2-((i <? rf- butyl(diphenyl)silyl)oxymethyl)piperazin-1-yl)pyrimidin-5-yl )methyl)carbamate

[0054S] To a solution of /erf-butyl N-((2-(4-benzyl-2-((reri- buty l(dipheny l)sily i)oxymethyl)piperazin-1-y f)pyrimiclin-5-y 1 )metlvyl)-N-/er/- butoxycarbonyi-carbamate (3.3 g, 4.39 mmol, 1.0 equiv) in EtOH (10 mL) was added Pd(OH) 2 /'C (1 g, 10 wt.%). The mixture was heated to 50 °C under II2 (30 psi) for 30 h. The mixture was then cooled to room temperature, filtered through Celite, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (20/1 to 3/1 EtOAc/EtOH) to give the product (1.44 g, 45.6% yield) as a yellow' solid. LCMS (ESI) m/z [M + H] calcd for CseHsiNsOsSi: 662.38; found 662.3. Building block H. 2-(4-(5-(((ieri-butoxycarbonyl)amino)methyl)pyrimidin-2-yl)- 3- (hydroxymethyI)piperazin-1-yl)pyriimdine-5-carboxylic add.

Step 1 : Synthesis of iert-butyl N--ftro-hutoxycarbonyl--N-((2--(2--(hydroxymethyl)piperazin- T- yl) pyrimidin-5-yl)melhyl)carbamaie

[00549] To a solution of feri-butyl N-((2-(4-benzyI-2 (hydroxymethyl)piperazin- l·· yl)pyrimidin-5-yl)metbyI)-N-ieri-butoxycarbonyl-carbamate (3 g, 5.84 mmol, 1.0 equiv) in EtOH (40 mL) was added Pd/C (2 g, 10 wt.%). The suspension was degassed and purged with ¾, then stirred under ¾ (50 psi) at 30 °C for 16 h. The reaction mixture was cooled to room temperature and filtered through Celite and then concentrated under reduced pressure to give the product (1.6 g, crude) as a yellow oil. LCMS (ESI) rn/z: [M + H] calcd for C 20 H 3 3N5O5: 424.26; found 424.3.

Step 2: Synthesis of ethyl 2-(4-(5-((bis(ieri-butoxycarbonyl)amino)methyl)pyrimidin-2-y l)-3- (hydroxymethyl)piperazin-1-yl)pyrimidine-5-carboxylate

[00550] To a solution of tert-butyl N-/er/-butoxycarbonyl-N-((2-(2- (hydiOxymethyl)piperazin-1-yl)pyrimidin-5-yl)methyl)carbamat e (1.4 g, 3.31 mmol 1.0 equiv) in MeCN (20 mL) was added K 2 CO 3 (2.28 g, 16.53 mmol, 5.0 equiv) and ethyl 2- chloropyrixnidine-5-carboxylate (616.84 mg, 3.31 mmol, 1.0 equiv). The solution was stirred at 80 °C for 4 h. The mixture was cooled to room temperature and poured into H 2 O (30 mL). The aqueous layer was extracted with EtOAc (2 x 30 mL) and the combined organic layer was washed with brine (20 mL), dried with NaiSCb, filtered and concentrated under reduced pressure. The mixture was purified by silica gel chromatography (20/1 to 3/1 petroleum

1 s ether/EtOAc) to give the product (1.6 g, 66.7% yield) as a light yellow solid. LCMS (ESI) m/z: [M + H] caicd for C 2 7H 3 9N7O7: 574.30; found 574.4.

Step 3: Synthesis of 2-(4-(5-(((t 4>utoxycarbonyl)amino)methyl)pyrimidin-2-yr)-3- (hydiOxymethyl)piperazin-1-yl)pyrimidine-5-carboxylic acid

[00551] To a solution of ethyl 2-(4-(5-((bis(iert-butoxycarbonyl)amino)methyl)pyrimidin- 2-yl)-3-(hydfoxymethyi)piperazin-1-yi)pyrimidine-5-carboxy3a le (1.4 g, 2.44 mmol, 1.0 equiv) in THF (6 mL) and EtOH (6 mL) at 0 °C was added a solution of LiOFFEkO (512.07 mg, 12.20 mmol, 5.0 equiv) in Hi>0 (3 mL). The reaction mixture was warmed to room temperature and stirred for 2 h. The mixture was then concentrated under reduced pressure to remove THF and EtOH. The aqueous phase was adjusted to pH 3 with 0.1 M HCl and the resulting suspension was filtered. The solid cake was dried under reduced pressure to give the product (613.14 mg, 55.6% yield) as a white solid. LCMS (ESI) n/'z [M + H] caicd for C 20 H 2 7N7O5: 446.22; found 446.2.

Building block I, tert-bu lyl N-[(ieri-butoxy)carbonyl]-N-({2-[(3/f)-3- (hydroxymethyl)piperazin-1-yI]pyrimidin-5-yI}methyl)carbamat e.

Step I : Synthesis of (R)-teri-butyl-N-½ri-butoxycarbonyl-((2-(3-(((ieri-butyldip henylsilyl)- oxy)methyl)piperazin-1-yl)pyrimidin-5-yl)methyl)carbamate

[00552] To a solution of ½rf-butyl-N-feri-butoxycarbonyl-((2-chloropyrimidin-5- yl)methyl)carbamate (24.24 g, 70.51 mmol, 1.0 equiv) in MeCN (300 mL) was added (R)-2- ((( tert-butyldiphenylsilyI)oxy)methyl)piperazine (25 g, 70.51 mmol, 1.0 equiv) and K 2 CO 3 (29.24 g, 211.53 mmol, 3.0 equiv). The mixture was stirred at 80 C C for 16 h. The reaction mixture was then cooled to room temperature, diluted with EtOAc (200 mL), filtered and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography (0 — > 1 ()()% EtOAc/petroleum ether) afforded the desired product (46.5 g, 94% yield) as a white solid.

Step 2: Synthesis of tert-butyl N-[(/i?r/-butoxy)carbonyl]-N-({2-[(3R )-3- (bydroxymethyl)piperazin-1-yl]pyrimidin-5-yl}methyl)carhamat e [00553] To a solution of (R )-iiri-butyl-N-ieri-butoxycarbonyl-((2-(3-(((½r/- butyldiphenylsilyl)oxy)methyl)piperazin-1-yl)pyrimidin-5-yl) methyl)carbamate (12 g, 18.13 mmol, 1.0 equiv) in THF (120 mL) was added TBAF (1 M, 23.93 mL, 1.3 equiv). The mixture was stirred at room temperature for 2 h. The reaction mixture was then poured into H 2 O (300 mL) and the aqueous phase was extracted with EtOAc (3 x 80 mL). The combined organic phases were combined, washed with brine (80 mL), dried, filtered and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography (0- 20% MeOH/DCM) afforded the desired product (5 g, 64% yield) as a yellow solid.

Building Muck J, 2-{4-[5-({[(#er#-butoxy)carbonyl]amino}methyl)pyrimidin-2- yl]piperazin-1-yl}pyrimidine-5-car boxylic add.

Step 1: Synthesis of (j?)-ethyl 2-(4-(5-(((di-ieri-butoxycarbonyl)amino)methyl)pyrimidin-2- yl)-2-(((n?/t-butyldiphenyIsilyl)oxy)methyl)piperazin-1-yl)p yrimidine-5-carboxylate

[00554] To a solution of (R )-feri-butyl-N-ieri-butoxycarbonyl-N-((2-(3-(((ieri- butyldiphenylsilyl)oxy)methyl)piperazin-1-yl)pyiimidin-5-yl) methyl)carbamate (31.5 g,

45.21 mmol 1.0 equiv) in MeCN (350 mL) was added ethyl 2-chloropyrimidine-5- carboxylate (8.44 g, 45.21 mmol, 1.0 equiv) and K 2 CO 3 (18.75 g, 135.63 mmol, 3.0 equiv). The mixture was stirred at 80 °C for 16 h. The reaction mixture was then cooled to room temperature, diluted with EtOAc (150 mL), and filtered to remove inorganic salts. The filtrate was then concentrated under reduced pressure. Purification by silica gel chromatography (Q 100% EtOAc/petroleum ether) afforded the deshed product (33.5 g, 89% yield).

Step 2: Synthesis of (j?)-ethyl 2-(4-(5-(((di-ieri-butoxycarbonyl)amino)methyl)pyrimidin-2- yl)-2-(hydroxymethyl)piperazin-1-yI)pyrimidine-5-carboxylate [00555] To a solution of (A’)--ethyl 2-(4-(5-(((di-firt- butoxycxirbonyl)amino)methyl)pyrimidin-2-yl)-2-(((fi t- butyldiphenyisiiyI)oxy)methyl)piperazin-1-yl)pyrimidine-5-ca rboxyIate (36.5 g, 44.95 mmol, 1.0 equiv) in THF (300 mL) was added TBAF (1 M, 59.33 mL, 1.32 equiv). The mixture was stirred at room temperature for 6 h, at which point the reaction mixture was poured into H 2 O (500 mL). The aqueous phase was separated and extracted with EtOAc (3 x 150 mL) and the combined organic layers were washed with brine (150 mL), dried, filtered, and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography (0-->100% EtOAc/petroleum ether) afforded the desired product (17 g, 64% yield) as a yellow oil.

Step 3: Synthesis of (f?)-2-(4-(5-(((firi-butoxycarbonyl)amino)methyl)pyrimidin-2 -yl)-2- (hydroxymethyl)piperazin-1-yl)pyrimidine-5-carboxylic acid

[00556] To a solution of(R)-ethyl 2-(4-(5-(((di- tert- butoxycarbonyl)amino)methyl)pyrimidin-2-yl)-2-(hydroxymethyl )piperazm-1-yl)pyrimidine- 5-carboxylate (17 g, 29.64 mmol, 1.0 equiv) in H 2 O (160 mL), EtOH (80 mL) and THF (160 mL) was added LiGFFHiO (4.97 g, 118.54 mmol, 4.0 equiv). The reaction mixture was stirred at 55 °C for 16 h. To the mixture was then added LiOIMIiO (1.01 g, 24.00 mmol,

0.81 equiv) and the reaction mixture was stirred at 55 °C for an additional 9 h. The mixture was cooled to room temperature, diluted with H 2 O (150 mL), and concentrated under reduced pressure to remove THF and EtOH. The mixture was acidified (pH = 5) with 1 N HCl, filtered, and the filter cake washed with H 2 O (2 x 30 mL). The filter cake was dried under reduced pressure to afford the desired product (9.2 g, 67% yield) as a white solid. LCMS (ESI) m/z: [M + H] calcd for C 20 H 27 N 7 O 5 : 446.22; found 446.1.

Building block K. fiat-butyl N-[(teri-butoxy)carbonyl]-N-({2-[(3S)-3- (hydroxymethyl)piperazin-1-yl]pyrimidin-5-yl]methyl)carbamat e.

[00557] This building block is prepared by a process similar to tha for Building block I by utilizing [(25)-piperazin-2-yl]methanoL Building block L, 2-[(2S)-4-[5-({[(i<?ri-butoxy)carbonyl]amino}methyl)pyrim idin-2-yl]-2- (hydroxymethyI)piperazin-1-yl]pyriimdme-5-carboxylic add.

[00558] This building block is prepared from Building block K by a process similar to that for Building block J.

Building Mock M, tenf-butyl 2-[(3R )-3-(hydroxymethyl)piperazin-1-yl]-5H,6H,7H,8H- pyrido[4,3-d]pyrimidine-6-carboxylate.

Step 1 : Synthesis of (R)-tert-buty] 2-(3-(((feri-butyldiphenylsilyl)oxy)-methyl)piperazin-1- yl)-7,8-dihydropyrido[4,3-d]pyrimidine-6(5H)-carboxylate

[00559] To a solution of (R )-2-(((i< ?/ t-butyldiphenylsilyl)oxy)meihyl)piperazine (25 g, 70.51 mmol, 1.0 equiv) in MeCN (250 mL) was added K 2 CO 3 (29.24 g, 211.53 mmol, 3.0 equiv) and ieri-butyl 2-chloro-7,8-dihydropyrido[4,3-d]pyrimidme-6(5H)-carboxylate (17.12 g, 63.46 mmol, 0.9 equiv). The mixture was stirred at 80 °C for 17 h. The reaction mixture was then cooled to room temperature, filtered, and the filtrated was concentrated under reduced pressure. Purification by silica gel chromatography (0-- 100% EtOAc/petroleum ether) afforded the desired product (31 g, 73.5% yield) as a white solid. LCMS (ESI) m/z [M + H] calcd for C 3 3H45N5O 3 S1: 588.34; found 588.2.

Step 2: Synthesis of (J?)-tert-butyl 2-(3-(hydroxymethyl)piperazin-1-yl)-7,8- dihydropyrido [4,3 -d]pyrimidine- 6(5H)- carboxy late

[00560] To a mixture of (R)-te ri-butyl 2-(3-(((tert- butyldiphenylsily])oxy)methyl)piperazin-1-yl)-7,8-dihydropyr ido[4,3-d]pyrimidine-6(5H)- carboxylate (12 g, 20.41 mmol, 1.0 eqtiiv) in THF (120 mL) was added TBAF (1.0 M, 24.50 mL, 1.2 equiv). The mixture was stirred at room temperature for 5 h. The mixture was poured into H 2 t) (100 mL) and the aqueous phase was extracted with EtOAc (2 x 100 mL). The combined organic phases were washed with brine (100 mL), dried, filtered, and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography (Q→ 10% MeOH/DCM) afforded the desired product (6 g, 84.1% yield) as a white solid. LCMS (ESI) m/z: [M + H] calcd for C 1 7H 2 7N5O 3 : 350.22; found 3502.

Building block N. 2-[(2R)-4-{6-[(ter -butoxy)carbonyl]-5H,6H,7H,8H-pyrido[4,3- d]pyrimidin-2-yl}-2-(hydroxyinethyl)piperaziii-1-yl]pyrimidi iie-5-carboxylic add.

[00561] This building block is prepared from Building block M by a process similar to that for Building block J.

Bollding block O, tert- butyl 2-[(3S)-3-(hydroxymethyl)piperazm-1-yl]-5H,6H,7H,8H- pyrido[4,3-€l]pyrimidine-6-earboxylate.

[00562] This building block is prepared by a process similar to that for Building block I by utilizing /erf-butyl 2-chloro-7,8-dihydiOpyrido[4,3-d]pyrimidine-6(5H)-carboxylat e and [(25)-piperazin-2-yl]methanoL

Building block P, 2-[(2S)-4-{6-[(ieri-butoxy)carhonyl]-5H,6H,7H,8H-pyrido[4,3- d]pyrimidin-2-yl}-2-(hydroxymethyl)piperazin-1-yl]pyrimidine -5-carboxylic add. [00563] This building block is prepared from Building block O by a process similar to that for Building block J.

Boll ing block , iert~\mi \ N-[(ieri-butoxy)carbonyl]-N-({2-((3S)-3- [(dirnethylamino)methyI]piperazin-1-yl]pyrimidin-5-yl}methyl )carbamate.

Step 1: Synthesis of (if)-dibenzyl 2-(dimethylcarbamoyl)piperazine-1 ,4-dicarboxylate

[00564] To a solution of CDI (1221 g, 75.30 mmol, 1.2 equiv) in DCM (300 mL ) at 0 °C was added (R)-1 ,4-bis((benzyloxy)carbonyl)piperazine-2-carboxy lie acid (25 g, 62.75 mmol, 1.0 equiv). The mixture was stirred at 0 °C for 0.5 h, at which time dimethylamine (8.51 mL, 92.87 mmol, 1.5 equiv, HCl) was added. The reaction mixture was warmed to room temperature and stirred for 12 h. The reaction mixture was then added to H 2 O (200 mL), and the aqueous layer was separated and extracted with DCM (2 x 200 mL). The combined organic phases were washed with brine (2 x 50 mL), dried, filtered, and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography (50→ 100% EtOAc/petroleum ether) afforded the desired product (23.5 g, 88.0% yield) as a yellow oil.

Step 2: Synthesis of (S) -dibenzyl 2-((dimethylamino)methyl)piperazine-1 ,4-dicarboxylate

[00565] To a solution of (R )-dibenzyl 2-(dimethylcarbamoyl)piperazine-1,4-dicarboxylate (28 g, 65.81 mmol, 1.0 equiv) in THF (300 mL) at 0 °C was added Blfb^ e S (10 M, 13.16 mL, 2.0 equiv). The reaction mixture was then stirred at 80 °C for 3 h. The reaction mixture was cooled to room temperature and then MeOH (50 L) was added. After stirring for an additional 1 h the mixture was concentrated under reduced pressure. Purification by silica gel chromatography (50→100% EtOAc/petroleum ether) afforded the desired product (18 g, 66.5% yield) as a yellow oil.

Step 3: Synthesis of (R )-N,N-dimethyl-1-(piperazin-2-yl)methanamine [00566] To a solution of (5) -dibenzyl 2-((dimethylamino)methyl)piperazine-1,4- dicarboxylate (18 g, 43.74 mmol, 1.0 equiv) in EtOAc (200 mL) was added Pd/C (1.5 g, 10 wt.%). The suspension was degassed under reduced pressure and purged with ¾ three times. The suspension was stirred under ¾ (30 psi) at 30 °C for 5 h. The reaction mixture was then filtered through celite and the filtrate was concentrated under reduced pressure to afford the desired product (6 g, 95.8% yield) as a yellow' solid.

Step 4: Synthesis of tert-butyl N-terf-butoxycarbonyl-N-((2-((3S)-3-

((dimethylamino)methyl)piperazin-1-yl)pyrimidin-5-yl)meth yl)carbamate

[00567] To a solution of (R )-N,N-dimetliyl-1-(piperazin-2-yl)methanamine (2.8 g, 19.55 mmol, 1.0 equiv) in MeCN (40 mL) was added ieri-butyl N-ieri-butoxycarbonyl-N-((2- chloropyrimidin-5-yl)methyl)carbamate (6.72 g, 19.55 mmol, 1.0 equiv) and K 2 CO 3 (5.40 g, 39.10 mmol, 2.0 equiv). The mixture was stirred at 80 °C for 24 h. The mixture was then cooled to room temperature, filtered, and the filter cake washed with EtOAc (3 x 10 mL). The filtrate was then concentrated under reduced pressure. Purification by silica gel chromatography (0→1(K)% MeOH/EtOAc) afforded the desired product (5.3 g, 57.8% yield) as a yellow oil. LCMS (ESI) m/z: [M + H] calcd for C 2 2H 3 S N0O4: 451.31; found 451.2.

Building block R, 2-[(2S)-4-[5-({[(ierf-butoxy)carbonyl]amino}methyl)pyrimidin -2-yl]-2- [(dimethylamino)methyl]piperazin-1-yl]pyriniidiiie-5-carboxy lic add.

Step 1: Synthesis of (S)-ethyl 2-(4-(5-(((bi-teri-butoxycarbonyl)amino)methyl) pyrimidin-2- yl)-2-((dimethylamino)methyl)piperazin-1-yl)pyrimidine-5-car boxylate

[00568] To a solution of (S)-ieri-butyl-N-ieri-butoxycarbonyl ((2-(3- ((dimethylamino)methyl) piperazin-1-yl)pyrimidin-5-yl)methyl)carbamate (3.26 g, 7.24 mmol, 1.0 equiv) in DMF (30 mL) 'as added EtsN (3.02 mL, 21.71 mmol, 3.0 equiv) and ethyl 2-chloropyrimidine-5-carboxylate (1.47 g, 7.86 mmol, I equiv). The mixture was stirred at 50 °C for 3 h and then concentrated under reduced pressure to afford the desired product (4.35 g, crude) as a solution in DMF (30 mL), which was used directly in the next step. LCMS (ESI) m/z;. [M + H] calcd for CcgELciNsCL: 601.35; found 601.5. Step 2: Synthesis of (S)-2-(4-(5-(((i<??7-butoxycarbonyl)amino)methyl)-pyrirni din-2-yl)-2- ((dimethylamino)methyl)piperazin-i-yl)pyrimidin e -5-carboxylic acid

[00569] To a solution of (S)-eihyl 2-(4-(5-(((bi-½rf-butoxycarbonyl)amino)methyr)- pyrimidin-2-yl)-2-((dimetliy{amino)metliy{)piperazin-1-yl)py rimicliiie-5-carboxylate (4.35 g, 7.24 mmol, 1.0 equiv) in DMF (30 mL) was added DMF (50 mL) EtOH (30 mL) and ¾0 (30 mL). To the solution was then added LiOfMLO (3 g, 71.50 mmol, 9.9 equiv) at 50 °C. The reaction was stirred at 50 °C for 36 h. The mixture was then cooled to room temperature, neutralized with 0.5 N HQ, and concentrated under reduced pressure. Purification by reverse phase chromatography (2 ~~ >30% MeCN/HiO) afforded the desired product (1.15 g, 34% yield) as a white solid. LCMS (ESI) m/z: [M + H] calcd for C 2 .2H 3 2N8O4: 473.26; found 473.3.

Building block S, fcrrf-butyl N-[(tenf-butoxy)carbonyl]-N-({2-[(3R )-3- [(dimethylamino )methyl]piperazm-1-yl]pyrimidm-5-yl}methyl)carbamate.

% NH

[00570] This building block is prepared by a process similar to that for Building block I by utilizing dimethyl( { [(2S)-piperazin-2-yl]metby 1 ] )amine.

Building block T, 2-[(2^)-4-[5-({[(ii?rf-buioxy)carboByl]amIiio}methyl)pyrimi l 2-yI]-2- [(dimetfay!asBbi0)melfayl]piperazm-Tyl]pyr!mMme-5-earboxy!k acid.

[00571] This building block is prepared from Building block S by a process similar to that for Bui lding block I. Building block U. tert-butyl 2-[(3S)-3-[(dimethylamino )methyl]piperazin-1-yl]- 5H,6H,7H,8H-pyrido[4,3-d]pyrimidine-6-carboxylate.

[00572] To a solution of tert-hutyl 2-chloro-7,8-dihydropyrido[4,3-d]pyrimidine-6(5H)- carboxylate (4.80 g, 17.80 mmol, 1.4 equiv) in MeCN (45 mL) was added K 2 CO 3 (10.42 g, 75.40 mmol, 3.0 equiv) and (R )-N,N -dimethyl-1-(piperazin-2-yl)methanamine (3.6 g, 25.13 mmol, 1.0 equiv). The mixture was stirred at 80 °C for 8 h. The mixture was then cooled to room temperature, filtered, and the filter cake was washed with EtOAc (50 mL). To the organic phase was added H 2 O (50 mL) and the aqueous phase was extracted with EtOAc (3 x 50 mL). The combined organic phases were washed with brine (5 mL), dried, filtered and concentrated under reduced pressure. Purification by silica gel chromatography (8→ 67% EtOAc/petroleum ether) afforded the desired product (6.5 g, 63.5% yield) as a yellow' oil.

Building block V. 2-[(2S)-4-{6-[(tert-butoxy)carbonyl]-5H,6H,7H,8H-pyrido[4^- d]pyri!nidin-2-yl}-2 [(dimeihyIamino)!iiefhyl]piperazin-1-yl]pyrimidine-5-carboxy Mc add.

Step 1: Synthesis of (S)-ieri-butyl 2-(3-((dimethylaimno)methyl)-4-(5-

(ethoxycarbonyI)pyrimidin-2-yl)piperazin-1-yl)-7,8-dihydr opyrido[4,3-d]pyrimidine-6(5H)- carboxylate

[00573] To a solution of (S)-i <? ri-butyl 2-(3-((dimethylaniino)methyl)piperazin-1-yl)-7,8- dihydropyrido[4,3-d]pyrimidine-6(5H)-carboxylate (3 g, 7.97 mmol, 1.0 equiv) in DMF (70 mL ) at 0 °C was added NaH (382.44 mg, 9.56 mmol, 60 wt.%, 1.2 equiv). The suspension was stirred at 0 °C for 0.5 h, then ethyl 2-cbIoropyrimidine-5-carhoxylate (1.49 g, 7.97 mmol, 1 equiv) in DMF (50 mL) was added, dropwise. The mixture was warmed to room temperature and stirred for 5 h. The mixture was then cooled to 0 °C and poured into H 2 O (360 mL). The suspension was filtered, and the filter cake washed with H 2 O (30 mL) and dried under reduced pressure. Purification by silica gel chromatography (6%→ 33% EtOAc/petroleum ether) afforded the desired product (1.8 g, 39.6'% yield) as a brown oil.

Step 2: Synthesis of (S)-2-(4-(6-(terf-butoxycarbonyl)-5,6,7,8-tetrahydropyrido[4 ,3- d]pyrimidin-2-yl)-2-((dirnethylarmno)methyl)piperazin-i-yl)p yrirmdine-5-carboxylic acid

[00574] To a solution of (5)-/e/7-butyl 2-(3-((dimethylamino)methyl)-4-(5- (ethoxycarbonyI)pyrimidin-2-yl)piperazin-1-yl)-7,8-dihydropy rido[4,3-d]pyrimidine-6(5H)- carboxylate (1.1 g, 2.09 mmol, 1.0 equiv) in THF (5 mL), EtOH (2.5 mL), and H 2 O (2.5 mL) was added LiOEFthO (175.30 mg, 4.18 mmol, 2.0 equiv). The mixture was stirred at room temperature for 2 h, at which point the pH was adjusted to 7 by the addition of 1 N HCl at 0 °C. The mixture was concentrated under reduced pressure to remove THF and MeOH. The resulting suspension was filtered, and the filter cake was washed with H 2 O (5 mL) and dried under reduced pressure to afford the desired product (680 mg, 65.3% yield) as a white solid. LCMS (ESI) m/z: [M + H] calcd for C 2 4H34N8O4: 499.28; found 499 2.

Building block W. ten- butyl 2-I(3tf)-3-[(dimethylamino)methyl]piperazin-1-yl]- 5H,6H,7H,8H-pyrido[4,3-d]pyrimidine-6-carboxylate.

[00575] This building block is prepared by a process similar to that for Building block I by utilizing feri-butyl 2-chloro-7,8-dihydropyrido[4,3 -d]pyrimidine-6(5H)-carboxylate and dimethyl({[(2S)-piperazin-2-yl]methyl})amme.

Building block , 2 (2R H6^eri-hi i y)c^rhoi^i]~5M -pjrkio\4,3- d]pyriimdm-2-yI}-2-[(dimetfaylamino)inethyl]piperazin-1-yl]p yriraidine-5-carboxylic add. [00576] This building block is prepared from Building block W by a process similar to that for Building block J.

Building block Y. tert-hutyl (2R )-4-{6-((iiri-butoxy)carbonyl]-5H,6H,7H,8H-pyrido[4,3- d]pyriimdin-2-yI}piperazine-2-carboxyIate.

Step 1: Synthesis of (R)~ 1 ,4-bis((benzyloxy)carbonyl)piperazine-2-carboxylic acid

[00577] To a solution of (R )-piperazine-2-carboxylic acid (70 g, 344.71 mmol, 1.0 equiv, 2HCl) in dioxane (1120 rnL) and H 2 O (700 mL) was added 50% aq. NaOH until the solution was pH---- i 1. Benzyl chloroformate (156.82 mL, 1.10 mol, 3.2 equiv) was added and the reaction mixture was stirred at room temperature for 12 h. To the solution was then added H 2 O (1200 mL) and the aqueous layer was washed with MTBE (3 x 800 mL). The aqueous layer was adjusted to pH =2 with concentrated HCl (12N) and extracted with EtOAc (2 x 1000 mL). The combined organic extracts were dried, filtered and the filtrate was concentrated under reduced pressure to afford the desired product (137 g, 99.8% yield) as a yellow solid. LCMS (ESI) rn/z [M + H] calcd for C 2 1H 2 2N 2 O6: 399.16; found 399.2.

Step 2: Synthesis of (R )-1,4-dibenzyl 2- tert-butyl piperazine-1,2,4-tricarboxy late

[00578] To a solution of (R )-h4-bis((benzyloxy)carbonyl)piperazine-2-carboxylic acid (50 g, 125.50 mmol, 1.0 equiv) in toluene (500 mL) at 80 °C was added l,1-di-/cr/-butoxy-N,N- dime thy Imethan amine (57.17 mL, 238.45 mmol, 1.9 equiv). The solution was stirred at 80 °C for 2 h, at which point the reaction mixture was cooled to room temperature and partitioned between EtOAc (300 mL) and H 2 O (500 mL). The aqueous layer was extracted with EtOAc (2 x 500 mL) and the combined organic layers were dried, filtered and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography (0%→25% EtOAc/petroleum ether) afforded the desired product (35 g 61.2% yield) as a white solid. LCMS (ESI) m/z: [M + Na] calcd for C 2 5H30N 2 O6: 477.20; found 477.1. Step 3: Synthesis of (R)-tert -butyl piperazine-2-carboxylate

[00579] To a solution of (R )-1,4-dibenzyl 2-feit-bulyl piperazine-1, 2, 4-tricarboxy late (35 g, 77.01 mmol, 1.0 equiv) in EtOAc (350 mL) was added Pd/C (10 g, 10 wt.%). The suspension was degassed under reduced pressure and purged with ¾ three times. The mixture was stirred under Ph (30 psi) at 30 °C for 4 h. The reaction mixture was then filtered through celite, the residue was washed with MeOH (5 x 200 mL), and the filtrate was concentrated under reduced pressure to afford the desired product (14 g, 79.6% yield) as yellow oil. LCMS (ESI) rn/z [M + H] calcd for C9H18N 2 O 2 : 187.15; found 187.1.

Step 4: Synthesis of {R)-tert-bxxty\ 2-(3-(terf-butoxycarbonyl)piperazin-1-yl)-7,8- dihydropyrido[4,3-d]pyrimidine-6(5H)-carboxylate

[00580] To a solution of t -butyl (2R)-piperazine-2-carboxylate (12 g, 64.43 mmol, 1.0 equiv) in MeCN (200 mL) was added K 2 CO 3 (17.81 g, 128.86 mmol, 2.0 equiv) and te rt- butyl 2-chloro-7,8-dihydropyrido[4,3-d]pyrimidine-6(5H)-carboxylat e (17.38 g, 64.43 mmol, 1.0 equiv). The reaction mixture was heated to 80 °C and stirred for 12 h. The reaction mixture was then cooled to room temperature and filtered, the residue was washed with EtOAc (3 x 150 L), and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography (0%- T00% EtOAc/petroleum ether) afforded the desired product (19 g, 69.2% yield) as a yellow' solid. LCMS (ESI) m/z: [M + H] calcd for C 2 1H 3 3N5O4: 420.26; found 420.2.

Building block Z. 4-amino -2-[i2R )-2-[(feri-but0xy)carbosiyI]-4-{6-[(a¾ri- butoxy)carbonyl]-5H,6H,7H,8H-pyrido[4,3-d]pyrimidin-2-yl}pip erazin-1-yl]pyrimidine- 5-carboxyIic acid.

Step 1 : Synthesis of (R)-tert-butyl 2-(4-(4-amino-5-(ethoxycarbonyl)pyrimidin-2-yl)-3-(/er/- butoxycarbonyl)piperazin-1-yl)-7,8-dihydropyrido[4,3-d]pyrim idine-6(5H)-carboxylate

[00581] To a stirred solution of {R)-tert-buiy\ 2-(3-(ieri-butoxycarbonyl)piperazin-1-yl)- 7,8-dihydiOpyrido[4,3-d]pyrimidine-6(5H)-carboxylate (12 g, 28.60 mmol, 1.0 equiv) in MeCN (150 mL) was added K 2 CO 3 (7.91 g, 57.20 mmol 2.0 equiv) and ethyl 4-amino-2- chloropyrimidine-5-carboxylate (692 g, 34.32 mmol, 1 2 equiv). The reaction mixture was stirred at 80 °C for 12 h, at which point the reaction mixture was filtered and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography (0%→17% EtOAc/petroleum ether) afforded the desired product (16 g, 91.6% yield) as a yellow solid LCMS (ESI) m/z: [M + H] calcd for C 28 H4oN 8 0 6 : 585.32; found 585.1.

Step 2: Synthesis of (R )-4-amino-2-(2-(½ri-butoxycarbonyl)-4-(6-(ieri-butoxycarbon yl)-

5.6.7.8-tetrahydropyrido[4,3-d]pyrimidin-2-yl)piperazin-1 -yl)pyrimidine-5-carboxylic acid

[00582] To two separate batches run in parallel each containing a solution of (R)-tert-butyl 2-(4-(4-amino-5-(ethoxycarbonyl)pyrimidin-2-yl)-3-(tiri-buto xycarbonyl)piperazin-1-yl)-

7.8-dihydropyrido[4,3-d]pyrimidine-6(5H)-carboxylate (7 g, 11.97 mmol, 1.0 equiv) in THE (70 mL), EtOH (35 mL) and H 2 O (35 mL) was added LiOH*H 2 O (2.01 g, 47.89 mmol, 4.0 equiv). The mixtures were stirred at 60 °C for 3 h, at which point the two reaction mixtures were combined, and were adjusted to pH=7 with 1 N HQ. The mixture was concentrated under reduced pressure to remove THF and EtOH, filtered, and the residue was dried under reduced pressure. The residue was stirred in MTBE (100 mL) for 10 min, filtered, and the residue was dried under reduced pressure to afford the desired product (8.02 g, 55.1% yield) as a white solid. LCMS (ESI) m/z [M + H] calcd for C 26 H 36 Ns0 6 :557.29; found 557.3.

Building block AA. ferf-butyl (2S)-4-{6-[(ieri-butoxy)carbonyl]-5H,6H,7H,8H- pyrido[4,3-d]pyrimidin-2-yl}piperazine-2-carboxylate.

[00583] dliis building block is prepared by a process similar to that for Building block 1 by utilizing ieri-butyl 2-chloro-7,8-dihydropyrido[4,3-d]pyrimidine-6(5H)-carboxylat e and tert- butyl (2S)-piperazine-2-carboxylate. Building block AB. 4-amino -2-[(2S)-2-[(f£rf-butoxy)carbonyl]-4-{6-[(f0rf- butoxy)carbonyl]-5H,6H,7H,8H-pyrido[4,3-d]pyrmndm-2-yl}piper azm-1-yl]pyrimidme- 5-carboxylic add.

[00584] This building block is prepared from Building block AA by a process similar to that for Building block J by utilizing ethyl 4-amino-2-chloropyrimidine-5-carboxylate.

Budding block AC. 4 amino-2 {4-f6-[{teri-butoxy)carbors I ] 5H,6H.7H.8B pyrido[4.3- d] py rimidin-2-yl }piperazin-1-y i)pyrimidine-5-carboxy lie add.

Step 1: Synthesis of tert-butyl 2-(4-(4-amino-5-(ethoxycarhonyl)pyrixnidin-2-yl)piperazin-1- yT-7,8-dihydropyrido[4,3-d]pyrimidine-6(5H)-carboxylate

[00585] To a solution of tert-bulyl 2-(piperazin-1-yl)-7,8-dihydropyrido[4,3-d]pyrimidine- 6(5H)-carboxylate (8.3 g, 25.99 mmol, 1.0 equiv) and ethyl 4-amino-2-chloropyrixnidine-5- carboxylate (5.24 g, 25.99 mmol, 1.0 equiv) in MeCN (100 mL ) was added to K 2 CO 3 (7.18 g, 51.97 mmol, 2.0 equiv). The reaction was stirred at 80 °C for 12 h. The reaction was then cooled to room temperature, DCM (100 mL) was added, and the reaction mixture was stirred for 30 min. The suspension was filtered, and the filter cake was washed with DCM (6 x 100 mL). The filtrate was concentrated under reduced pressure and the residue was triturated with EtOAc (30 mL), filtered and then the filter cake was dried under reduced pressure to afford the desired product (8.7 g, 65.9*% yield) as light yellow solid.

Step 2: Synthesis of 4-amino-2-(4-(6-(feri-butoxycarbonyl)-5,6,7,8-tetrahydropyri do[4,3- d]pyrimidin-2-yl)piperazin-1-yl)pyrimidine-5-carboxylic acid

[00586] To a solution of /erf-butyl 2-(4-(4-amino-5-(ethoxycarbonyl)pyrimidm-2- yl)piperazin-1-yl)-7,8-dihydiOpyrido[4,3-d]pyrimidine-6(5H)- carboxylate (8.7 g, 17.95 mmol, 1.0 equiv) in THF (120 mL), EtOH (60 mL), and H 2 O (60 mL) was added LiOH » H 2 O (1.51 g, 35.91 mmol, 2.0 equiv). The mixture was stirred at 55 °C for 12 h. The reaction mixture was then concentrated under reduced pressure to remove EtOH and THF, and the reaction mixture was adjusted to pH=6 by the addition of 1 N HCl. The precipitate was filtered, and the filter cake was washed with H 2 O (3 x 50 mL) and then dried under reduced pressure to afford the desired product (7 3 g, 89.1% yield) as light yellow solid. LCMS (ESI) m/z: [M + H] calcd for C - d LsNTO.:: 457.23; found 457.2.

Building block AD. 4-amino-2-{4-[5-({[(ierf-butoxy)carboiiyl]amino }methyl)pyriinidin- 2-yl]piperazin-1-yl}pyrmndine-5-carboxylie acid.

Step 1: Synthesis of ethyl 4-amino-2-(4-(5-(((di-feri- hutoxyearhony])antino)methy])pyrirmdin-2-y])piperazin-1-yl)p yrimidine-5-carhoxy]ate

[00587] To a solution of feri-butyl-N-feri-butoxycai¾onyl-N-((2-(piperazin-1- yl)pyrimidin-5-yl)methyl)carbamate (8.3 g, 21.09 mmol, 1.0 equiv) in MeCN (100 mL) was added ethyl 4-amino-2-chloropyrimidine-5-carboxylate (4.04 g, 20.04 mmol, 0.95 equiv) and K 2 CO 3 (8.75 g, 63.28 mmol, 3.0 equiv). The mixture was stirred at 80 °C for 3 h. The reaction was then cooled to room temperature, DCM (150 mL) w'as added, and the reaction mixture was stirred for 30 min. The suspension was filtered, the filter cake was washed with DCM (3 x 100 mL), and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography (0%- 100% EtOAc/petroleum ether) afforded the desired product (8.35 g, 67% yield) as a white solid.

Step 2: Synthesis of 4-amino-2-(4-(5-((( tert-butoxycarbonyl)amino)methyl)pyrimidm-2- y 1 )pi perazin-1- y l)pyrimidine- 5 -c arboxy li e acid

[00588] To a solution of ethyl 4-amino-2-(4-(5-(((di-/er/- butoxycarbonyl)amino)methyl)pyrimidin-2-yl)piperazin-1-yl)py rimidine-5-carboxylate (8.3 g, 14.86 mmol, 1.0 equiv) in H 2 O (70 mL), EtOH (36 mL) and THF (80 mL) was added LiOH*H 2 O (2.49 g, 59.43 mmol, 4.0 equiv). The reaction mixture was stirred at 55 °C for 16 h. The mixture was then concentrated under reduced pressure to remove THF and EtOH. The mixture was diluted with I¾0 (55 mL) and was adjusted to pH=6 by the addition of 1 N HCl. The mixture was filtered, and the filter cake was washed with H 2 O (2 x 20 mL). The solid cake was dried under reduced pressure to afford the desired product (5.5 g, 84% yield) as a white solid. LCMS (ESI) rn/z [M + H] calcd for CwfeNsCL: 431.22; found 431.4.

Building block AE , 4-amino-2-[(21?)-4-{6-[(ieri-butoxy)carbonyl]-5H,6H,7H,8H- pyrido[4,3-d]pyrimidm-2-yl}-2-(hydroxymethyl)piperazin-1-yI] pyrimidine-5-carbox Iic add.

Step 1: Synthesis of (R)-tert-butyl 2-(4-(4-amino-5-(ethoxycarhonyl)pyrimidin-2-yl)-3- (((/eri-buty ldiphenylsilyl)oxy )methyl)piperazin-1-yl)-7 , 8-dihydropyrido [4,3 -d ] pyrimidine- 6(5H)-carboxylate

[00589] To a solution of (R)-tert-butyl 2-(3-(((/er/-hutyldiphenylsilyl)oxy)methyI) piperazin-1-yl)-7,8-dihydropyrido[4,3-d]pyrimidine-6(5H)-car boxylate (17.2 g, 29.26 mmol, 1.0 equiv) in MeCN (200 mL) was added K 2 CO 3 (12.13 g, 87.78 mmol, 3.0 equiv) and ethyl 4-amino-2-chloropyrimidine-5-carboxylate (6.37 g, 31.60 mmol, 1.08 equiv). The mixture was stirred at 80 °C for 18 h. The reaction mixture was then cooled to room temperature filtered and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography (0%→33% EtOAc/petroleum ether) afforded the desired product (20.3 g, 90.6% yield) as a white solid. LCMS (ESI) rn/z: [ + H] calcd for CLoifeNsOsSi: 753.39; found 753.4.

Step 2: Synthesis of(R)-4-amino-2-(4-(6-(r -butoxycarbonyl)-5,6,7,8 -tetrahydropyrido[4,3- d]pyrimidin-2-yl)-2-(hydroxymetbyl)piperazin-1-yl)pyrimidine -5-carboxylic acid

[00590] To a solution of ( R)- ter i-hutyl 2-(4-(4-amino-5-(ethoxycarbonyl)pyrimidin-2-yl)- 3-(((f -hutyldiphenylsilyl)oxy)methyl)piperazin-1-yI)-7,8-dihydropy rido[4,3-d]pyrimidine- 6(5H)-carboxylate (20.3 g, 26.96 mmol, 1.0 equiv) in THF (200 mL) was added TBAF (1.0 M, 50.75 mL, 1.9 equiv). The reaction mixture was stirred at room temperature for 5 h. The mixture was then poured into I¾0 (200 mL) and the aqueous phase was extracted with EtOAc (2 x 150 mL). The combined organic phases were washed with brine (2 x 100 mL ), dried, filtered and concentrated under reduced pressure. Purification by silica gel chromatography (0%→20% EtOAc/petroleum ether) afforded the desired product (12 g, 85 7% yield) as a white solid. LCMS (ESI) m/z: [M + H] calcd for C 2 4H34N8O5: 515.28; found 515.4.

Step 3: Synthesis of (R )-4-amino-2-(4-(6-(ieri-butoxycarbonyl)-5,6,7,8 -tetrahydropyrido[4,3- d]pyrimidin-2-yl)-2-(hydroxymethyl)piperazm-1-yl)pyrimidine- 5-carboxylic acid

[00591] To a solution of(R)-4-amino-2-(4-(6-(/e/7-biitoxycarbonyl)-5,6,7,8- tetrahydiOpyrido[4,3-d]pyrimidin-2-yl)-2-(hydroxymethyl)pipe razm-1-yl)pyrimidine-5- carboxylic acid (12 g, 23.32 mmol, 1.0 equiv) in THE (100 mL), EtOH (30 mL), and H 2 O (30 mL) was added LiOH^EbO (5.87 g, 139.92 mmol, 6.0 equiv). The mixture was stirred at 50 °C for 22 h. The mixture was then concentrated under reduced pressure to remove THE and EtOH. The aqueous phase was neutralized with 1 N HC 3 and the resulting precipitate was filtered. The filter cake was washed with H 2 O (50 mL and dried under reduced pressure. The filtrate was extracted with DCM (8 x 60 mL) and the combined organic phases were washed with brine (2 x 50 mL), dried, filtered, and concentrated under reduced pressure. The resulting residue was combined with the initial filter cake and the solid was dissolved in DCM (150 mL) and concentrated under reduced pressure to afford the desired product (9.76 g, 85.2% yield) as a white solid. LCMS (ESI) m/z [M + H] calcd for C 2 2H30N8O5: 487.24; found 487.2.

Building block AF. 4-amino -2-[(2S)-4-{6-[(i<?r#-butoxy)carbonyl]-5H,6H,7H,8H- pyrido[4,3-d]pyriniidin-2-yl}-2-(hydroxymethyl)piperazm-1-yl ]pyrimidine-5-carboxyUc add

[00592] This building block is prepared from Building block O by a process similar to that for Building block J by utilizing ethyl 4-amino-2-chloropyrimidine-5-carboxylate.

1 s o Building block AG. 2-((2-(4-(5-((di-(ieri-butoxycarbonyl)arnmo)methyl)pyrimidin -2- yl)piperazm-I-yl)-2-oxoethyl)(methyl)amino)acetic add.

[00593] To a solution of t -butyl N-i?/t-butoxycarbonyI-N-((2-piperazin-1-ylpyrimidm- 5-yl)methyl)carbamate (4.88 g, 12.39 mmol, 1.0 equiv) in EtOAc (40 mL) was added 4- methylmorpboline-2,6-dione (1.6 g, 12.39 mmol, 1.0 equiv). The reaction was stirred at room temperature for 2 h then reaction mixture was concentrated under reduced pressure to give the crude product. The residue was triturated with EtOAc (15 mL) and filtered to give the produc (5.65 g, 87.2% ' yield) as a white solid. LCMS (ESI) m/z: [M + H] calcd for C 2 4H39N6O7: 523.28; found 523.3.

Building block AH. / ri-butyl N-firf-butoxycarbonyI-N-((2-(4-(3-(2-piperazm-1- ylethoxy)propanoyl)piperazm-1-yl)pyrimidm-5-yl)methyl)carbai iiate.

Step 1: Synthesis of benzyl 4-(2-(3-(feri-butoxy)-3-oxopropoxy)ethyl)piperazine-1- carboxylate

[00594] To a solution of tert-butyl 3-(2-bromoethoxy)propanoate (35 g, 138.27 mmol, 1.0 equiv) and benzyl piperazine-1-carboxylate (31.14 L, 138.27 mmol, 1.0 equiv, HCl) in MeCN (420 mL) was added K 2 CO 3 (57.33 g, 414.80 mmol, 3.0 equiv). The reaction was stirred at 80 °C for 20 h. The reaction mixture was cooled to room temperature and the suspension was filtered. The filter cake was washed with EtOAc (3 x 50 mL) and the combined filtrates were concentrated under reduced pressure to give crude product. The residue was purified by silica gel chromatography (5/1 to 0/1 petroleum ether/EtOAc) to give the product (46 g, 84.8% yield) as a yellow oil.

Step 2: Synthesis of 3-(2-(4-((benzyloxy)carbonyl)piperazin-1-yl)ethoxy)propanoic acid

[00595] A solution of benzyl 4-(2-(3-(ierf-butoxy )-3-oxopropoxy)ethyl)piperazine-1- carboxylate (21 g, 53.50 mmol, 1.0 equiv) in TFA (160 mL) was stirred at room temperature for 2 h and then concentrated under reduced pressure. The residue was purified by silica gel chromatography (1/0 to 4/1 EtOAc/MeOH) to give the product (20.4 g, 84.7% yield) as a yellow oil. LCMS (ESI) m/z: [M + H] calcd for C 1 7H 2 4N 2 O5: 337.18; found 337.1.

Step 3: Synthesis of benzyl 4-(2-(3-(4-(5-(((di-ieri-butoxycarbonyl)amino)methyl)pyritni din- 2-yl)piperazin-1-yl)-3 -oxopropoxy)etliyl)piperazine-1-carboxylate

[00596] To a solution of 3-(2-(4-((benzyloxy)carbonyl)piperazin-1-yl)ethoxy)propanoic acid (20.2 g, 44.85 mmol, 1.0 equiv, TFA) in DCM (500 mL) was added HATH (25.58 g, 67.27 mmol, 1.5 equiv) and DIPEA (17.39 g, 134.55 mmol, 23.44 mL, 3.0 equiv). The reaction was stirred at room temperature for 30 min, and then lerl-butyl N -tert- butoxycarbonyl-N-((2-piperazin-1-ylpyrimidin-5-yl)mefliyl)ca rbamate (14.12 g, 35.88 mmol, 0.8 equiv) was added. The reaction mixture was stirred at for 2 h and then quenched with sat. NH4CI (500 mL). The aqueous phase was extracted with DCM (3 x 300 mL) and the combined organic phase was washed with brine (30 mL), dried with anhydrous Na .'. SO:. filtered and concentrated under reduced pressure to give crude product. The residue was purified by silica gel chromatography (0/1 petroleum ether/EtOAc to 10/1 DCM/MeOH) to give the product (29 g, 90.8% yield) as a yellow oil. LCMS (ESI) m/z [M + IT] calcd for C 3 6H53N7O8: 712.41; found 712.4.

Step 4: Synthesis of / -butyl N-½ri-butoxycarbonyl-N-((2-(4-(3-(2-piperazin-1- ylethoxy)propanoyl)piperazin-1-yl)pyrimidin-5-yI)methyl)carb amate

[00597] To a solution of 4-(2-(3-(4-(5-(((di-ii ?/ t-butoxycarbonyl)amino)methyl)pyrimidin- 2-y])pip era zin-1-yl)-3-oxopropoxy)ethyl)piperazine-1-carboxylate (5 g, 7.02 mmol, 1.0 equiv) in EtOAc (150 mL) was added Pd/C (2 g, 10 wt.%). The suspension was degassed and purged with ¾ and then stirred under IT (30 psi) at 30 °C for 3 h. The suspension was then cooled to roo temperature and filtered through Celite. The filter cake was washed with MeOH (15 x 100 mL) and the combined filtrates were concentrated under reduced pressure to give the product (12 g, 89.9% yield) as a light yellow oil. LCMS (ESI) m/z [M + IT] calcd for C 2 8H47N7O6: 578.37; found 578.5. Building block AI. ethyl 2-(piperazin-1-yl)pyrimidine-5-carboxyIate.

Step 1: Synthesis of ethyl 2-(4-( ri-butoxycarbonyl)piperazin-1-yl)pyrimidine-5-carboxylate

[00598] To a solution of /erf-butyl piperazine-1-carboxy late (11.94 g, 53.59 mmol, 1.0 equiv, HCI) and ethyl 2-ch!oropyrimidine-5-carboxylate (10 g, 53.59 mmol, 1.0 equiv) in MeCN (100 mL) was added K 2 CO 3 (7.41 g, 53.59 mmol, 1.0 equiv). The mixture was stirred at 80 °C for 17 h and then poured into H 2 O (200 mL). The mixture was filtered and the filter cake was washed with H 2 O (80 mL) and dried under reduced pressure to give the product (15.76 g, 82% yield) as a white solid.

Step 2: Synthesis of ethyl 2-(piperazin-1-yi)pyrimidine-5-carboxylate

[00599] To a solution of ethyl 2-(4-(/er/-butoxycarbonyl)piperazin-1-yl)pyrimidine-5- carboxylate (15.7 g, 46.67 mmol, 1.0 equiv) in EtOAc (150 mL) was added HCl/EtOAc (150 mL) at 0 °C. The resulting mixture was stirred at room temperature for 9 h. The reaction mixture was filtered and the filter cake was washed with EtOAc (100 mL). The solid was dried under reduced pressure to give the product (12.55 g, 96% yield, HCI) as a white solid. LCM8 (ESI) m/z [M + H] calcd for C, O C.NAL: 237.14; found 237.3.

Building block AJ. 2-(4-(2-(3-(4-(5-(((di-iiri-butoxycarbonyl)amino)methyl)pyri midm-2- yI)piperazin-1-yl)-3-oxopropexy)ethyl)piperazm-1-yl)pyrimidm e-5-carboxylic add.

Step 1: Synthesis of ethyl 2-(4-(2-(3-(i<eri-butoxy)-3-oxopropoxy)ethyl)piperazin-1- yl)pyrimidine-5-carboxylate [00600] To a solution of ethyl 2-piperazin-1-ylpyrimidine-5-carboxylate (17.92 g, 75.85 mmol, 1.2 equiv) and crt-butyl 3-(2-bromoetboxy)propanoate (16 g, 63.21 mmol, 1.0 equiv) in MeCN (200 mL) was added K 2 CO 3 (17.47 g, 126.42 mmol, 2.0 equiv). The reaction was stirred at 80 °C for 12 h and then the reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The crude product was suspended in petroleum ether (200 mL) and stirred for 20 min at 0 °C and then filtered. The solid was dried under reduced pressure to give the product (19.4 g, 75.1% yield) as a yellow' solid.

Step 2: Synthesis of 3-(2-(4-(5-(ethoxycarbonyl)pyrimidin-2-yl)piperazin-1- yl)ethoxy)propanoic acid

[00601] A solution of ethyl 2-(4-(2-(3-(¾r/-butoxy )-3-oxopropoxy)etbyl)piperazin-1- yl)pyrimidine-5-carboxylate (19.4 g, 47.49 mmol, 1.0 equiv) in TFA (200 mL) was stirred at room temperature for 30 min. The reaction mixture was then concentrated under reduced pressure and the residue was purified by silica gel chromatography (50/1 to 1/1 EtOAc/MeOH) to give the product (18 g, 81.3% yield) as a yellow' oil.

Step 3: Synthesis of ethyl 2-(4-(2-(3-(4-(5-(((di-i <? ri- butoxycarbonyl)amino)methyl)pyiimidin-2-yl)piperazin-1-yl)-3 -oxopropoxy)ethyl)piperazin- 1-y l)pyrimidine- 5 -c arboxy la te

[00602] To a solution of 3-(2-(4-(5-(ethoxycarbonyl)pyrimidin-2-yl)piperazin-1- yl)ethoxy)propanoic acid (13 g, 27.87 mmol, 1.0 equiv) in DCM (200 mL) was added HATU (15.90 g, 41.81 mmol, 1.5 equiv) and DIPEA (19.42 mL, 111.49 mmol, 4.0 equiv). The reaction was then stirred at room temperature for 30 min and then tert-butyl N-tert- butoxycarbonyl-N-[(2-piperazin-1-ylpyrimidin-5-yl)methyl]car bamate (10.97 g, 27.87 mmol, 1.0 equiv) was added. The mixture was stirred at for 2 h and then poured into a sat. NH4CI solution (200 mL). The aqueous phase was extracted with DCM (2 x 200 mL) and the combined organic phase was washed with brine (2 x 20 mL), dried with anhydrous Na SCL, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (100/1 to 9/1 EtOAc/MeOH) to give the product (17 g, 79.0% yield) as a yellow oil.

Step 4: Synthesis of 2-(4-(2-(3-(4-(5-(((di-ii ? ri-butoxycarbonyl)amino)methyl)pyrimidin-2- yl)piperazin-1-yl)-3-oxopropoxy)ethyl)piperazin-1-yl)pyrimid ine-5-carboxylic acid

[00603] To a solution of ethyl 2-(4-(2-(3-(4-(5-(((di-teri- butoxycarbonyl)amino)methyl)pyrimidin-2-yl)piperazin-1-yl)-3 -oxopropoxy)ethyl)piperazin- 1-yl)pyrimidine-5-carboxylate (11 g, 15.11 mmol, 1.0 equiv) in THF (40 mL), EtOH (10 mL), and H 2 O (20 mL) was added LiOH^HiO (1.27 g, 30.23 mmol, 2.0 equiv). The mixture was then stirred at 35 °C for 1.5 h. The reaction mixture was extracted with EtOAc (30 mL) and the aqueous phase was adjusted to pH = 7 by addition of HCl (1 N). The mixture was then concentrated under reduced pressure. The crude product was purified by reversed-phase chromatography (20/1 to 3/1 H 2 O/MeCN) to give the product (6.1 g, 67.3% yield) as a white solid. LCMS (ESI) m/z : [M + H] calcd for C 3 3H49N9O8: 700.38; found 700.4.

Building block AK. 2-(4-(2-(3-(4-(5-(((ii?ri-butoxycarbonyl)amino)methyl)pyriim din-2- yl)piperazm-1-yI)-3-oxopropoxy)ethyl)piperazin-1-yl)pyrimidi ne-5-carboxyIic acid.

[00604] A solution of ethyl 2-(4-(2-(3-(4-(5-(((di-tert- hidox yearhony 1 )amino)methyl )py riimdin-2-y 1 )pi perazin-1-yl)-3 -oxopropox y )ethyl)piperazin - 1-yl)pyrimidine-5-carboxylate (5.4 g, 7.42 mmol, 1.0 equiv) in THF (40 mL), EtOH (10 mL), and H 2 O (10 mL) was added LiOIMLO (933.92 g, 22.26 mmol, 3.0 equiv). The mixture was then stirred at 30 °C for 12 h. The reaction mixture was then extracted with EtOAc (2 x 50 mL) and the aqueous phase was adjusted to pH = 7 by addition of HCl (1 N). The solution was then concentrated under reduced pressure. The crude product was purified by reversed- phase chromatography (20/1 to 3/1 EhG/MeCN) to give the product (1.01 g, 22.5% yield) as a white solid. LCMS (ESI) m/z [M + H] calcd for C 2 8H41N9O6: 600.33; found 600.2.

Building block AL. 4-{4-[2-(3-{4-[5-({[(iiri-butoxy)earbonyl]amino}methyl)pyrim idin-2- yI]piperazm-1-yl}-3-oxopropoxy)ethyl]piperazin-1-yl}-4-oxobu tanoic add.

[00605] To a solution of ten-butyl N-ieri-butoxycarbonyl-N-((2-(4-(3-(2-piperazin-1- ylethoxy)propanoyl)piperazin-1-yl)pyrimidin-5-yl)methyI)carb amate (1.0 equiv) in DCM is added succinic anhydride (1.2 equiv) and EtsN (2.0 equiv). The reaction is stirred at room temperature until consumption of starting material, as determined by LCMS analysis. The reaction mixture is then concentrated under reduced pressure to give the crude product. The residue is purified by silica gel chromatography to afford the product.

Building block AM. 2-(4-(4-(4-(5-(((i<?ri-butoxycarbonyl)amino)rnethyl)pyrim idm-2- yl)piperazin-1-yl)-4-oxobutyl)piperazm-1-yl)pyrimidme-S-carb oxyBc add.

Step I : Synthesis of ethyl 2-(4-(4-(/¾r/-butoxy)-4-oxobutyl)piperazin-1-yl)pyrirmdine- 5- carboxylate

[00606] To a solution of ethyl 2-(piperazin-1-yl)pyrimidine- 5 -carboxylate hydrochloride (10 g, 36.67 mmol, 1 0 equiv, HCl) and fenf-butyl 4-bromobutanoate (8.18 g, 36.67 mmol, 1.0 equiv) in DMF (100 mL) was added EtsN (15.31 mL, 110.00 mmol, 3.0 equiv). The mixture was stirred at 130 °C for 14 h. The mixture was then poured into H 2 O (400 mL) and the solution was extracted with EtOAc (3 x 150 L). The combined organic layer was washed with brine (200 mL), dried over NaiSCb and concentrated under reduced pressure. The residue was purified by silica gel chromatography (5/1 to 1/1 petroleum ether/EtGAc) to give the product (9.5 g 68.5% yield) as a yellow solid. LCMS (ESI) m/z: [M + H] calcd for C 1 9H 3 0N4O4: 379.24; found 379.2, 380.2.

Step 2: Synthesis of 4-(4-(5-(ethoxycarbonyl)pyrimidin-2-yl)piperazin-1-yl)butano ic acid hydrochloride

[00607] To a solution of ethyl 2-(4-(4-(ter -butoxy)-4-oxobutyl)piperazin-1-yl)pyrimidine- 5-carboxylate (9.5 g, 25.10 mmol, 1.0 equiv) in EtOAc (100 mL) was added HCl/EtOAe (500 mL). The mixture was stirred at room temperature for 10 h and then the solution was concentrated under reduced pressure to give the product (9.6 g, 96.8% yield) as a white solid. LCMS (ESI) /z [M + H] calcd for C 1 5H 2 2N4O4: 323.17; found 323.2. Step 3: Synthesis of ethyl 2-(4-(4-(4-(5-(((di-½r/-butoxycarbonyl)amino)methyl)pyrimid in-2- y])piperazin-1-yl)-4-oxobutyl)piperazin-1-yl)pyrimidine-5-ca rboxylie acid

[00608] To a solution of 4-(4-(5-(etiioxycarbonyl)pyrimidin-2-yl)piperazin-1-yl)butan oic acid hydrochloride (5 g, 15.51 mmol, 1.0 equiv) and feri-butyl N-½r/-butoxycarbonyl-N-((2- piperazin-i-ylpyrimidin-5-yl)methyl)carbamate (6.10 g, 15.51 mmol, 1.0 equiv) In DMF (150 mL) was added DIPEA (8.11 mL , 46.53 mmol, 3.0 equiv) and HATU (7.08 g, 18.61 mmol, 1.2 equiv). The mixture was stirred at room temperature for 3 h and then the solution was poured into H 2 O (600 mL). The aqueous layer was extracted with EtOAc (3 x 200 mL) and then the combined organic layer was washed with brine (100 mL), dried with IS a . ’SC).: and concentrated under reduced pressure. The residue was purified by silica gel chromatography (50/1 to 15/1 DCM/MeOH) to give the product (6.3 g, 58.2% yield) as a yellow solid. LCM8 (ESI) m/z: [M + H] calcd for (AH , ; NAT: 698.40; found 698.6.

Step 4: Synthesis of 2-(4-(4-(4-(5-(((ieri-butoxycarbonyl)arnino)xnethyl)pyrimidi n-2- yl)piperazin-i-yl)-4-oxobutyl)piperazin-i-yl)pyrimidine-5-ca rboxylic acid

[00609] To a solution of ethyl 2-(4-(4-(4-(5-((bis(½n- butoxycarbonyl)amino)methyl)pyrimidin-2-yl)piperazin-1-yl)-4 -oxo-butyl)piperazin-1- yl)pyrimidine-5-carboxylate (4.5 g, 6.45 mmol, 1.0 equiv) in EtOH (7 mL) and THF (28 mL) was added a solution of LiOIMIaO (541.17 mg, 12.90 mmol, 2.0 equiv) in H 2 O (7 mL). The mixture was stirred at 30 °C for 8 h, then additional LiOH H 2 O (541 mg, 12.90 mmol, 2.0 equiv) was added. After stirring for an additional 8 h at 30 °C, the solution was concentrated under reduced pressure. H 2 O (20 mL) was added and solution was adjusted to pH 3 with IN HCl. The suspension was filtered and the solid dried under reduced pressure to give the product (3.2 g, 79.1% yield) as a white solid. LCM8 (ESI) m/z: [M + H] calcd for C 2 7H 3 9N9O5: 570.32; found 570.3.

Building Block AN. 2-(4-(2-(2-(4-(6-(teri-butoxycarbonyI)-5,6,7,8-tetrahydropyr ido[4,3- d]pyrimidm-2-yl)piperazin-1-yl)ethoxy)ethyl)piperazm-1-yl)py rimidme-5-carboxyIic add.

Step 1: Synthesis of benzyl 4-(2-hydiOxyethyl)piperazine-1-carboxylate

[00610] To a solution of benzyl piperazine-1-carboxy late hydrochloride (41.09 g, 160.04 mmol, 1.0 equiv, HCI) in MeCN (200 rnL) was added K 2 CO 3 (66 36 g, 480.13 mmol, 3.0 equiv) and 2-bromoethanol (20 g, 160.04 mmol, 1.0 equiv). The reaction mixture was stirred at 80 C C for 16 h, at which point it was cooled to room temperature and filtered. The filter cake was washed with EtOAc (100 mL ) and the filtrate then washed with H 2 O (100 mL). The aqueous phase was extracted with EtOAc (3 x 50 mL) and the combined organic phases were washed with brine (50 mL), dried, and concentrated under reduced pressure. Purification by silica gel chromatography (5→25% MeOH/EtOAc) afforded the desired product as a yellow solid (20 g, 47% yield). LCMS (ESI) m/z: [M + H] calcd for C 1 4H 2 0N2O 3 : 265.16; found 264.9.

Step 2: Synthesis of tert-butyl 4-(2-hydroxyethyl)piperazine-1-carboxylate m s: [00611] To a solution of tert-hutyl piperazine-1-carboxy late (198.72 g, 1.07 mol, 1.0 equiv) in MeCN (1500 mL) was added 2-bromoethanol (240 g, 1.92 mol, 1.8 equiv) and K 2 CO 3 (221.19 g, 1.60 mol, 1.5 equiv). The reaction mixture was stirred at 80 °C for 16 h, at which point the mixture was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography (0→14% MeOH/EtOAc) afforded the desired product as a white solid (146 g, 59% yield).

Step 3: Synthesis of trn-butyl 4-(2-bromoethyl)piperazine-1-carboxylate

[00612] To a solution of tert-b\xiy\ 4-(2-hydroxyethyl)piperazine-1-carboxylate (45 g, 195.39 mmol, 1.0 equiv) in THF (600 mL) was added triphenylphosphine (97.38 g, 371.25 mmol, 1.9 equiv) and CBI * 4 (116.64 g, 351.71 mmol, 1.8 equiv). The mixture was stirred at room temperature for 3 h. Two separate batches were combined, and the reaction mixture was filtered, and the filtrate concentrated under reduced pressure. Purification by silica gel chromatography (1→25% EtOAc/petroleum ether) afforded the desired product as a light- yellow solid (31 g, 27% yield).

Step 4: Synthesis of benzyl 4-(2-(2-(4-(ieri-butoxycarbonyl)piperazin-1- yl)ethoxy)ethyl)piperazine-1-carboxylate

[00613] To a solution of benzyl 4-(2-hydroxyethyl)piperazine-1-carboxylate (18 g, 68.10 mmol, 1.0 equiv) in toluene (200 mL) was added NaNfL (26.57 g, 680.99 mmol, 10.0 equiv). tert-Butyl 4-(2-bromoethyl)piperazine-1-carboxylate (25 g, 85.27 mmol, 1.25 equiv) was added and the mixture was heated to 90 °C for 18 h. The mixture was cooled to room temperature and poured into H 2 O (700 mL) at 0 °C. The aqueous phase was extracted with EtOAc (3 x 240 mL) and the combined organic phases were washed successively with H 2 O (350 mL) and sat. brine (2 x 200 mL), dried, filtered, and concentrated under reduced pressure. Purification by silica gel chromatography (0→12% MeOH/EtOAc) afforded the desired product as a light-yellow oil (20 g, 62% yield).

Step 5: Synthesis of teri-butyl 4-(2-(2-(piperazin-1-yl)ethoxy)ethyl)piperazine-1-carboxylat e

[§§614] To a solution of benzyl 4-(2-(2-(4-(/er/-butoxycarbonyl)piperazin-1- yl)ethoxy)ethyl)piperazine-1-carboxylate (20 g, 41.96 mmol, 1.0 equiv) in EtOAc (180 mL) was added Pd/C (8 g, 10 wt.%). The suspension was degassed under reduced pressure and purged with ¾ three times. The mixture was stirred under ¾ (30 psi) at 35 °C for 12 h. The reaction mixture was then filtered, and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography (0→100% MeOH/EtOAc) afforded the desired product as a colorless oil (10.8 g, 75% yield).

Step 6: Synthesis of ethyl 2-(4-(2-(2-(4-(feri-butoxycarbonyl)piperazin-1-yl)etihoxy)- ethyl)piperazin-1-yl)pyrimidine-5-carboxylate

[00615] To a solution of feri-butyl 4-(2-(2-(piperazin-1-yl)ethoxy)ethyI)piperazine-1- carboxylate (10.8 g, 31.54 mmol, 1.0 equiv) in MeCN (100 mL) was added K 2 CO 3 (13.08 g, 94.61 mmol, 3.0 equiv) and ethyl 2-chloropyrimidine-5-carboxylate (5.88 g, 31.54 mmol, 1.0 equiv). The mixture was stirred at 80 °C for 12 h, at which point the reaction was cooled to room temperature, filtered, and the filtrate concentrated under reduced pressure. Purification by silica gel chromatography (0→ 9% MeOH/DCM) afforded the desired product as a white solid (13.6 g, 85% yield).

Step 7: Synthesis of 2-(4-(2-(2-(4-(6-(teri-butoxycarbonyl)-5,6,7,8-tetrahydropyr ido-[4,3- d]pyrimidin-2-yl)piperazin-1-yl)ethoxy)ethyl)pipera in-1-yl)pyrimidine-5-carboxylic acid

[00616] To a solution of ethyl 2-(4-(2-(2-(4-(tert-butoxycarbonyl)piperazin-1- yl)ethoxy)ethyl)piperazin-1-yl)pyrimidine-5-carboxylate (13.6 g, 27.61 mmol, 1.0 equiv) in MeOH (50 mL) was added a solution of HCl in MeOH (4 M, 150 mL, 21.7 equiv). The reaction was stirred at room temperature for 4 h, at which point the mixture was concentrated under reduced pressure to afford the crude desired product as a white solid (13.8 g, 4HCl) that was taken directly onto the next step. LCMS (ESI) m/z [M + H] calcd for C 1 9H32N6O 3 : 393.26; found 393.3.

Step 8: Synthesis of tert-butyl 2-(4-(2-(2-(4-(5-(ethoxycarbonyl)pyiimidin-2-yl)-piperazin-1 - yl)ethoxy)ethyl)piperazin-1-yl)-7,8-dihydropyrido[4,3-d]pyri midine-6(5H)-carboxylate

[00617] To a stirred solution of 2-(4-(2-(2-(4-(6-(/er/-butoxycarbonyl)-5, 6,7,8- tetrahydropyrido[4,3-d]pyrimidin-2-yl)piperazin-1-yl)ethoxy) ethyl)piperazin-1- yl)pyrimidine-5-carboxylic acid (10.2 g, 18.95 mmol, 1.0 equiv, 4HCl) and DIPEA (16.50 mL, 94.74 mmol, 5.0 equiv) in DMF (100 mL) was added feri-butyl 2-chloro-7,8- dihydropyrido[4,3-d]pyrimidine-6(5H)-carboxylate (5.11 g, 18.95 mmol, 1.0 equiv). The reaction mixture was stirred at 90 °C for 12 h. The reaction mixture was then cooled to room temperature and added to EtOAc (200 mL) and H 2 O (400 mL). The aqueous phase was extracted with EtOAc (2 x 100 mL) and the combined organic phases were washed with aqueous NH4CI (4 x 100 mL), brine (2 x 100 mL), dried, filtered and concentrated under reduced pressure. Purification by silica gel chromatography (0→9% MeOH/DCM) afforded the desired product as a white solid (5.4 g, 45% yield). LCMS (ESI) m/z: [M + H] calcd for C 3 1H47N9O5: 626.38; found 626.3.

Step 9: Synthesis of 2-(4-(2-(2-(4-(6-(i<2ri-butoxycarbonyl)-5,6,7,8-tetrahydr opyrido[4,3- d]pyrimidin-2-yl)piperazin-1-yl)etihoxy)etihyl)piperazin-1-y l)pyrimidine-5-carboxylic acid

[00618] To a solution of t -butyl 2-(4-(2-(2-(4-(5-(ethoxycarbonyl)pyrimidin-2- yl)piperazin-1-yl)ethoxy)ethyl)piperazin-1-yl)-7,8-dihydropy rido[4,3-d]pyrimidine-6(5H)- carboxylate (5.4 g, 8.63 mmol, 1.0 equiv) in THF (50 mL), EtOH (20 mL), and H 2 O (20 mL) was added EΐOH·H 2 q (1.09 g, 25.89 mmol, 3.0 equiv). The reaction mixture was stirred at 35 °C for 12 h, at which point the mixture was concentrated under reduced pressure to remove THF and EtOH. The aqueous phase was neutralized to pH = 7 with 0.5N HCl and concentrated under reduced pressure. Purification by reverse phase chromatography afforded the desired product as a white solid (4.72 g, 92% yield). LCMS (ESI) m/z [M + H] calcd for C 2 9H43N9O5: 598.35; found 598.3.

Building block AO. r-[(tert -butoxy)carbonyl]-[1,4'-bipiperidine]-4-carboxylic add.

[00619] At the time of this application this building block was commercially available

(CAS # 201810-59-5).

Building block AP. 2-((2-(piperazm-1-yl)pyriimdm-5-yl)inethyI)isoindoline-1,3-d ione hydrochloride salt.

Step 1: Synthesis of 2-chloro-5-(dibromometbyl)pyrimidine [00620] To a solution of 2-chloro-5-methylpyrimidine (100 g, 777.85 mmol, 1.0 equiv) in CC (1200 rnL) was added NBS (304.58 g, 1.71 mol, 2.2 equiv) and AIBN (51.09 g, 311.14 mmol, 0.4 equiv). The mixture was stirred at 80 °C for 16 h. The reaction solution was then cooled to room temperature, filtered, and the filtrate was poured into H 2 O (1500 mL). The solution was diluted with DCM (3 x 250 mL) and the organic layer washed with brine (300 mL), dried with anhydrous Na2SC , filtered and concentrated under reduced pressure to give the crude product as a brown oil, which was used directly in the next step.

Step 2: Synthesis of 5-(bromomethyl)-2-chloropyrixnidine

[00621] To a solution of 2-chloro-5-(dibromomethyl)pyrimidine (229 g, 799.72 mmol, 1.0 equiv) in THF (600 L) was added DIPEA (111.44 mL, 639.77 mmol, 0.8 equiv) and 1- ethoxyphosphonoyloxyethane (82.57 mL, 639.77 mmol, 0.8 equiv). The mixture was stirred at room temperature for 19 h. The mixture was then poured into H 2 O (1200 rnL) and the aqueous phase was extracted with EtOAc (3 x 300 mL). The combined organic phase was washed with brine (300 mL), dried with anhydrous Na SOi, filtered and concentrated under reduced pressure. The residue was purified b silica gel chromatography (1/0 to 0/1 petroleum ether/EtOAc) to give the product as a brown oil, which was used directly for the next step.

Step 3: Synthesis of 2-((2-chloropyrimidin-5-yl)methyl)isoindoline-1,3-dione

[00622] To a mixture of isoindoline-1,3-dione (15 g, 101.95 mmol, 1.0 equiv) in DMF (126 mL) was added Nall (4.89 g, 122.34 mmol, 60 wt.%, 1.2 equiv) a 0 °C. The mixture was stirred at 0 °C for 30 min, then a solution of 5-(bromomethyl)-2-chloro-pyrimidine (30.21 g, 101.95 mmol, 1.0 equiv) in DMF (24 mL) was added dropwise to the above mixture at room temperature. The mixture was stirred at room temperature for 2 h and was then cooled to 0 °C and quenched with sat. NH + Cl (600 mL). The suspension was filtered and the solid dried under reduced pressure to give the crude product (27.4 g, 98.2% yield) as a grey- solid, which was used directly in the next step. LCMS (ESI) m/'z: [M + H] ealed for C 1 3H8CIN3O 2 : 274.04; found 274.0.

Step 4: Synthesis of tert-butyl 4-(5-((1,3-dioxoisoindolm-2-yl)metbyi)pyrimidin-2- yl)piperazine-1-carboxylate

[00623] To a solution of 2-((2-chloropyrimidin-5-yl)methyl)isoindoline-1,3-dione (27 g, 98.66 mmol, 1.0 equiv) and tert-butyl piperazine-1-carboxylate (20.21 g, 108.52 mmol, 1.1 equiv) in DMF (270 mL) was added K 2 CO 3 (34.09 g, 246.64 mmol, 2.5 equiv). The mixture

1 o was stirred at 80 °C for 3 h and then the reaction was cooled to room temperature and poured into H 2 O (1200 mL). The suspension was filtered and the solid was dried under reduced pressure to give the crude product (35.58 g, 85.2% yield) as a white solid, which was used directly in the next step.

Step 5: Synthesis of 2-((2-(piperazin-1-yl)pyrimidin-5-yl)methyl)i soindoline-1 ,3-dione

[00624] A solution of iert-butyl 4-(5-((l ,3 lioxoisoindolin-2-yl)methyl)pyrmiidin-2- yl)piperazine-1-carboxyl ate (15 g, 35.42 mmol, 1 equiv) in HCl/EtOAc (150 mL) was stirred at room temperature for 2 h. The mixture was filtered and then the filter cake was washed with EtOAc (20 mL) and dried under reduced pressure to give the product (42.53 g, 92.5% yield) as a white solid.

Building block AQ. 2-[(2-{4-[2-(3-{4-[5-({bis[(ferf- butoxy)carbonyl]amino}methyl)pyrimidin-2-yl]piperazm-1-yl}-3 - oxopropoxy)ethyl]piperazm-I-yl}-2-oxoethyl)(methyl)amino]ace tic add.

[00625] To a solution of tert-\miy\ N-[(teri-butoxy)carbonyl]-N-{ [2-(4-{3-[2-(piperazin-1- yl)ethoxy]propanoyl}piperazin-1-yl)pyrimidin-5-yi]methyl [carbamate (300 mg, 519 μmol, 1.0 equiv) in pyridine (8 mL) at 0 °C was added 4-methylmorpholine-2,6-dione (80.3 mg,

622 μmol, 1.2 equiv). The reaction mixture was stirred at 0 °C for 1 li and then warmed to room temperature and stirred for an additional 12 h. The solvent was concentrated under reduced pressure and the solid was partitioned between DCM and H 2 O. The organic layer was separated, dried over MgSOr and the solvent was concentrated under reduced pressure to give the product (23.0 mg, 6.28% yield) LCMS (ESI) m/z: [M + H] ealcd for C 3 3H54NSO9: 707.41; found 707.4. Building Blnck AR. 2-(4-(2-(3-(4-(6-(teri-butoxycarbonyl)-5,6,7,8-tetrahydropyr ido[4,3- d]pyrimidm-2-yl)piperazm-1-yl)-3-oxopropoxy)ethyl)piperazin- 1-yl)pyrmndine-5- carhoxylic add.

Step 1: Synthesis of tert-butyl 2-(4-(3-(2-(4-(5-(ethoxycarbonyl)pyrimidin-2-yl)piperazin-1- yl)ethoxy)propanoyl)piperazin-1-yl)-7,8-dihydropyrido[4,3-d] pyrimidine-6(5H)-carboxylate

[00626] To a solution of 3-(2-(4-(5-(etboxycarbonyl)pyrimidin-2-yl)piperazin-1-yl)eth oxy) propanoic acid (6 g, 12.86 mmol, 1.0 equiv, TEA) in DMF (55 mL) was added HATH (6.36 g, 16.72 mmol, 1.3 equiv) and DIPEA (11.20 mL, 64.32 mmol, 5.0 equiv). After 0.5 h, tert- butyl 2-ipipexazin-1-yl)-7 8-dihydropyrido[4,3-d]pyrimidine-6(5H)-carboxylaie (4.11 g,

12.86 mmol, 1.0 equiv) was added. The mixture was stirred for 3 h, at which point it was filtered and the solid cake was dried under reduced pressure to afford the desired product as a white solid (7.5 g, 89% yield). LCMS (ESI) m/z: [M + H] calcd for C 3 2H47N9O6: 654.37; found 654.4.

Step 2: Synthesis of 2-(4-(2-(3-(4-(6-(f<2rf-butoxycarbonyl)-5,6,7,8-tetrahydr opyrido[4,3- d]pyriinidin-2-yl)piperazin-1-yl)-3-oxopropoxy)ethyl)piperaz in-1-yl)pyriinidine-5-carboxylic acid

[00627] To a solution of tert-butyl 2-(4-(3-(2-(4-(5-(ethoxycarbonyl)pyrimidin-2- yl)piperazin-1-yl)ethoxy)propanoyl)piperazin-1-yl)-7,8-dihyd ropyrido[4,3-d]pyrimidme- 6(5H)-carboxylate (7.2 g, 11.01 mmol, 1.0 equiv) in THF (72 mL), EtOH (36 mL) and H 2 O (36 mL) was added Li0H ® H 2 O (1.85 g, 44.05 mmol, 4.0 equiv). The reaction mixture was stirred at room temperature for 2.5 h, at which point the mixture was filtered and the filtrate was concentrated under reduced pressure to remove THF and EtOH. The aqueous phase was neutralized to pH = 7 with IN HCl, and then concentrated under reduced pressure. Purification by reverse phase chromatography (30% MeCN/HbO) afforded the desired product as a white solid (3.85 g, 54% yield). LCMS (ESI) m/z: [M + H] calcd for C 3 0H43N9O6: 626.34; found 626.3.

Building Block AS. 2-(4-(2-(2-(3-(4-(5-((di-i £ rf-butoxycarbonyIamino)methyl)pyrmndhi- 2-yl)piperazm-1-yl)-3-oxo-propoxy)ethoxy)ethyl)piperazin-1-y l)pyriraidine-5-carboxylic add.

Step 1: Synthesis of 3-(2-(2-(4-(5-(ethoxycarbonyl)pyrimidin-2-yl)piperazin- 1 yI)ethoxy)ethoxy)propanoic acid

[00628] A solution of ethyl 2-(4-(2-(2-(3-( tert-butoxy)-3- oxopropoxy)ethoxy)ethyl)piperazm-1-yl)pyrimidine-5-carboxyla te (4 g, 8.84 mmol, 1.0 equiv) in TFA (12.29 mL, 166.00 mmol, 18.8 equiv) was stirred at room temperature for 3 h. The reaction mixture was concentrated under reduced pressure. Purification by silica gel chromatography (0 20% MeOH/EtOAc) afforded the desired product as a brown oil (4.35 g, 95% yield, TFA salt).

Step 2: Synthesis of ethyl 2-(4-(2-(2-(3-(4-(5-((bis(terf- butoxycarbonyl)amino)methyl)pyrimidin-2-yl)piperazin-1-yl)-3 - oxopropoxy)ethoxy)ethyl)piperazin-1-yl)pyrimidine-5-carboxyl ate

[00629] To a solution of 3-(2-(2-(4-(5-ethoxycarbonylpyrimidin-2-yl)piperazm-1- yl)ethoxy)ethoxy)propanoic acid (3.8 g, 7.44 mmol, 1.0 equiv, TFA) in DCM (30 mL) was added HATU (4.25 g, 11.17 mmol, 1.5 equiv) and DIPEA (6.48 mL, 37.22 mmol, 5.0 equiv). The reaction was stirred at room temperature for 30 min, and then terf-butyl N-tert- butoxycarbonyl-iY-((2-piperazin-1ylpyrimidin-5-yl)metliyl)ca rbamate (2.93 g, 7.44 mmol, 1.0 equiv) was added. The mixture was stirred at room temperature for 3.5 h, at which point the reaction mixture was concentrated under reduced pressure. Purification by silica gel > chromatography (0→20% MeOH/EtOAc) afforded the desired product as a brown oil (4.14 g, 70% yield).

Step 3: Synthesis of 2-(4-(2-(2-(3-(4-(5-((di-tert-butoxycarbonylamino)methyl)pyr imidin-2- yl)piperazin-1-yl)-3-oxo-propoxy)ethoxy)ethyl)piperazin-1-yl )pyrimidine-5-carboxylic acid

[00630] To a solution of ethyl 2-(4-(2-(2-(3-(4-(5-((bis( tert- butoxycarbony l)amino)methy 1 )pyrimidin-2-y l)piperazin-1-yl)-3-oxo- propoxy)ethoxy)ethyl)piperazin-1-yl)pyrimidine-5-carboxylate (1.4 g, 1.81 mmol, 1.0 equiv) in THE (28 mL), EtOH (14 mL) and H 2 O (14 mL) was added LiOi-MLO (304.44 mg, 7.25 mmol, 4.0 equiv). The mixture was stirred at 40 °C for 30 min, at which point the reaction mixture was concentrated under reduced pressure. Purification by reverse phase chromatography (10→40% MeCN/EbO) afforded the desired product as a yellow solid (500 mg, 43% yield).

Building block AT, 2-{4-[2-(2-{4-[5-({[(ieri-butoxy)carbonyl]amino}methyl)pyrin iidin-2- yl]piperazin-1-yl}ethoxy)ethyl]piperazin-1-yl}pyrimidme-5-ca rboxyIic acid.

Step 1: Synthesis of ethyl 2-(4-(2-(2-(4-(5-(((di-tert- butoxycarbonyl)amino)rnethyl)pyrimidin-2-yl)piperazin-1-yl)e thoxy)ethyl)piperazin-1- yl)pyrimidine-5-carboxylate

[00631] To a solution of ethyl 2-(4-(2-(2-(piperazin-1-yl)ethoxy)ethyl)piperaziii-1- yl)pyrimidine-5-carboxyIate hydrochloride (7.3 g, 13.56 mmol, 1.0 equiv, 4HCl) in DMF (75 mL ) was added DIPEA (14.17 mL, 81.36 mmol, 6.0 equiv) and tert-hutyl-N-tert- butoxycarbonyl-N-[(2-chIoropyrimidin-5-yl)methyl]carbamate (5.59 g, 16.27 mmol, 1.2 equiv). The mixture was stirred at 80 °C for 12 h. The mixture was then cooled to room temperature and poured into HrO (300 mL). The aqueous phase was extracted with EtOAe (3 x 80 mL). The combined organic phases were washed with sat. NH Cl (4 x 80 mL) and brine (150 mL), dried, filtered and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography (0%→17% MeOH/EtOAc) afforded the desired product (7.7 g, 81 1% yield) as a light yellow oil. LCMS (ESI) m/z [ + Na] calcd for C 3 4H53N9O7: 722.40; found 722.4.

Step 2: Synthesis of 2-(4-(2-(2-(4-(5-(((i« ?/ -butoxycarbonyl)amino)methyl)pyrimidin-2- yl)piperazin-1-yl)ethoxy)ethyl)piperazin-1-yl)pyrimidine-5-c arboxylic acid

[00632] To a solution of ethyl 2-(4-(2-(2-(4-(5-(((di-teri- butoxycarbonyl)amino)methyl)pyrimidin-2-yl)piperazin-1-yl)et hoxy)ethyl)piperazin-1- yl)pyrimidine-5-carboxylate (7.7 g, 11.00 mmol, 1.0 equiv) in THE (80 mL ), EtOH (20 mL), and H 2 O (40 mL) was added LiOtbbhO (2.31 g, 55.01 mmol, 5.0 equiv). The mixture was stirred at 50 °C for 26 h. The mixture was then concentrated under reduced pressure to remove THE and EtOH. The aqueous phase was neutralized with 0.5 N HO, and concentrated under reduced pressure. Purification by reverse phase chromatography afforded the desired product (4.67 g, 74.3% yield) as a white solid. LCMS (ESI) m/z: [M - H] calcd for C 2 7H41N9O5: 570.31; found 570.3.

Building block All. (R. )~ieri~\mi \ 4-(5-(((terf-butoxycarbonyl-N-firf- butoxycarbonyl)amino)methyl)pyrimidin-2-yl)piperazine-2-carb oxylate.

Step 1: Synthesis of (R )-1,4-bis((benzyloxy)carbonyl)piperazine-2-carboxylic acid

[00633] To two separate batches containing a solution (2R )-piperazine-2-carboxylic acid (70 g, 344.71 mmol, I equiv, 2HCl) in H 2 O (700 mL) and dioxane (1120 mL) was added 50% aq. NaOH until pH = 11. Benzyl chloroformate (156.82 mL, 1.10 mol, 3.2 equiv) was added and the reaction was stirred at room temperature for 12 h. The two reaction mixtures were combined and H 2 O (1200 L) was added. The aqueous layer was extracted with MTBE (3 x 1000 mL), adjusted to pH = 2 with con. HCl, and then extracted with EtOAc (2 x 1000 mL). The combined organic phases were dried, filtered, and concentrated under reduced ro pressure to afford the desired product (280 g, 86% yield). LCMS (ESI) m/z: [M + H] calcd for C 2 1H 2 2N 2 O6: 399.16; found 399.0.

Step 2: Synthesis of(R)-1 ,4-dibenzyl 2- er -butyl piperaz e-1,2, 4-tricarboxylate

[00634] To a solution of(R)-1 ,4-bis((benzyloxy )carbony l)piperazine-2-carboxy lie acid (70 g, 175.70 mmol, 1.0 equiv) in toluene (700 mL ) at 80 °C was added 1,1-di-ii ? ri-butoxy-iV,A ? - dimethyl-methanamine (80.04 mL, 333.83 mmol, 1.9 equiv). The reaction was stirred at 80 °C for 2 h, at which point it was cooled to room temperature and partitioned between EtOAc (300 mL) and H 2 O (500 mL). The aqueous layer was extracted with EtOAc (2 x 500 mL) and the combined organic layers were dried, filtered, and concentrated under reduced pressure. Purification by silica gel chromatography (0→25 EtOAc/petroleum ether) afforded the desired product as a white solid (50 g, 57% yield). LCMS (ESI) m/z: [M + Na] calcd for C 2 5H 3 0N 2 O : 477.20; found 476.9.

Step 3: Synthesis of (R)-tert-butyl piperazine-2-carboxylate

[00635] To a solution of(R)-1,4-dibenzyl 2-tert-butyl piperazine-1,2, 4-tricarboxylate (50 g, 110.01 mmol, 1 equiv) in EtOAc (20 mL) was added Pd/C (15 g, 10 wt.%). The suspension was degassed under reduced pressure and purged with ¾ three times. The suspension was stirred under ¾ (30 psi) at 30 °C for 4 h. The reaction mixture was then filtered, the residue was washed with MeOH (5 x 200 mL), and the filtrate concentrated under reduced pressure to afford the desired product as a yellow oil (17 g, 81% yield). LCMS (ESI) m/z [M + H] calcd for C9H18N2O 2 : 187.15; found 187.1.

Step 4: Synthesis of (R)-tert-hulyl 4-(5-(((feri-butoxycarbonyl-/V-ieri- butoxycarbonyl)amino)methyl)pyrimidin-2-yl)piperazine-2-carb oxylate

[00636] To a suspension of (R)-tert-hnty\ piperazine-2 -carboxylate (8 g, 23.27 mmol, 1.0 equiv) and tert-bulyfoY-/ -buloxycarbonyl ((2-chloropyrimidin-5-yl)methyl)carbamate (5.20 g, 27.92 mmol, 1.2 equiv) in MeCN (100 mL) was added K 2 CO 3 (6.43 g, 46.54 mmol, 2.0 equiv). The reaction mixture was heated to 80 °C for 12 h, at which point it was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography (0→ lQ0% EtOAc/petroleum ether) afforded the desired product as a yellow solid (9.2 g, 73% yield). LCMS (ESI) m/z:. [M + H] ealed for C.',:s S WN ' ; ( ) {.:4 4.30: found 494.1. Building block AV. (S)-tert-butyl 4-(5-(((ierf-butoxycarbonyl-N-feri- butoxycarbonyl)a ino)methyl)pyrimidin-2-yl)piperazine-2-carboxylate.

[00637] This building block is prepared by a process similar to that for Building block AU by utilizing (2S)-piperazine-2-carboxylic acid.

Building block AW. (J?)-2-(4-(2-(3-(2-(terf-butoxycarbonyl)-4-(5-(((£<?ri-b utoxycarbonyl- A-icri-butoxycarbonyl)amino)methyl)pyrimidin-2-yl)piperazin- 1-yl)-3- oxopropoxy)ethyl)piperazin-1-yI)pyrimidine-5-carboxylic acid.

Step 1: Synthesis of (R )-eihyl 2-(4-(2-(3-(2-(tert-butoxycarbonyl)-4-(5-(((te/f- butoxycarboriyl-yV-tert-butoxycarbony])aimno)methyl)pyrimidi ri-2-y])piperazin-1-yl)-3- oxopropoxy)ethyl)piperazin-1-yl)pyrimidine-5-carboxyIate

[00638] To a solution of (R )-/i?ri-butyl 4-(5-(((ie i-butoxycarbonyl-V-ieri- butoxycarbonyl)amino)methyl)pyrimidin-2-yl)piperazine-2-carb oxylate (5.3 g, 11.36 mmol, 1.0 equiv, TFA) in DCM (80 mL) was added HATH (6.48 g, 17.05 mmol, 1.5 equiv) and DIPEA (7.92 mL, 45.45 mmol, 4.0 equiv). The reaction was stirred at room temperature for 30 min and then tert-butyl (2R )-4-(5-((bis(ieri-butoxycarbonyl)amino)methyl)pyrimidin-2- yl)piperazine-2-carboxylate (5.61 g, 11.36 mmol, 1.0 equiv) was added. The mixture was stirred for 1 h, at which point sat. NILCl (80 mL) was added. The organic phase was washed with sat. NBLCl (5 x 80 mL ), dried, filtered, and concentrated under reduced pressure. Purification by silica gel chromatography (0→ 9% MeOH/EiOAc) afforded the desired product as a yellow solid (8.4 g, 85% yield).

Step 2: Synthesis of(R)-2-(4-(2-(3-(2-(ieri-butoxycarbonyl)-4-(5-(((ieri-butox ycarbonyl-N- /€nf-butoxycarbonyl)amino)metbyl)pyrimidin-2-yl)piperazin- 1-yl)-3- oxopropoxy)ethyl)piperazin-1-yl)pyrimidine-5-carboxylic acid

[00639] To two separate batches containing a solution a solution of(R)-ethyl 2-(4-(2-(3- (2-(i¾r/-butoxycarbonyl)-4-(5-(((/iir/-butoxycarbonyl-Y-ter /- butoxycarbonyl)amino)methyl)pyrimidin-2-yl)piperazin-1-yl)-3 -oxopropoxy)ethyl)piperazin- 1-yl)pyrimidine-5-carboxylate (3.4 g, 4.11 mmol, 1.0 equiv) in THF (16 mL), EtOH (8 mL) and H 2 O (8 mL) was added LiCfflMfcO (344.61 mg, 8.21 mmol, 2.0 equiv). The mixture was stirred at room temperature for 2 h. The two reaction mixtures were then combined and were adjusted to pH = 7 with IN HCl. The solution was concentrated under reduced pressure to remove THF and EtOH. The solution was then filtered, and the resulting solid was purified by reverse phase chromatography to afford the desired product as a white solid (4 g, 59% yield). LCMS (ESI) ni/z [M + H] calcd for C 3 8H57N9O10: 800.43; found 800.3

Biuldiiig block AX. (S) -2-(4-(2-(3-(2-(tert -butoxycarbonyl)-4-(5-(((fi?ri-butoxycarbonyl- A-terf-butoxycarbonyI)amino )methyl)pyrimidin-2-yl)piperazin-1-yl)-3- oxopropoxy)ethyl)piperazin-1-yl)pyrimidine-5-carboxylic add.

[00640] This building block is prepared from Building block AV by a process similar to that for Building block AW. i o o Building block AY, l'-(2-(3-(4-(5-(((feri-butoxycarbonyl)amino)inetfayl)pyrimid m-2-yl) piperazin-1-yl)-3-oxopropoxy)ethyl)-(1,4'-bipiperidine]-4-ca rboxylic add.

Step 1: Synthesis of G-teri-butyl 4-ethyl [ 1 , 4 '-bipiperidine]-1’,4-dicarboxylate

[00641] To a solution of ethyl piperidine-4-carboxylate (30 g, 150.57 mmol, 1.0 equiv) and feri-butyl 4-oxopiperidine-1-carboxylate (23.67 g, 150.57 mmol, 1.0 equiv) in DCM (300 mL) was added HOAc (6.00 mL, 104.95 mmol, 0.7 equiv). The mixture was stirred at room temperature for 30 min, then NaBH(OAc)3 (63.82 g, 301.13 mmol, 2.0 equiv) was added.

The mixture was stirred for 16 h, at which point ¾0 (50 mL) was added. The aqueous phase was extracted with DCM (3 x 15 mL) and the combined organic phases were washed with brine (10 mL), dried, filtered, and concentrated under reduced pressure. Purification by silica gel chromatography (8→100 MeOH/EtOAc) afforded the desired product as a yellow oil (30 g, 59% yield).

Step 2: Synthesis of ethyl [ 1 ,4’-bipiperidine]-4-carboxylate

[00642] To a solution of HCI in EtOAc (200 mL) was added l'-lerl-butyl 4-ethyl [1,4'- bipiperidine]-r,4-dicarboxylate (20 g, 58.74 mmol, 1.0 equiv). The mixture was stirred at room temperature for 3 h. The mixture was then concentrated under reduced pressure to afford the desired crude product as a white solid (15 g, HCi salt).

Step 3: Synthesis of ethyl r-(2-(3-(ierf-butoxy)-3-oxopropoxy)ethyl)-[1,4'-bipiperidine ]-4- carhoxylate r o [00643] To a solution of tert-hutyl 3-(2-bromoethoxy)propanoate (6.46 g, 25.54 mmol, 1.0 equiv) in DMF (240 rnL) was added K 2 CO 3 (10.59 g, 76.61 mmol, 3.0 equiv) and ethyl [1,4'- bipiperidine]-4-carboxylate (8 g, 25.54 mmol, 1.0 equiv, 2HCI). The mixture was stirred at 120 °C for 12 h, at which point the reaction was cooled to room temperature, filtered, the filter cake washed with H 2 O (20 rnL), and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography (O- ll MeOH/EtOAc) afforded the desired product as a yellow oil (6.6 g, 63% yield).

Step 4: Synthesis of 3-(2-(4-(ethoxycarbonyl)-[1,4’-bipiperidin]-r-yl)ethoxy (propanoic acid

[00644] To the solution of HCl in EtOAc (70 mL) was added ethyl l’-(2-(3-(ieri-butoxy)- 3-oxopropoxy) ethyl)-[1,4’-bipiperidine]-4-carboxylate (6.6 g, 16.00 mmol, 1.0 equiv). The mixture was stirred at room temperature for 3 h, at which point the reaction was concentrated under reduced pressure to afford the desired product as a white solid (6.5 g, 95% yield,

2HCl).

Step 5: Synthesis of ethyl l'-(2-(3-(4-(5-(((lV,A-di- tert- buioxycarbonyl)amino)methyl)pyrimidin-2-yl)piperazin-1-yl)-3 -oxopropoxy)ethyl)-[1,4’- bipiperidine] -4-carboxylate

[00645] To a solution of t« ?/ -butyl-ii?/t-butoxycarbonyl((2-(piperazin-1-yI)pyrimidin-5- yl)methyl)carbamate (2.49 g, 6.33 mmol, 1.5 equiv) in DMF (40 L) was added DIPEA (9.74 mL, 55.89 mmol, 6.0 equiv) and HATU (5.31 g, 13.97 mmol, 1.5 equiv). The mixture was stirred at room temperature for 30 min, and then 3-(2-(4-(ethoxycarbonyl)-[ 1 ,4'- bipiperidin]-r-yl)ethoxy) propanoic acid (4 g, 9.32 mmol, 1.0 equiv, 2HCI) was added. The mixture was stirred at for 1.5 h, at which point H 2 O (5 mL) and EtOAc (20 mL) were added. The aqueous phase was extracted with EtOAc (3 x 10 mL) and the combined organic phases were washed with brine (5 mL), dried, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography afforded the desired product as a brown oil (1.6 g, 23% yield). LCMS (ESI) m/z: [M + H] calcd for Ca T HeiNyOs: 732.47; found 732.6.

Step 6: Synthesis of r-(2-(3-(4-(5-(((ieri-butoxycarbonyl)amino)metiiyl)pyrimidin -2- yl spiperazin-1-yl)-3-oxopropoxy)ethyl)-[ 1 ,4'-bipiperidine]-4-carboxylic acid

[00646] To a solution of ethyl r-(2-(3-(4-(5-((( V,/V-di-/er/- butoxycarbonyl)amino)methyl)pyrimidin-2-yl)piperazin-1-yl)-3 -oxopropoxy)ethyl)-[1,4'- bipiperidine]-4-carboxylate (1.4 g, 1.91 mmol, 1.0 equiv) in THF (7.5 mL), EtOH (3.8 mL), and H 2 O (3.8 mL) was added LiOH ® H 2 O (321.07 mg, 7.65 mmol, 4.0 equiv). The mixture was stirred at room temperature for 2 h, at which point the mixture was concentrated under reduced pressure. Purification by reverse phase chromatography (5-- 38% MeCN/HfiO) afforded the desired product as a yellow solid (325 mg, 22% yield). LCMS (ESI) m/z [M + H] calcd for C 3 0H49N7O6: 604.38; found 604.3.

Building block AZ. 1-(4-{2-|2-(2-

{[(benzyloxy)carbonyl]aimno}ethoxy)ethoxy]etfayl}piperazi n-1-yl)-3,6,9,12- tetraoxapentadecan-15-oic acid.

Step i: Synthesis of benzyl (2-(2-(2-bromoethoxy)ethoxy)ethyl)carbamate

[00647] To a solution of benzyl (2-(2-(2-hydroxyethoxy)ethoxy)ethyl)carbamate (10 g, 35.30 mmol, 1.0 equiv) in DCM (300 mL) at 0 C was added PPI13 (13.79 g, 52.59 mmol, 1.49 equiv) and CB¾ (17.44 g, 52.59 mmol, 1.49 equiv). Then the mixture was warmed to room temperature and stirred for 12 h. The reaction mixture was then filtered, and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography (1 % 25% EtOAc/petroleum ether) afforded the desired product (10.8 g, 88.4% yield) as yellow oil.

Step 2: Synthesis of teri-butyl 4-(3-oxo-1-pheny 1-2,7, 10-trioxa-4-azadodecan-12- yl)piperazine-1-carboxylate

[00648] To a solution of benzyl (2-(2-(2-bromoethoxy)ethoxy)ethyl)carbamate (10.8 g, 31.19 mmol, 1.0 equiv) and /erf-butyl piperazine-1-carboxylate (5.81 g, 31.19 mmol, 1.0 equiv) in MeCN (100 mL) was added K 2 CO 3 (4.31 g, 31.19 mmol, 1.0 equiv). The mixture was stirred at 80 °C for 1 h. The reaction mixture was then filtered, and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography (0%→50% MeOH/EtOAc) afforded the desired product (13.1 g, 93.0% yield) as yellow oil.

1 m s Step 3: Synthesis of benzyl (2-(2-(2-(piperazin-1-yl)ethoxy)ethoxy)ethyl)carbamate

[00649] A solution of tert- butyl 4-(3-oxo-1-phenyl-2,7,10-irioxa-4-azadodecan-12- yl)piperazine-1-carhoxylate (5.64 g, 12.49 mmol, 1.0 equiv) in HCl/EtOAc (50 mL, 4 M) was stirred at room temperature for 1 h. The reaction mixture was then concentrated under reduced pressure to afford the desired product (5.23 g, crude, HCl salt) as yellow oil.

Step 4: Synthesis of /eri-butyl 1-(4-(3-oxo-1-phenyl-2,7, 10-trioxa-4-azadodecan-12- yl)piperazin-1-yl)-3 ,6,9, 12-tetraoxapentadecan-15 -oate

[00650] A solution of benzyl (2-(2-(2-(piperazin-1-yl)eihoxy)ethoxy)ethyl)carbantate

(13.3 g, 31.34 mmol, 1.0 equiv, 2HCl) and tert-butyl 1-bromo-3,6,9,12-tetraoxapentadecan- 15-oate in MeCN (150 mL) was added K 2 CO 3 (21.66 g, 156.71 mmol, 5.0 equiv). The mixture was stirred at 80 °C for 12 h. The reaction mixture was then filtered, and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography (1%— >17% MeOH/DCM) afforded the desired product (5.4 g, 26.3% yield) as a yellow' oil.

Step 5 : Synthesis of 1-(4-(3-oxo-1-phenyl-2,7,10-trioxa-4-azadodecan-12-yl)pipera zin-1-yl)- 3,6,9,12-tetraoxapentadecan-15-oic acid

[00651] A solution of tert-butyl 1-(4-(3-oxo-1-phenyl-2,7, 10-lrioxa-4-azadodecan-12- yl)piperazin-1-yl)-3,6,9,12-tetraoxapentadecan-15-oate (2.4 g, 3.66 mmol, 1.0 equiv) in TFA (20 mL) was stirred at room temperature for 30 min. The reaction mixture was then concentrated under reduced pressure to afford the desired product (3.03 g, TFA salt) as yellow oil.

Building Block BA, (R)-2-(2-(f£r6-butoxycarbonyl)-4-(6-(ieri-butoxycarbonyl)-5 ,6,7,8- tetrahydropyrido[4,3-d]pyrimidin-2-yl)piperazin-1-yl)pyriini dine-5-carboxylic acid,

Step 1: Synthesis of (R)-tert-buty] 2-(3-(i<eri-butoxycarbonyl)-4-(5-

(eihoxycarbonyl)pyrimidin-2-yl)piperazin-1-yl)-7,8-dihydr opyrido[4,3-d]pyriinidine-6(5H)- carhoxylate

[00652] To two separate batches run in parallel each containing a solution of (K)-tert-butyl 2-(3-(i< ?/ -butGxycarbonyl)piperazin-1-yl)-7,8-dihydropyrido[4,3-d]pyri midine-6(5H)- carboxylate (6 g, 14.30 mmol, 1.0 equiv) and K 2 CO 3 (3.95 g, 28.60 mmol, 2.0 equiv) in MeCN (80 mL) was added ethyl 2-chloropyrimidine-5-carboxylate (3.20 g, 17.16 mmol, 1.2 equiv). The reaction mixtures were stirred at 80 °C for 12 h. The two reactions mixtures were combined and filtered, the residue was washed with EtOAc (3 x 50 mL), and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography (0%→17% MeOH/EtOAc) afforded the desired product (15 g, 91.5% yield) as a yellow solid. LCMS (ESI) rn/z: [M + H] calcd for C 2 8H 3 9N7O6: 570.31; found 570.1.

Step 2: Synthesis of(R)-2-(2-(tert-butoxycarbonyl)-4-(6-(ter/-butoxycarbonyl)- 5,6,7,8- tetrahydropyrido[4,3-d]pyrimidin-2-yl)piperazin-1-yl)pyrimid me-5-carboxylic acid

[§§653] To a solution of (R )-tert-butyl 2-(3-(ieri-butoxycarbonyl)-4-(5- (ethoxycarbonyl)pyrimidin-2-yl)piperazin-1-yl)-7,8-diliydrop yrido[4,3-d]pyrimidine-6(5H)- carboxylate (15 g, 26.33 mmol, 1.0 equiv) in THE (80 mL), EtOH (40 mL) and H 2 O (40 mL) was added LiOHeEhG (2.21 g, 52.66 mmol, 2.0 equiv). The mixture was stirred at room temperature for 6 h. The reaction mixture was then adjusted to pH=6 with 1 N HCl. The resulting suspension was filtered, and the solid cake was dried under reduced pressure to afford the desired product (10.87 g, 75.9% yield) as a white solid. LCMS (ESI) rn/z: [M + H] calcd for C 2 6H 3 5N7O6: 542.27; found 542.1.

Building Block BB, (S) -2-(2-(ieri-hutoxycarbonyl)-4-(6-(tert -butoxycarbonyI)-5,6,7,8- tetrahydropyrido[4,3-d]pyrimidin-2-yl)piperazm-1-yl)pyrimidm e-5-carboxylic add.

[§§654] This building block is prepared from Building block AA by a process similar to that for Building block BA.

Building Block BC. 2-[(2R )-2-[(i<?ri-butoxy)carbonyl]-4-[5-({[(feri- butoxy)carbonyl]amino}methyI)pyrimidm-2-yI]piperazin-1-yl]py riimdine-5-carboxylic acid. [00655] This building block is prepared from Building block AU by a process similar to that for Building block B A.

Building Block BD. 2-[(2,S)-2-[(ieri-butoxy)carbonyl]-4-[5-({[(ieri- butoxy)carbonyl]amino }methyl)pyrimidin-2-yl]piperazin-1-yl]pyrimidine-5-carboxyli c add.

[00656] This building block is prepared from Building block AV by a process similar to that for Building block BA.

Building block BE. 15-(6-((4-amino -3-(2-amino benzo|d]oxazoI-5-yl)-1H-pyrazolo[3,4- d]pyriimdm-1-yI)metfayl)-3,4-dihydroisoquinoIin-2(1H)-yl)-1- ((lS,4S)-5-(2-(2-(2- aminoethoxy)ethoxy)ethyl)-2,5-diazabicydo[2.2.1]heptan-2-yl) -3,6,9,12- tetraoxapentadecan-15-one.

Step I: Synthesis of (15,45)-ter/-butyl 5-(3-oxo-1-phenyl-2,7, 10-trioxa-4-azadodecan-12-yl)- 2,5-diazabicyclo[2.2.1]heptaiie-2-carboxylate

[00657] To a solution of (lS,45)-/er -butyl 2,5-diazabicyclo[2.2.1]heptane-2-carboxylate (2.85 g, 14.37 mmol, 1.0 equiv) in MeCN (50 mL) was added K 2 CO 3 (3.97 g, 28.75 mmol, 2.0 equiv) and benzyl (2-(2-(2-bromoethoxy)ethoxy)ethyl)carbamate (4.98 g, 14.37 mmol,

1.0 equiv). The mixture was stirred at 80 °C for 24 h. The reaction mixture was then filtered, and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography (0→1()% MeOH/EtOAc) afforded the desired product (6.2 g, 93.0% yield) as colorless oil. LCMS (ESI) m/z [M + H] caicd for C 2 4H37N3O6: 464.27; found 464.2. L Step 2: Synthesis of benzyl (2-(2-(2-((15,45)-2,5-diazabicyclo[2.2.1]heptan-2- yl)ethoxy)ethoxy)etbyI)carbamate

[00658] To a solution of (15,45)-i?ri-butyl 5-(3-oxo-1-phenyl-2,7, 10-trioxa-4-azadodecajn- 12-yl)-2,5-diazabicyclo[2.2. l]heptane-2-carboxylate (6 2 g, 13.37 mmol, 1.0 equiv) in DCM (60 mL) was added TFA (20.7 ml., 279.12 mmol, 20.9 equiv). The reaction was stirred for 2 h, at which point the mixture was concentrated under reduced pressure at 45 °C to afford the desired crude product (10.5 g, 4TFA) as light brown oil, which was used the next step directly. LCMS (ESI) rn/z: [M + H] calcd for C 1 9H 2 9N3O4: 364.22; found 364.2.

Step 3: Synthesis of tert-butyl 1-((15',4S)-5-(3-oxo-1-phenyl-2,7,10-trioxa-4-azadodecan-12- yl)-2,5-diazabicyclo[2.2.1]heptan-2-yl)-3,6,9,12-tetraoxapen tadecan-15-oate

[00659] To a solution of benzyl (2-(2-(2-(( 1 S,4S)-2,5-diazabicyclo[2.2. l]heptan-2- yl)ethoxy) ethoxy)ethyl)carbamate (5 g, 6 10 mmol, 1.0 equiv, 4TFA) in MeCN (80 mL) was added K 2 CO 3 (5.06 g, 36.61 mmol, 6.0 equiv) and ten-butyl 1-bromo-3,6,9,12- tetraoxapentadecan-15-oate (2.35 g, 6.10 mmol, 1.0 equiv). The reaction mixture was stirred at 80 °C for 12 h. The reaction mixture was then filtered, and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography (0→15% MeOH/EtOAc) afforded the desired product (5.2 g, 92.8% yield) as light yellow oil LCMS (ESI) m/z: [M + H] calcd for C 3 4H57N3O10: 668.4; found 668.4.

Step 4: Synthesis of 1-((l£,4S)-5-(3-oxo-1-phenyl-2,7, 10-trioxa-4-azadodecan-12-yl)-2,5- diazabic clo[2.2. l]heptan-2-yl)-3,6,9, 12-tetraoxapentadecan-15-oic acid

[00660] A solution of tert-butyl 1-((lS,4S)-5-(3-oxo-1-phenyl-2,7,10-trioxa-4-azadodecan- 12-yl)-2,5-diazabicyclo[2.2.1]heptan-2-yl)-3,6,9,12-tetraoxa pentadecan-15-oate (5 2 g, 5.66 mmol, 1.0 equiv) in TFA (47.3 mL, 638.27 mmol, 112 75 equiv) was stirred at room temperature for 30 min. The mixture was then concentrated under reduced pressure at 45 °C. Purification by reverse phase chromatography (2 ~ 35% MeCN/EfcC O.OS % NH4OH)) afforded the desired product (1.88 g, 54.3% yield) as light brown oil. LCMS (ESI) m/z. : [M + H] calcd for C 3 0H49N3O10: 612.34; found 612.3.

Building block BF. 21-(6-((4-amino -3-(2-aimnobenzo[d]oxazol-5-yl)-1H-pyrazolo[3,4- d]pyrimidin-1-yl)methyl)-3,4-dihydroisoquinolin-2(1H)-yI)-1- (piperazin-1-yI)-

1 r nc 3,6,9,12,15,18-hexaoxahenicosan-21-one.

Step 1: Synthesis of benzyl 4-(23 ,23-dimethyl-21-oxo-3 ,6,9, 12, 15 , 18,22-heptaoxatetracosyl) piperazine·· 1-carboxylate

[00661] To a solution of mri-butyl 1-bromo-3,6,9,12,15,18-hexaoxahenicosan-21-oate (5 g, 10.56 mmol, 1.0 equiv) and benzyl piperazine-1-carboxylate (2.62 mL, 11.62 mmol, 1.1 equiv HCl) in MeCN (50 mL) was added K 2 CO 3 (4.38 g, 31.69 mmol, 3.0 equiv). The reaction mixture was stirred at 80 °C for 10 h. The mixture was then filtered, the solid cake washed with EtOAc (3 x 3 mL), and the filtrate concentrated under reduced pressure. Purification by silica gel chromatography (0— ->10% MeOH/EtOAc) afforded the desired product (4 g, 61.8% yield) as a red liquid.

Step 2: Synthesis of 1-(4-((benzyloxy)carbonyl)piperazin-1-yl)-3 ,6, 9, 12, 15,18- hexaoxahenicosan-1-oic acid

[00662] To a solution of benzyl 4-(23,23-dimethyl-21-oxo-3,6,9,12,15,18,22- heptaoxatetracosyl)piperazine-1-carboxylate (1.8 g, 2.94 mmol, 1.0 equiv) in DCM(10 mL) was added TFA (10 mL). The solution was stirred for 0.5 h. The solution was then concentrated under reduced pressure. To the residue was added DCM (30 mL) and then the solution was concentrated under reduced pressure to afford the desired product (1.6 g, 2.87 mmol, TFA) as a red liquid.

Preparation of Rapamycin Monomers,

Monomer 1. 40(J?)-O-(4-mtrophenyl)carbonate rapamycin. [00663] To a solution of rapamycin (30.10 g, 32.92 mmol, 1.0 equiv) in DCM (148.9 mL ) was added pyridine (29.6 mL, 367 mmol, 11.1 equiv). The solution was cooled to -78 °C and then p-nitrophenyl chloroformate (12.48 g, 61.92 mmol, 1 .9 equiv) was added. The reaction was stirred at -78 C C for 2 li. To the reaction mixture was then added DCM and the solution was then poured into ¾0. The aqueous layer was extracted with DCM and the combined organic layers were dried over MgSCb, and concentrated under reduced pressure. The crude material was purified by silica gel chromatography (0→ 50% EtOAc/hexanes) to provide the product (23.1 g, 59.2% yield) as a white solid. LCMS (ESI) rn/z: [M + Na] calcd for C58H82N2O17: 1101.55; found 1101.6.

Monomer 2. 32( i)-hydroxy 40(i¾ -O-(4-nitrophenyl)carbonate rapamycin.

Step i: Synthesis of 32(R)-hydroxy rapamycin

[00664] A solution of 32(R )-hydroxy-28,40-bistriethylsilyl rapamycin (3.64 g, 3.18 mmol, 1 equiv) in THE (41.8 mL) was treated with pyridine (20.8 mL, 258 mmol, 81 equiv) and the reaction mixture was cooled to 0 °C. The solution was treated dropwise with 70% HF- pyridine (4.60 mL, 159 mmol, 50 equiv) and the reaction mixture was stirred at 0 °C for 20 min followed by wanning to room temperature. After 5 h, the reaction mixture was cooled hack to 0 °C and carefully added to ice cold sat. NaHCO 3 solution (400 mL). The mixture was extracted with EtOAc (2 x 100 mL) and the organic phases were washed with 75 mL portions of Hi>O, sat. NaHCO 3 solution and brine. The organic solution was dried over Na2SC>4, filtered and concentrated to yield a light yellow oil that produced a stiff foam under reduced pressure. The crude material was purified by silica gel chromatography (2Q→ 40% acetone/hexanes) to yield the desired product as a white amorphous solid (1.66 g, 57% yield). LCMS (ESI) m/z: [M + Na] calcd for C 5 iHgiNOo: 938.56; found 938.7; m/z: [M - H] calcd for C 5 iHsiNOn: 914.56; found 914.7.

Step 2: Synthesis of 32(R)-hydroxy 40(R)-O-(4-nitrophenyl)carbonate rapamycin

[00665] To a suspension of powdered 4A molecular sieves (6.0 g) in DCM (130 mL) was added 32(R)-hydroxy rapamycin (6.00 g, 6.55 mmol, 1.0 equiv). After stirring at room temperature for 45 min, pyridine (5.99 mL, 74.0 mmol, 11.3 equiv) was added. The suspension was cooled to -15 °C and then 4-nitrophenylchloroformate (1.78 g, 8.84 mmol,

1.4 equiv) was then added. The reaction mixture was stirred at -10 °C for 2 h and then filtered, and the filter pad washed with DCM (140 mL). The filtrate was washed with sat. NaHCCfi (130 mL) H 2 O (130 mL) and brine (130 mL), dried over NaiSCL, filtered and concentrated under reduced pressure. The crude material was purified by silica gel chromatography (2Q→5Q% EtOAc./hexanes) to give the product (4.44 g, 63% yield) as an off-white stiff foam. LCMS (ESI) m/z [M + Na] calcd for C58H84N2O17: 1103.57; found 1103.5.

Monomer 3. 32(R)-methoxy 40(R)-O-(4-nitrophenyl)carbonate rapamycin.

1 m

Step 1: Synthesis of 32(R)-methoxy-28,40-bistriethylsilyl rapamyein

[00666] To a stirred solution of 32(R )-hydroxy-28,40-bistriethylsilyl rapamyein (3 83 g, 3.34 mmol, 1.0 equiv) in chloroform (95.8 mL) was added Proton Sponge© (7.17 g, 33.5 mmol, 10.0 equiv) along with freshly dried 4 A molecular sieves (4 g). The solution was stirred for 1 h prior to the addition of trimethyloxonium telrafluoroborate (4.95 g, 33.5 mmol, 10.0 equiv, dried by heating under reduced pressure at 50 °C for 1 h before use) at room temperature. The reaction mixture was stirred for 18 h, and then the reaction mixture was diluted with DCM and filtered through Celite. The filtrate was washed sequentially with aqueous 1 M HCl (2x), sat. aqueous NaHCOa solution, then dried and concentrated under reduced pressure. Purification by silica gel chromatography (10→20% EtOAc/hexanes) afforded the desired product as a yellow oil that was contaminated with 3 wt.% Proton Sponge®. The residue was taken up in MTBE and washed with aqueous 1 M HCl, sat. aqueous NaHC()3 solution, dried, and then concentrated under reduced pressure to furnish a yellow foam (3.15 g, 81.2% yield). LCMS (ESI) rn/z: [M - TES + H 2 O] calcd for .J l : I i KOivSie: 1061.68; found 1061.9.

Step 2: Synthesis of 32(R)-methoxy rapamyein

[00667] To a stirred solution of 32(R )-methoxy-28,40-bistriethylsilyl rapamyein (1.11 g, 0.958 mmol, 1.0 equiv) in THE (12.6 mL) and pyridine (6.30 mL) in a plastic vial was added 70% HF-pyridine (2.22 mL, 76.6 mmol, 80.0 equiv) dropwise at 0 °C. The reaction mixture was stirred at 0 °C for 20 min before being warmed to room temperature for 3 h, when HPLC showed complete consumption of starting material. The reaction mixture was cooled to 0 °C and poured slowly into ice cold sat. aqueous NaHCO 3 solution (50 mL). The aqueous layer was extracted with EtOAc (3x) and the combined organics were washed with sat. aqueous NaHCOa solution, brine, dried, and concentrated under reduced pressure. The yellow residue was dissolved in MeOH (5 mL) and added dropwise to H 2 O (50 mL) to produce a white precipitate. After stirring for 15 min the slurry was filtered on a medium porosity funnel and the cake washed with H 2 O (2x) The solids were then dissolved in MeCN (50 mL) and lyophilized overnight to provide the product as a white solid (780 mg, 87% yield). LCMS (ESI) m/z [M + Na] calcd for C52H83NO13: 952.58; found 952.4.

Step 3: Synthesis of 32(R)-methoxy 40(R)-O-(4-nitrophenyl)carbonate rapamycin

[00668] To a solution of 32(f?)-methoxy rapamycin (4.50 g, 4.84 mmol, 1.0 equiv) in DCM (180 mL) was added pow'dered 4A molecular sieves (6.0 g). The mixture was stirred at room temperature for 1 h and then pyridine (3.91 mL, 48.4 mmol, 10 equiv) was added. The mixture was cooled to -10 °C and 4-nitrophenylchloroforaiate (0.990 g, 4.91 mmol, 1.0 equiv) was added in one portion. The reaction was allowed to slowly warm to room temperature and after 3 h the reaction mixture was cooled to 0 °C and 4- nitrophenylchloroformate (250 mg, 1.24 mmol, 0.3 equiv) was added. The mixture was warmed to roo temperature and after 1 h the reaction mixture was filtered through a pad of celite and the pad was washed with DCM (140 mL). The filtrate was washed with H 2 O (120 mL) and sat NaHCOi (2 x 120 mL). The organic phase was dried over N 3286)4, filtered and concentrated under reduced pressure. The crude material was purified by flash chromatography (20→50% EtOAc/hex) to yield a white stiff foam. The material was taken up in MeCN during which time a white solid formed. The soiid was filtered, washed with additional MeCN and allowed to air dry to provide the product (4 51 g, 85% yield). LCMS (ESI) m/z [M + Na] calcd for C 5 gHseNLOr / : 1117.58; found 1117.6. Monomers 4. 32(j?)-ethoxy 40(fl)-O-(4-nitropfaenyl)carbonate rapamycin.

Step 1: Synthesis of 32(j?)-ethoxy-28,40-bistriethylsilyl rapamycin

[00669] A solution of 32(R )-hydroxy-28,40-bistriethylsilyl rapamycin (773 mg, 0.675 mmol, 1.0 equiv) in chloroform (19 mL) was treated with N, N,N',N'-tetramethyl- 1,8- naphthalenediamine (1.85 g, 8.63 mmol, 12.8 equiv) along with freshly dried 4A molecular sieves. The mixture was stirred for 1 h at room temperature and treated with triethyloxonium tetrafluoroborate (1.51 g, 7.95 mmol, 11.8 equiv) in one portion at room temperature. The reaction mixture was stirred for 3 h, at which point the reaction mixture was diluted with DCM and filtered through Celite, washing the filter pad with additional DCM. The combined filtrates were washed twice with 1 M HCl, once with saturated NaHCO 3 solution, and dried over NaiiSOr. The solution was filtered and concentrated to a residue. The crude residue was treated with MTBE and filtered to remove polar insoluble material. The filtrate was concentrated and purified by silica gel chromatography (5→25% EtOAc/hex) to afford the product as a foam (516 mg, 65% yield). LCMS (ESI) m/z: [M + Na] calcd for C 6 sHmNOisSri 1194.77; found 1194.6. Step 2: Synthesis of 32(A)--ethoxy rapamycin

[00670] To a solution of 32(R )-ethoxyethoxy-28,40-bistriethylsilyl rapamycin (131 mg, 0.112 mmol 1.0 equiv) in THE (1.3 mL) at 0 °C was added pyridine (271 mΐ,, 3.35 mmol, 3.4 equiv) followed by 70% HF-pyridine (51 mΐ,, 1.8 mmol, 1.8 equiv). The reaction flask was capped and stored in the fridge for 3 days, at which point the reaction mixture was poured into cold saturated NaHCO 3 (20 mL). The aqueous layer extracted with EtOAc (3 x 20 mL) and the combined organic layers were washed with 1 M HCl (2 x 20 mL), saturated NaHCO 3 solution (20 mL), and brine. The solution was dried over NaaSCfl, filtered, and concentrated. The residue was taken up in MeOH (1.5 mL) and added dropwise to H 2 O (20 mL). The solids were filtered and washed with additional H 2 O to provide the product (53 mg, 51% yield) as a white powder. LCMS (ESI) m/z: [M + Na] calcd for C53H85NO33: 966.59; found 966.5.

Step 3: Synthesis of 32(R)-ethoxy 40(R )-O-(4-nitrophenyl)carbonate rapamycin

[00671] To a 0.03 M solution of 32(R)-ethoxy rapamycin (1.0 equiv) in DCM is added powdered 4 A molecular sieves. The mixture is stirred at room temperature for 1 h and then pyridine (10 equiv) is added. The mixture is cooled to -10 °C and 4-nitrophenylchloroformate (1.0 equiv) is added in one portion. The reaction is warmed to room temperature and stirred until consumption of 32(R)-ethoxy rapamycin, as determined by LCMS analysis. The mixture is filtered through a pad of celite and the pad washed with DCM. The filtrate is washed with H 2 O and sat NaHCO 3. The organic phase is then dried over Na 2 S0 4 , filtered and concentrated under reduced pressure. The crude material is purified by flash chromatography (20→50% EtOAc/hex) to provide the product.

Monomer 5. 40(J?)-O-(4-mtrophenyl)thiocarbonate rapamycin.

[00672] To a solution of rapamycin (4.00 g, 4.38 mmol, 1.0 equiv) in DCM (20 mL) at -78 °C w'as added pyridine (4.0 mL, 49 mmol, 11.2 equiv), followed by a solution of 0-(4- nitrophenyl)chlorothiocarbonate (1.19 g, 5.47 mmol, 1.3 equiv) in DCM (8.0 mL). The reaction mixture was warmed to -20 °C and stirred for 48 h. Hexane (40 mL) was then added and the resulting suspension was purified by silica gel chromatography (15/25/60 EtOAc/DCM/hexane then 20/25/55 EtOAc/DCM/hexane) to provide the product (3.09 g, 64.4% yield) as an off-white solid. LCMS (ESI) rn/z )M + Na] calcd for C 5 sHsaNaOieS: 1117.53; found 1117.5.

Monomer 6. 28-O-(4-nitrophenyl)carbonate rapamycin.

Step 1: Synthesis of 40-O-/¾r/-butyldimethylsilyl rapamycin

[00673] To a solution of rapamycin (1.00 g, 1.09 mmol, 1.0 equiv) in DMF (4 mL) at room temperature was added imidazole (0.22 g, 3.2 mmol, 2.9 equiv) followed by tert- butyldimethylsilyl chloride (0.176 g, 1.17 mmol, 1.07 equiv). The reaction mixture was stirred for 18 h. The reaction mixture was then diluted with DCM (100 mL) and washed with 20% aq. LiCl (3 x 20 mL). The organic layer was dried over NaaSC , filtered and concentrated under reduced pressure. The residue was purified by silica gei chromatography (20→4Q% EtOAc/hexanes) to give the product (950 mg, 75% yield) as a faint yellow glass. LCMS (ESI) rn/z: [M + H 2 O] calcd for Cv / ffeNOisSi: 1045.65; found 1045.9.

Step 2: Synthesis of 28-O-(4-nitrophenoxycarbonyl)-40-O-(tof-butyldimethylsilyl) rapamycin

[00674] To a solution of 40-O-f -butyldimethylsiIyl rapamycin (0.845 g, 0.822 mmol, 1.0 equiv) in DCM (10 mL) at room temperature was added pyridine (0.9 mL, 10 mmol, 12.1 equiv) followed by 4-nitrophenyl chloroformate (0.373 g, 1.85 mmol, 2.25 equiv). The reaction mixture was stirred for 2 h. The reaction mixture was then diluted with DCM (150 mL) and the solution sequentially washed with sat. NaHCO 3 (20 mL ), 10% citric acid (2 x 20 mL), and brine (20 mL). The organic layer was dried over NaaSCL, filtered, and concentrated under reduced pressure. The residue was purified by reverse phase chromatography (3Q-->10O% MeCN/HcO) to give the product (930 mg, 95% yield) as a pale yellow foam. LCMS (ESI) m/z: [M + H] calcd for C^HgeNrCft / Si: 1193.66; found 1193.7.

Step 3: Synthesis of 28-O-(4-nitrophenoxycarbonyl) rapamycin

[00675] To a solution of 28-O-(4-nitrophenoxycarbonyl)-40-O-(ieri- butyldimethylsilyl)rapamycin (0.930 g, 0.779 mmol, 1.0 equiv) in THF (10.7 mL) was added pyridine (3.78 mL, 46.8 mmol, 60.1 equiv) followed by the dropwise addition of 70% HF- pyridine (0.91 mL, 31.2 mmol, 40.0 equiv). The reaction mixture was stirred at room temperature for 48 h. The mixture was then poured slowly into ice cold sat. aqueous NaHCCL (20 mL). The aqueous layer was extracted with EtOAc (3 x 20 mL) and the combined organic layer was washed with sat. NaHCO 3 (10 mL) and brine (10 mL), dried over Na-rSCk, filtered, and concentrated under reduced pressure. The residue was purified by reverse phase chromatography (30→1()0% MeCN/H 2 O) to give the product (200 mg, 24% yield) as a faint yellow powder. LCMS (ESI) m/z [M + Na] calcd for C 5 sifeNcGi ? : 1101.55; found 1101.3.

Monomer 7, 32(R)-hydroxy 40(R)-O-(4-nitrophenyl)thiocarbonate rapamycin.

[00676] To a solution of 32(R )-hydroxy rapamycin (2.80 g, 3.06 mmol, 1.0 equiv) in DCM (28 mL) w'as added pyridine (27.6 mL, 34 mmol, 11 equiv) and dried 4 A molecular sieves (2.8 g). The suspension was stirred at room temperature for 1 h, at which point the mixture W ' as cooled to -25 °C and a solution of 0-(4-nitrophenyl)chlorothioformate (0.798 g, 3.67 mmol, 1.2 equiv) in DCM (6 mL) was added. The reaction was warmed to room temperature and after 21 h was filtered through Celite. The filtrate was partitioned between DCM and H 2 O and the aqueous layer was extracted with DCM. The combined organic layers were dried and concentrated under reduced pressure. Purification by silica gel chromatography (EtOAc/hexanes) afforded the desired product as a white solid (2.15 g, 64% yield). LCMS (ESI) m/z [M + Na] calcd for CfolfoN dfoS: 1119.54; found 1120.0.

Monomer 8. 32(R)-methoxy 40(R )-O-(4-mtrophenyl)thiocarbonate rapamycin.

[00677] To a solution of 32(R)-methoxy rapamycin (6.69 g, 7.19 mmol, 1.0 equiv) in DCM (67 mL) was added pyridine (6.6 mL, 81 mmol, 11 equiv) and dried 4 A molecular sieves (6.7 g). The suspension was stirred at room temperature for 1 h, at which point the mixture was cooled to -25 °C and a solution of 0-(4-nilrophenyI)chlorothioformate (1 88 g, 8.63 mmol, 1.20 equiv) in DCM (13 L) was added. The reaction was warmed to room temperature and after 21 h was filtered through Celite. The filtrate was partitioned between DCM and H 2 O and the aqueous layer was extracted with DCM. The combined organic layers were dried and concentrated under reduced pressure. Purification by silica gel chromatography (EtOAc/hexanes) afforded the desired product as a white solid (5.1 g, 64% yield). LCMS (ESI) rn/z: [M + Na] calcd for C 5 gHseNaOieS: 1133.56; found 1134.1.

Monomer 9. 32-deoxy 40(if)- -(4-nitrophenyI)carbonate rapamycin.

[0067S] To a solution of 32-deoxy rapamycin (0623 g, 0.692 mmol, 1.0 equiv) in DCM (24.7 mL) was added 4 A molecular sieves (600 mg). The suspension was stirred for 1 h and then pyridine (557 μL, 6.92 mmol, 10 equiv) was added. The reaction mixture was cooled to 0 °C and then 0-(4-nitrophenyl)chloroformate (175 mg, 1.03 mmol, 1.7 equiv) was added. The reaction warmed to room temperature and stirred for 2 h, at which point the reaction mixture was concentrated under reduced pressure. Purification by silica gel chromatography (0→10% MeOH/DCM) afforded the desired product as a white solid (0.61 g, 82% yield). LCMS (ESI) m/z: [M + Na] ealcd for C 5 sHg^Oie: 1087.57; found 1087.6.

Monomer 10. 32-deoxy 40(R )-O-(4-nitrophenyl)thiocarbonate rapamycin.

[00679] To a 0.2 M solution of 32-deoxy rapamycin (1.0 equiv) in DCM at -78 °C is added pyridine (11.2 equiv), followed by a 0.7 M solution of 0-( 4- nitrophenyl)chlorotihiocarbonate (1.3 equiv) in DCM. The reaction mixture is warmed to -20 °C and stirred until consumption of the stalling material, as determined by LCMS analysis. Hexane is then added and the resulting suspension is purified by silica gel chromatography to provide the product.

Monomer 11, 28(R)-raethoxy 32i/£)-hydroxy 40(J?)-(4-nItropheoyl)carbonate rapamyc .

Step 1: Synthesis of 28(Z?)-methoxy 40(R)-O-½ri-butyldimethylsilyl rapamycin

[00680] To a solution o 40(R )-O-/er -butyldimethylsilyl rapamycin (4.00 g, 4.89 mmol, 1.0 equiv) in chloroform (67 mL) was added proton sponge (11.2 mL, 52.3 mmol, 13 equiv) and dried 4 A molecular sieves (5.8 g). The suspension was stirred at room temperature for 1 h, at which point trimethyloxonium tetrafluoro borate (7.21 g, 48.8 mmol, 12.5 equiv) was added. After 4 h the suspension was filtered through Celiie. The filtrate was washed sequentially with aqueous 2 N HCl, H 2 O, sat. aqueous NaHCO 3 , then dried and concentrated under reduced pressure. Purification by silica gel chromatography (EtOAe/hexane) afforded the desired product as a white solid (2.1 g, 52% yield). LCMS (ESI) m/z: [M + Na] calcd for C58H95NO13S1: 1064.65; found 1065.26.

Step 2: Synthesis of 28(R )-methoxy 32(A)-hydroxy 40(R )-O-ieri-butyldimethylsilyl rapamycin [00681] To a solution of 28(R)-methoxy 40(A’) O-ie/f-butyldimethyIsiiyl rapamycin (2.13 g, 2.04 mmol, 1.0 equiv) in THF (31 mL) at -20 °C was added a solution of lithium ίή-tert- butoxyaluminumhydride in THF (1 M, 4.09 L, 4.09 mmol, 2.0 equiv), dropwise. The reaction mixture was warmed to room temperature and after 3 h was added to a solution of H 2 O (4 mL), EtOAc (31 mL), and 2M aqueous citric acid (4 mL) at 0 °C. After 5 min the mixture was partitioned, and the aqueous layer was extracted with EtOAc. The combined organic layers were poured into a sat. aqueous NaHCO 3 solution (60 mL) at 0 °C. The layers were partitioned, and the organic layer was dried and concentrated under reduced pressure to provide a crude white solid (2.32 g). The crude solid was dissolved in DCM (12 mL) and then pyridine (241 μL, 2.98 mmol, 1.5 equiv), dried 4 A molecular sieves (2.1 g), and cupric acetate (0.27 g, 1.49 mmol, 0.7 equiv) were added. The suspension was stirred at room temperature for 1 h. The suspension was sparged with O 2 and then kept under an O 2 atmosphere for 30 min. After 2 h the mixture was filtered through Celite and the filtrate was concentrated under reduced pressure. Purification by silica gel chromatography

(EtO Ac/hexane) afforded the desired product as a white solid (307 mg, 14% yield). LCMS (ESI) m/:. [M + Na] calcd for CVsl .NOuSi: 1066.66; found 1067.0.

Step 3: Synthesis of 28(R)-methoxy 32(R)-hydroxy rapamycin

[00682] To a solution of 28(f?)-methoxy 32(R)-hydroxy 40(/6)-O-ierAhutyldimethylsiiyl rapamycin (0.307 g, 0.294 mmol, 1.0 equiv) in THF (4 mL) in a polypropylene vial at 0 °C was added pyridine (1.42 mL, 17.6 mmol, 60.0 equiv) followed by 70% HF-pyridine (0.34 mL, 11.7 mmol, 40 equiv). The solution was warmed to room temperature and stirred for 21 h, at which point the solution was poured into sat. aqueous NaHCCL at 0 °C. The aqueous layer was extracted with EtOAc (2x) and the combined organic layers were washed with sat. aqueous NaHCO 3 and brine, then dried and concentrated under reduced pressure. Purification by silica gel chromatography (EtO Ac/hexane) afforded the desired product as a white solid (146 mg, 53% yield). LCMS (ESI) m/z: [M + Na] calcd for C52H83NO13: 952.58; found 952.8.

Step 4: Synthesis of 28(R )-methoxy 32(R )-hydroxy 40(R )-(4-nitrophenyl)carbonate rapamycin

[00683] To a solution of 28(f?)-methoxy 32(R)-hydroxy rapamycin (0.66 g, 0.71 mmol, 1.0 equiv) in DCM (3 mL) was added pyridine (0.64 mL, 7.9 mmol, 11 equiv) and dried 4 A molecular sieves (0.66 g). The suspension was stirred at room temperature for 1 h, at which point the mixture was cooled to -35 °C and 0-(4-nitrophenyl)chlorofom ate (0.17 g, 0.85 mmol, 1.2 equiv) was added. After 3 h, DCM (5 mL) was added and the suspension was filtered through Celite. The filtrate was washed with H 2 O, dried, and concentrated under reduced pressure. Purification by silica gel chromatography (EtOAc/hexanes) afforded the desired product as a white solid (0.44 g, 57% yield). LCMS (ESI) m/z: [M + Na] calcd for

C 59 H 86 N 2 O 17 : 1117.58; found 1118.0.

Monomer 12. 28(R )-methoxy 32(R)-methoxy 40(R)-(4-nitrophenyl)carbonate rapamycin.

Step 1: Synthesis of 28(R)-methoxy 32(R)-methoxy 40(R )-O-tert-butyldimethylsilyl rapamycin

[00684] To a solution of 28(R)-methoxy 32(R )-hydroxy 40(R)-O-tert-butyldimethylsilyl rapamyein (1.15 g, 1.10 mmol, 1.0 equiv) in chloroform (19 mL) was added proton sponge (3.22 mL, 15.0 mmol, 14 equiv) and dried 4A molecular sieves (2,3 g). The suspension was stirred at room temperature for 1 h, at which point trimethyloxonium tetrafluoroborate (2.07 g, 14.0 mmol, 12.7 equiv) was added. After 4 h the suspension was filtered through Celite and the filtrate was washed with IN HCl, H 2 O, and sat. aqueous NaHCO 3 , dried and concentrated under reduced pressure. Purification by silica gel chromatography (EtOAc/hexane) afforded the desired product as a white solid. LCMS (ESI) m/z: [M + Na] calcd for C 59 H 99 NO 13 Si: 1080.68; found 1081.2.

Step 2: Synthesis of 28(R)-methoxy 32(R )-methoxy rapamyein [00685] To a solution of 28(R)-methoxy 32(R)-methoxy 40(R )-O-tert-butyldimethylsilyl rapamycin in THF (4 mL) in a polypropylene vial at 0 °C was added pyridine (1.13 mL, 14.2 mmol, 12.9 equiv) followed by 70% HF-pyridine (0.27 mL, 9.42 mmol, 8.6 equiv). The solution was warmed to room temperature and stirred for 41 h, at which point the solution was poured into sat. aqueous NaHCO 3 at 0 °C. The aqueous layer was extracted with EtOAc (2x) and the combined organic layers were washed with sat. aqueous NaHCO 3 and brine, then dried and concentrated under reduced pressure. Purification by silica gel chromatography (EtG Ac/hexane) afforded the desired product as a white solid (516 mg, 49% yield 2-steps). LCMS (ESI) rn/z: [M + Na] calcd for C 53 H 85 N O 13 : 966.59; found 967.0.

Step 3: Synthesis of 28(R)-methoxy 32(R)-methoxy 40(R )-(4-nitrophenyl)carbonate rapamycin

[00686] To a solution of 28(R)-methoxy 32(R )-methoxy rapamycin (0.30 g, 0.32 mmol,

1.0 equiv) in DCM (1.4 mL) was added pyridine (0.29 mL, 3.5 mmol, 11 equiv) and dried 4 A molecular sieves (0.30 g). The suspension was stirred at room temperature for 1 h, at which point it was cooled to -35 °C and 0-(4-nitrophenyl)chloroformate (0.08 g, 0.38 mmol, 1.2 equiv) was added. After 3 h, DCM (2 mL) was added and the suspension was filtered through Celite. The filtrate was washed with H 2 O, dried and concentrated under reduced pressure. Purification by silica gel chromatography (EtOAc/hexanes) afforded the desired product as an off-white solid (0.20 g, 57% yield). LCMS (ESI) m/z: [M + Na] calcd for C 60 H 88 N 2 O 17 : 1131.60; found 1132.1.

Monomer 13, 32(R)-(4-nitrophenyl)carbonate rapamycin.

Step 1: Synthesis of 28,40-O-bis(triethylsilyl) 32(R)-(4-nitrophenyl)carbonate rapamycin

[00687] To a solution of 28,40-O-bis(trietbyisilyl) 32(R)-hydroxy rapamycin (0.602 g, 0.526 mmol, 1.0 equiv) in DCM (16 mL) at -20 °C was added pyridine (0.82 mL, 10 mmol, 19 equiv) followed by O-(4-nitrophenyl)chloroformate (0.36 g, 1.8 mmol, 3.4 equiv). The reaction mixture was warmed to room temperature and stirred for 1 h, at which point the solution was diluted with DCM (50 mL) and poured into H 2 O (30 mL). The aqueous layer was extracted with DCM (50 mL) and the combined organic layers were washed with brine (20 mL), dried and concentrated under reduced pressure to afford a faint yellow foam that was used directly in the next step.

Step 2: Synthesis of 32(R )-(4-nitrophenyl)carbonate rapamycin

[00688] To a solution of 28,40-O-bis(triethylsilyl) 32(R )-(4-nitrophenyl)carbonate rapamycin in THF (10 mL) in a polypropylene vial at 0 °C was added pyridine (1.70 mL,

21.0 mmol, 40.0 equiv) followed by 70% HF-pyridine (0.46 mL, 15.8 mmol, 30.0 equiv).

The solution was warmed to room temperature and stirred overnight, at which point the solution was poured into sat. aqueous NaHCO 3 at 0 °C. The aqueous layer was extracted with EtOAc (3x) and the combined organic layers were washed with sat. aqueous NaHCO 3 and brine, then dried and concentrated under reduced pressure. Purification by reverse phase chromatography (20→100% MeCN/H 2 O) afforded the desired product as an off-white powder (420 mg, 74% yield 2-steps). LCM8 (ESI) m/z: [M + Na] calcd for C 58 H 84 N 2 O 17 : 1103.57; found 1104.0.

Monomer 14. 32(S) -azido 40-(4-nitrophenyl)carbonate rapamycin.

Step 1: Synthesis of 28,40-O-bis(triethylsilyl) 32(R )-O-methanesulfonyl rapamycin

[00689] To a solution of 28,40-O-bis(triethylsilyl) 32(R)-hydroxy rapamycin (2.50 g, 2.18 mmol, 1.0 equiv) in DCM (25 mL) at 0 °C was added Et 3 N (0.912 mL, 6.54 mmol, 3.0 equiv) followed by methanesulfonyl chloride (0.338 mL, 4.36 mmol, 2.0 equiv). The solution was stirred at 0 °C for 3 h, at which point the EtOAc was added and the solution was washed with sat. aqueous NaHCO 3 . The combined organic layers were washed with brine, dried and concentrated under reduced pressure to give a yellow oil which was used directly in the next step.

Step 2: Synthesis of 28-O-triethylsilyl 32(S)-azido rapamycin

[00690] To a solution of 28,40-O-bis(triethylsilyl) 32(R)-O-methanesulfonyl rapamycin in THF (40 mL) was added DIPEA (0.761 mL, 4.37 mmol, 2.0 equiv) and tetrabutylammonium azide (3.72 g, 13.1 mmol, 6.0 equiv). The reaction solution heated to reflux for 5.5 h and then cooled to room temperature. The solution was diluted with EtOAc and washed with sat. aqueous NaHCO 3 . The combined organic layers were washed with brine, dried and concentrated under reduced pressure. Purification by reverse phase chromatography (30→100% MeCN/H 2 O) afforded the desired product as a clear glass (746 mg, 33% yield 2- steps). LCMS (ESI) m/z: [M + Na] calcd for C 57 H 94 N 4 O 12 Si: 1077.65; found 1077.8.

Step 3: Synthesis of 28-O-triethylsilyl 32(S)-azido 40(R)-(4-nitrophenyl)carbonate rapamycin

[00691] To a solution of 28-O-triethylsilyl 32(S)-azido rapamycin (0.505 g, 0.478 mmol,

1.0 equiv) in DCM (15 mL) was added pyridine (0.75 mL, 9.3 mmol, 19 equiv) and 4 A molecular sieves. The suspension was cooled to -20 °C and 0-(4-nitrophenyl)chloroformate (0.32 g, 1.6 mmol, 3.4 equiv) was added. The suspension was stirred at -20 °C for 2 h, at which point the it was diluted with DCM (50 mL), filtered and poured into H 2 O (20 mL). The aqueous layer was extracted with DCM (50 mL) and the combined organic layers were washed with brine (20 mL), dried and concentrated under reduced pressure to give a pale- yellow foam which was used directly in the next step.

Step 4: Synthesis of 32(S)-azido 40-O-(4-nitrophenyl)carbonate rapamycin

[00692] To a solution of 28-O-triethylsilyl 32(S)-azido 40(R)-(4-nitrophenyl)carbonate rapamycin in THF (10 mL) in a polypropylene vial at 0 °C was added pyridine (1.55 mL,

19.1 mmol, 40.0 equiv) followed by 70% HF-pyridine (0.42 mL, 14.4 mmol; 30.0 equiv).

The solution was warmed to room temperature and stirred overnight, at which point the solution was poured into sat. aqueous NaHCO 3 at 0 °C. The aqueous layer was extracted with EtOAc (3x) and the combined organic layers were washed with sat. aqueous NaHCO 3 and brine, then dried and concentrated under reduced pressure. Purification by reverse phase chromatography (30→100% MeCN/H 2 O) afforded the desired product as an off-white powder (410 mg, 77% yield 2-steps). LCMS (ESI) m/z : [M + Na] calcd for C 58 H 83 N 5 O 16 : 1128.57; found 1129.0. Monomer 15. 28-methoxy-40-O-(4-nitrophenoxy)carbonyl rapamycin.

Step 1: Synthesis of 28-methoxy rapamycin

[00693] To a solution of 28-methoxy-40-O-(tert-butyldimethyI)siIyl rapamycin (0.500 g, 0.480 mmol, 1.0 equiv) in MeOH (1.6 mL ) at -20 °C was added H 2 SO 4 (1.28 μL, 0.024 mmol, 0.05 equiv). The reaction mixture was stirred at -20 °C for 48 h. The reaction mixture was then poured into sat. aqueous NaHCO 3 (4 mL)/H 2 O (4 mL ). The aqueous layer was extracted with MTBE (2 x 6 mL), and the combined organic phases were dried, filtered, and concentrated under reduced pressure. Purification by reverse phase chromatography (30→100% MeCN/H 2 O) afforded the desired product as a yellow powder (270 mg, 61% yield). LCMS (ESI) m/z: [M + Na] calcd for C 52 H 81 NO 13 : 950.5; found 950.7.

Step 2: Synthesis of 28-methoxy-40-O-(4-nitrophenoxy)carbonyl rapamycin

[00694] To a solution of 28-methoxy rapamycin (0.210 g, 0.226 mmol, 1.0 equiv) in DCM (7.1 mL) at -20 °C was added pyridine (0.35 mL, 4.4 mmol, 19 equiv) and then p-nitrophenyl chloroformate (0.15 g, 0.76 mmol, 3.4 equiv). The reaction mixture was stirred at -20 °C for 30 min and then warmed to room temperature. After stirring overnight, p-nitrophenyl chloroformate (0.15 g, 0.76 mmol, 3.4 equiv) was added and the reaction was stirred for an additional 2 h. The reaction mixture was diluted with DCM (20 mL) and poured into H 2 O (10 mL). The aqueous layer was extracted with DCM (20 mL), and the combined organic layers were washed with brine (9 mL), dried, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography (50→100% MeCN/H 2 O) afforded the desired product as a pale yellow powder (200 mg, 81% yield). LCMS (ESI) m/z: [M + Na] calcd for

C 59 H 84 N 2 O 17 : 1115.6; found 1115.8.

Monomer 16. 32(R),40-O,O-bis|(4-nitrophenoxy)carbonyl] rapamycin.

[00695] To a solution of 32(R)-O-[(4-nitrophenoxy)carbonyl] rapamycin (675 mg, 0.624 mmol, 1.0 equiv) in DCM (13 mL) was added powdered 4A molecular sieves (675 mg). The suspension was stirred for 1 h, at which point pyridine (0.56 mL, 6.90 mmol, 11.1 equiv) was added. The mixture was cooled to -15 °C and then p-nitrophenyl cbloroformate (132 mg, 0.655 mmol, 1.05 equiv) was added in one portion. The mixture was wanned to 0 °C, stirred for 4 h, and then warmed to room temperature. The reaction mixture was filtered and washed with DCM (25 mL). The filtrate was washed with sat. aqueous NaHCO 3 (15 mL), H 2 O (15 mL), and brine (10 mL), dried, filtered, and concentrated under reduced pressure. Purification by silica gel chromatography (25→45% EtOAc/hexanes) afforded the desired product as a faint yellow solid (566 mg, 73% yield). LC-MS (ESI) rn/z: [M + Na] calcd for C 65 H 87 N 3 O 21 : 1268.57; found 1269.3.

Monomer 17. 28(R),32(R),40(R)-O,O,O-tris[(4-nitrophenoxy)carbonyl] rapamycin.

[00696] To a solution of 32(R)-hydroxy rapamycin (1.00 g, 1.09 mmol, 1.0 equiv) in DCM (22 mL) was added powdered 4Å molecular sieves (1.0 g). The suspension was stirred for 45 min, at which point pyridine (0.97 mL, 12.0 mmol, 11.0 equiv) was added. The mixture was cooled to -15 °C and then p-nitrophenyl chloroforrnate (550 mg, 2.73 mmol, 2.5 equiv) was added in one portion. The mixture was warmed to room temperature over 4 h and stirred overnight. The mixture was cooled to 0 °C and additional p-nitrophenyl chloroforrnate (220 mg, 1.09 mmol, 1.0 equiv) was added in one portion. The reaction mixture was stirred for 1 h, warmed to room temperature, and then stirred for 2 h. The mixture was once again cooled to 0 °C and additional p-nitrophenyl chloroforrnate (660 mg, 3.27 mmol, 3.0 equiv) was added. The reaction mixture was stirred for 15 min and then at room temperature for 1 h. The reaction mixture was filtered and washed with DCM (25 mL). The filtrate was washed with sat. aqueous NaHCO 3 (20 mL), H 2 O (20 mL), and brine (15 mL), dried, filtered, and concentrated under reduced pressure. Purification by silica gel chromatography (5→15% EtOAc/DCM) afforded the desired product as a faint yellow solid (550 mg, 36% yield). LC- MS (ESI) m/z: [M + Na] calcd for C 72 H 90 N 4 O 25 : 1433.58; found 1434.3.

General Procedures and Specific Examples,

General Procedure 1 : Coupling of a carboxylic add and an amine followed by N-Boc deprotectlon.

Step 1:

[00697] To a 0.1 M solution of carboxylic acid (1.0 equiv) in DMA was added an amine (1.2 equiv), DIPEA (4.0 equiv) and PyBOP (1.3 equiv). The reaction was allowed to stir until consumption of the carboxylic acid, as indicated by LCMS. The reaction mixture was then purified by silica gel chromatography to afford the product.

Step 2:

[00698] To a 0.07 M solution of N-Boc protected amine (1.0 equiv) in dioxane was added HCl (4 M in dioxane) (50 equiv). The reaction was allowed to stir until consumption of N- Boc protected amine, as indicated by LCMS. Then the reaction was concentrated to an oil, which was then dissolved in H 2 O and lyophilized to afford the product. Intermediate A1-7. 1-amino-27-(6-{[4-amino-3-(2-amino-1,3-benzoxazol-5-yl)-1H- pyrazoIo[3,4-d]pyrimidin-1-yl]methyl}-1,2,3,4-tetrahydroisoq uinoHn-2-yl)-

3,6,9,12,15,18,21,24-octaoxaheptacosan-27-one

Step 1 : Synthesis of tert-butyl N-[27-(6-{[4-amino-3-(2-amino-1,3-benzoxazol-5-yl)-1H- pyrazolo[3,4-d]pyrimidin-1-yl]methyl)-1,2,3,4-tetrahydroisoq uinolin-2-yl)-27-oxo-

3,6,9,12,15,18,21,24-octaoxaheptacosan-1-yl]carbamatecarb amate

[00699] To a solution of 1-{[(tert-butoxy)carbonyl]amino}-3,6,9,12,15,18,21,24- octaoxaheptacosan-27-oic acid (102 mg, 189 μmol, 1.0 equiv) and 6-{[4-amino-3-(2-ammo- l,3-benzoxazol-5-yl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl]methyl }-1,2,3,4- tetrahydroisoquinolin-2-ium (120 mg, 227 μmol, 1.2 equiv) in DMA (1.88 mL ) was added DIPEA (131 μL, 756 μmol, 4.0 equiv) followed by PyBOP (127 mg, 245 μmol, 1.3 equiv). The reaction was stirred at room temperature. After 2 h, the reaction mixture was concentrated under reduced pressure and the crude residue was purified by silica gel chromatography (0→20% MeOH/DCM) to give the product (161.5 mg, 91% yield) as a pale yellow oil. LCMS (ESI) m/z: [M + H] calcd for C 46 H 65 N 9 O 12 : 936.49; found 936.3.

Step 2: Synthesis of 1-amino-27-(6-{[4-amino-3-(2-amino-1,3-benzoxazol-5-yl)-1H- pyrazolo[3 ,4-d]pyrimidin-1-yl]methyl}-1,2,3,4-tetrahydroisoqumolin-2-y l)-

3.6.9.12.15.18.21.24-octaoxaheptacosan-27-one

[00700] To a solution of tert-butyl N-[27-(6-{ [4-amino-3-(2-amino-1,3-benzoxazol-5-yl)- 1H-pyrazolo[3 ,4-d]pyrimidin-1-yl]methyl}-1,2,3,4-tetrahydroisoquinolin-2- yl)-27-oxo-

3.6.9.12.15.18.21.24-octaoxaheptacosan-1-yl]carbamate (0.9 g, 0.9614 mmol, 1.0 equiv) in dioxane (3.20 mL) was added HCl (4 M in dioxane, 2.40 mL, 9.61 mmol, 10.0 equiv). The reaction stirred for 2 h and then was concentrated under reduced pressure to an oil. The oil was azeotroped with DCM (3 x 15 mL) to provide the product (881 mg, 105% yield, HCl) as a tan solid, which was used directly in the next step. LCMS (ESI) m/z: [M + H] calcd for C 41 H 57 N 9 O 10 : 836.43; found 836.3. [00701] Following General Procedure 1 but using the appropriate amine-containing active site inhibitor in Table 2 and PEG carboxylic acid, the Intermediates A1 in Table 5 were prepared:

Table 5. Additional amines prepared

[00702] Following General Procedure 1, but using the appropriate Intermediate A1 in Table 5 and PEG carboxylic acid, the Intermediates A2 in Table 6 were prepared:

Table 6. Additional amines prepared

General Procedure 2: Coupling of a 4-nitrophenyl carbonate containing rapamycin monomer and an active site inhibitor containing intermediate having a primary or secondary amine.

[00703] To a 0.02 M solution of 4-nitrophenyl carbonate containing rapamycin monomer (1.0 equiv) and an active site inhibitor containing intermediate (2.0 equiv) in DMA was added DIPEA (4.0 equiv). The resulting solution was stirred at room temperature under nitrogen. Upon completion as determined by LCMS analysis, the crude reaction mixture was purified by preparative HPLC to provide the product.

Example 2: Synthesis of Series 1 bivalent rapamyem compound.

[00704] To a solution of 40(R)-O-(4-nitrophenyl carbonate) rapamycin (25 mg, 23.16 μmol, 1.0 equiv) and Intermediate A1-7 (42.0 mg, 46.32 μmol 2.0 equiv) in DMA (1.15 mL ) was added DIPEA (16.0 μL, 92.64 μmol, 4.0 equiv). The reaction was stirred for 18 h, at which point the reaction mixture purified by reverse phase chromatography (10→40→95% MeCN + 0.1% formic acid/H 2 O + 0.1 % formic acid) to give the product (9.92 mg, 24% yield) as a white solid. LCMS (ESI) m/z: [M + H] ealed for C 93 H 134 N 10 O 24 : 1775.97; found 1775.7.

[00705] Following General Procedure 2, but using the appropriate 4-nitrophenyl carbonate containing rapamycin monomer in Table 1 and Intermediates A1 and A2 from Tables 5 and 6, the Series 1 bivalent analogs in Table 7 were synthesized: Table 7. Series 1 Bivalent Compounds:

General Procedure 3: Coupling of a halide containing PEG ester and an amine containing pre-linker followed by ester deprotection. Step 1:

[00706] To a 0.1 M solution of amine containing pre-linker (1.0 equiv) in MeCN was added K 2 CO 3 (2.0 equiv) followed by halide containing PEG ester (1.0 equiv). The reaction was stirred at 80 °C until consumption of amine containing pre-linker, as indicated by LCMS analysis. The reaction was then purified by silica gel chromatography to afford the product.

Step 2:

[00707] To a 0.1 M solution of PEG tert-butyl ester (1.0 equiv) in EtOAc was added a solution of HCI in EtOAc, The resulting suspension was stirred at room temperature until consumption of the PEG ester, as indicated by LCMS analysis. The reaction was then concentrated under reduced pressure to afford the product,

Intermediate B1-1. 1-(4-(5-((1,3-dioxoisoindolin-2-yl)methyl)pyrimidin-2-yl)pip erazin-1- yI)-3,6,9,12-tetraoxapentadecan-15-oic add

Step 1: Synthesis of tert-butyl 1-(4-(5-((1,3-dioxoisoindolin-2-yl)methyl)pyrimidin-2- yl)piperazin-1-yl)-3 ,6,9, 12-tetraoxapentadecan-15-oate

[00708] To a mixture of 2-((2-(piperazin-1-yl)pyrimidin-5-yl)methyl)isoindoline-1 ,3-dione (7,97 g, 24,66 mmol, 1.0 equiv) in MeCN (200mL) was added K 2 CO 3 (6.82 g, 49,31 mmol, 2.0 equiv) followed by tert-butyl 1-bromo-3 ,6,9,12-tetraoxapentadecan-15 -oate (9.5 g, 24.66 mmol, 1.0 equiv). The reaction mixture was heated to 85 °C and stirred for 15 h, The mixture was then cooled to room temperature and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel chromatography (0→20% EtOAc/MeOH) to give the product (11.5 g, 74.3% yield) a light yellow liquid.

Step 2: Synthesis of 1-(4-(5-((1,3-dioxoisoindolin-2-yl)methyl)pyrimidin-2-yl)pip erazin-1- yl)-3,6,9,12-tetraoxapentadecan-15-oic acid

[00709] To a solution of tert-butyl 1-(4-(5-((1,3-dioxoisoindolin-2-yl)methyl)pyrimidin-2- yl)piperazin-1-yl)-3,6,9,12-tetraoxapentadecan-15-oate (3.5 g, 5.58 mmol, 1.0 equiv) in EtOAc (50 mL) was added a solution of HCI in EtOAc (500 mL ). The mixture was stirred at room temperature for 3 h. The mixture was then concentrated under reduced pressure to give the product (5.3 g, 78.2% yield, HCl) as a white solid. LCMS (ESI) m/z: [M + H] calcd for C 28 H 37 N 5 O 8 : 572.27; found 572.4.

[00710] Following General Procedure 3, but using the appropriate halide containing PEG and amine containing pre-linkers in Table 4, the Intermediates B1 in Table 8 were prepared: Table 8. Additional protected amines prepared

General Procedure 4: Coupling of a PEG carboxylic acid and an amine containing active site inhibitor followed by amine deprotection.

Step 1:

[00711] To a 0.15 M solution of PEG carboxylic acid (1.0 equiv) in DMF was added HATU (1.3 equiv) and DIPEA (5.0 equiv). After stirring for 30 min, the amine containing active site inhibitor (1.2 equiv) was added. The reaction was stirred at room temperature until consumption of PEG carboxylic acid, as indicated by LCMS. The reaction was then purified by reverse phase chromatography to afford the product.

Step 2:

[00712] To a 0.1 M solution of phthalimide protected amine (1.0 equiv) in MeOH at 0 °C was added NH 2 NH 2 •H 2 O (4.0 equiv). The resulting mixture was stirred at 60 °C until consumption of the phthalimide protected amine, as indicated by LCMS analysis. The reaction was then purified by reverse phase chromatography to afford the product. Intermediate B2-1. N-(4-(4-amino-3-(2-aminobenzo[d]oxazol-5-yl)-1H-pyrazolo[3,4 - d]pyrimidm-1-yl)butyl)-1-(4-(5-(aminomethyl)pyrimidm-2-yl)pi perazin-1-yl)-3,6,9,I2- tetraoxapentadecan-15-amide

Step 1 : Synthesis of N-(4-(4-amino-3-(2-ammobenzo[d]oxazol-5-yl)-1H-pyrazolo[3,4- d]pyrimidin-1-yl)butyI)-1-(4-(5-((1,3-dioxoisoindolin-2-yl)x nethyl)pyrimidin-2-yl)piperazin- 1-yl)-3,6,9,12-tetraoxapentadecan-15-amide

[00713] To a mixture of 1-(4-(5-((1,3-dioxoisoindolin-2-yl)methyl)pyrimidin-2- yl)piperazin-1-yl)-3,6,9,12-tetraoxapentadecan-15-oic acid (3 g, 4.93 mmol, 1.0 equiv, HCl) in DMF (30 mL) was added HATU (12.11 μL, 6.41 mmol, 1.3 equiv) and DIPEA (4.30 mL, 24.67 mmol, 5.0 equiv). After 30 min, 5-(4-amino-1-(4-aminobutyl)-1H-pyrazolo[3,4- d]pyrimidin-3-yl)henzo[d]oxazol-2-amine (4.03 g, 5.92 mmol, 1.2 equiv, 3TFA) was added. The mixture was stirred at room temperature for 3 h. The reaction mixture was then purified by prep-HPLC (MeCN/H 2 O) to give the product (5.4 g, 81.2% yield, 4TFA) as a light red solid. LCMS (ESI) m/z: [M + 2H]/2 calcd for C 44 H 53 N 13 O 8 : 446.71; found 447.0.

Step 2: Synthesis of N-(4-(4-amino-3-(2-ammobenzo[d]oxazol-5-yl)-1H-pyrazolo[3,4- d]pyrimidin-1-yl)butyl)-1-(4-(5-(aminomethyl)pyrimidin-2-yl) piperazin-1-yl)-3,6,9,12- tetraoxa pen tadec an-15-amide

[00714] To a mixture of N-(4-(4-ammo-3-(2-aminobenzo[d]oxazol-5-yl)-1H-pyrazolo[3,4- d]pyrimidin-1-yl)butyl)-1-(4-(5-((1,3-dioxoisoindolin-2-yl)m ethyl)pyrirmdin-2-yl)piperazin- 1-yl)-3, 6, 9, 12-tetraoxapentadecan-15-amide (4 g, 2.97 mmol, 1.0 equiv, 4TFA) in MeOH (25 mL) at 0 °C was added NH 2 NH 2 •H 2 O (588.63 μL, 11.87 mmol, 4.0 equiv). The mixture was stirred at 60 °C for 2 h. The mixture was then cooled to room temperature and filtered, and the filter cake was washed with MeOH (5 mL). The filtrate was concentrated under reduced pressure and the residue was purified by prep-HPLC (MeCN/H 2 O) to give the product (700 mg, 24.5% yield, TFA) as a white solid. LCMS (ESI) m/z: [M + 2H]/2 calcd for C 36 H 51 N 13 O 6 :381.71; found 381.8.

[00715] Following General Procedure 4, but using the appropriate Intermediate B1 in Table 8 and amine containing active site inhibitors in Table 2, the Intermediates B2 in Table 9 were prepared:

Table 9. Additional amines prepared

General Procedure 5: Coupling of a halide containing PEG carboxylic add and an amine containing active site inhibitor.

[00716] To a 0.1 M solution of amine containing active site inhibitor (1.0 equiv) and PEG containing carboxylic acid (1.2 equiv) in DMA was added DIPEA (4.0 equiv) followed by PyBOP (1.3 equiv). The reaction was stirred until consumption of amine containing active site inhibitor, as indicated by LCMS. The reaction was then purified by reverse phase HPLC to afford the product.

Intermediate B3-1. 18-{6-[(4-amino-3-{1H-pyrrolo[2,3-b]pyridin-5-yI}-1H-pyrazol o[3,4- d]pyrimidin-1-yl)methyI]-1,2,3,4-tetrahydroisoquinolin-2-yI} -1-bromo-3,6,9,12,15- pentaoxaoctadecan-18-one

[00717] To a solution of 1-bromo-3 ,6,9, 12,15-pentaoxaoctadecan-18-oic acid (105 mg, 282 μmol, 1.2 equiv) and 3-{ 1H-pyrrolo[2,3-b]pyridin-5-yl}-1-[(1,2,3,4- tetrahydroisoquinolin-6-yl)methyl]-1H-pyrazolo[3,4-d]pyrimid in-4-amine (120 mg, 235 μmol, 1.0 equiv) in DMA (2.34 mL) was added DIPEA (163 μL, 940 μmol, 4.0 equiv) followed by PyBOP (158 mg, 305 μmol, 1.3 equiv). The resulting solution was stirred at room temperature for 3 h then purified by reverse phase HPLC (10→ 98% MeCN + 0.1% formic acid/H 2 O + 0.1% formic acid) to afford the product (82.7 mg, 47% yield). LCMS (ESI) m/z: [M + H] calcd for C 35 H 43 BrN 8 O 6 : 751.26; found 751.2.

[00718] Following General Procedure 5, but using the appropriate halide containing PEG carboxylic acid and amine containing active site inhibitors in Table 2, the Intermediates B3 in Table 10 were prepared:

Table 10. Additional PEG halides prepared

General Procedure 6: Displacement of a PEG halide with an amine containing post tinker and deprotection of the amine.

Step 1:

[00719] To a 0.1 M solution of halide containing PEG (1.0 equiv) in MeCN was added K 2 CO 3 (3.0 equiv) followed by amine containing post linker (1.2 equiv). The resulting suspension was heated to 80 °C and stirred until consumption of the PEG halide, as indicated by LCMS analysis. The reaction was cooled to room temperature and then purified by silica gel chromatography to afford the product.

Step 2:

[00720] To a 0.07 M solution of N-Boc protected amine (1.0 equiv) in dioxane w'as added HCl (4 M in dioxane, 10.0 equiv). The reaction was stirred until consumption of N-Boc protected amine, as indicated by LCMS analysis. The reaction was then concentrated under reduced pressure to afford the product. intermediate B2-4, 18-{6-[(4-amino-3-{1H-pyrrolo[2,3-b]pyridin-5-yI}-1H-pyrazol o[3,4- d]pyrimidin-1-yl)methyl]-1,2,3,4-tetrahydroisoquinolm-2-yl}- 1-(4-{5H,6H,7H,8H- pyrido[4,3-d]pyrimidm-2-yl}piperazm-1-yl)-3,6,9,12,15-pentao xaoctadecan-18-one

Step 1: Synthesis of tert-butyl 2-[4-(18-{6-[(4-amino-3-{ 1H-pyrrolo[2,3-b]pyridin-5-yl}-1H- pyrazolo[3,4-d]pyrimidin-1-yl)rnethyl]-1,2,3,4-tetrahydroiso qmnolm-2-yl}-18-oxo- 3,6,9,12,15-pentaoxaoctadecan-1-yl)piperazin-1-yl]-5H,6H,7H, 8H-pyrido[4,3-d]pyrimidine- 6-carboxylate [00721] To a suspension of 18-{6-[(4-amino-3-{1H-pyrrolo[2,3-b]pyridin-5-yl}-1H- pyrazolo[3 ,4-d]pyrimidin-1-yl)methyl] -1,2,3 ,4-tetrahydroisoquinolin-2-yl}-1-bromo- 3,6,9, 12,15-pentaoxaoctadecan-18-one (82.7 mg, 110 μmol, 1.0 equiv) in MeCN (1.09 mL) was added K 2 CO 3 (45.6 mg, 330 μmol, 3.0 equiv) followed by tort-butyl 2-(piperazin-1-yl)- 5H,6H,7H,8H-pyrido[4,3-d]pyrimidine-6-carboxylate (42.1 mg, 132 μmol, 1.2 equiv). The resulting suspension was heated to 80 °C for 8 h, then purified by silica gel chromatography (O→20% MeOH/DCM) to afford the product (75.1 mg, 70% yield). LCMS (ESI) m/z: [M + H] calcd for C 51 H 67 N 13 O 8 : 990.53; found 990.5.

Step 2: Synthesis of 18-{6-[(4-amino-3-{1H-pyrralo[2,3-b]pyridin-5-yl}-1H-pyrazol o[3,4- d]pyrimidin-1-yl)methyl]-1,2,3,4-tetrahydroisoquinolin-2-yl} -1-(4-{5H,6H,7H,8H- pyrido[4,3-d]pyrimidin-2-yl}piperazin-1-yl)-3,6,9,12,15-pent aoxaoctadecan-18-one

[00722] To a solution of tort-butyl 2-[4-(18-{6-[(4-amino-3-{1H-pyrrolo[2,3-b]pyridin-5- yl}-1H-pyrazolo[3,4-d]pyrimidin-1-yl)methyl]-1,2,3,4-tetrahy droisoquinolin-2-yl}-18-oxo- 3,6,9,12,15-pentaoxaoctadecan-1-yl)piperazin-1-yl]-5H,6H,7H, 8H-pyrido[4,3-d]pyrimidine- 6-carboxylate (75.1 mg, 75.8 μmol, 1.0 equiv) in dioxane (1 mL) was added HCl (4 M in dioxane, 472 μL, 1.89 mmol, 10.0 equiv). The solution was stirred at room temperature for 45 min, then concentrated under reduced pressure to afford the product. LCMS (ESI) m/z: [M + Na] calcd for C 46 H 59 N 13 O 6 : 912.46; found 912.5.

[00723] Following General Procedure 6, but using the appropriate PEG carboxylic acid and amine containing active site inhibitors in Table 2, the Intermediates B2 in Table 11 were prepared:

Table 11. Additional amines prepared

[00724] Following General Procedure 1, but using the appropriate carboxylic acid PEG tort-butyl ester and amine containing active site inhibitors in Table 2, the Intermediates B4 in Table 12 were prepared:

Table 12. Additional amines prepared

[00725] Following General Procedure 1, but using the appropriate Intermediates B4 in Table 12 and amine containing pre-linkers in Table 4, the Intermediates B2 in Table 13 were prepared:

Table 13. Additional amines prepared

[00726] Following General Procedure 1, but using the appropriate Intermediates A1 and amine containing pre-linkers in Table 4, the Intermediates B2 in Table 14 were prepared:

Table 14. Additional amines prepared

[00727] Following General Procedure 2, but using the appropriate 4-nitrophenyl carbonate containing rapamycin monomer in Table 1 and Intermediates B2 from Tables 9, 11, and 13 and 14, the Series 2 bivalent analogs in Table 15 were synthesized:

Table 15. Series 2 Bivalent Compounds:

[00728] Following General Procedure 1, but using the appropriate amine containing active site inhibitors in Table 2 and amine containing pre-linkers in Table 4, the Intermediates C1 in Table 16 were prepared:

Table 16. Additional amines prepared

[00729] Following General Procedure 1, but using the PEG carboxylic acids and Intermediates C1 in Table 16, the Intermediates C2 in Table 17 were prepared:

Table 17. Additional amines prepared

[00730] Following General Procedure 2, but using the appropriate 4-nitrophenyI carbonate containing rapamycin monomer in Table 1 and Intermediates C2 from Table 17, the Series 3 bivalent analogs in Table 18 were synthesized:

Table 18. Series 3 Bivalent Compounds

[00731] Following General Procedure 1, but using the appropriate Intermediates C2 in Table 17 and amine containing pre-linkers in Table 4, the Intermediates D1 in Table 19 were prepared:

Table 19. Additional amines prepared [00732] Following General Procedure 1, but using the appropriate amine containing active site inhibitors in Table 2 and amine containing pre-linkers in Table 4, the intermediates D1 in Table 20 were prepared:

Table 20. Additional amines prepared

[00733] Following General Procedure 2, but using the appropriate 4-nitrophenyl carbonate containing rapamycin monomer in Table 1 and Intermediates D1 from Tables 19 and 20, the Series 4 bivalent analogs in Table 21 were synthesized:

Table 21. Series 4 Bivalent Compounds

1 s

[00734] Following General Procedure 1, but using the appropriate Intermediates C1 in Table 16 and amine containing pre-linkers in Table 4, the Intermediates E1 in Table 22 were prepared:

Table 22. Additional amines prepared

[00735] Following General Procedure 2, but using the appropriate 4-nitrophenyl carbonate containing rapamycin monomer in Table 1 and intermediates El from Table 22, the Series 5 bivalent analogs in Table 23 were synthesized: Table 23. Series 5 Bivalent Compounds Table 24. Additional amines prepared

[00736] Following General Procedure 2, but using the appropriate 4-nitrophenyl carbonate containing rapamycin monomer in Table 1 and Intermediates FI from Table 24, the Series 6 bivalent analogs in Table 25 were synthesized:

Table 25. Series 6 Bivalent Compounds

[00737] Following General Procedure 1, but using the appropriate Intermediates A1 in Table 5 and amine containing pre-linkers in Table 4, the Intermediates G1 in Table 26 were prepared:

Table 26. Additional amines prepared

[00738] Following General Procedure 6, but using the appropriate Intermediates B3 in Table 10 and amine containing pre-linkers in Table 4, the Intermediates G1 in Table 27 were prepared:

Table 27. Additional amines prepared

[00739] Following General Procedure 2, but using the appropriate 4-nitrophenyl carbonate containing rapamycin monomer in Table 1 and Intermediates G1 from Tables 26 and 27, the Series 7 bivalent analogs in Table 28 were synthesized:

Table 28. Series 7 Bivalent Compounds

[00740] Following General Procedure 1, but using the appropriate Intermediates D1 in Tables 19 and 20 and PEG carboxylic acids, the Intermediates H1 in Table 29 were prepared:

Table 29. Additional amines prepared

[00741] Following General Procedure 2, but using the appropriate 4-nitrophenyl carbonate containing rapamycin monomer in Table 1 and Intermediates H1 from Table 29, the Series 8 bivalent analogs in Table 30 were synthesized:

Table 30. Series 8 Bivalent Compounds:

1 /· "* 7/L Biological Examples

Cell Based AlphaLISA Assays For Determining IC50 For Inhibition of P-Akt (S473), P- 4E-BP1 (T37/46), and P-P70S6K (T389) in MDA-MB-468 Cells mTOR Kinase Cellular Assay

[00742] To measure functional activity of mTORC 1 and mTORC2 in cells the phosphorylation of 4EBP1 (Thr37/46) and P70S6K (Thr389), and AKT1/2/3 (Ser473) was monitored using AlphaLisa SureFire Ultra Kits (Perkin Elmer), MDA-MB-468 cells (ATCC© HTB-132) were cultured in 96-well tissue culture plates and treated with compounds in the disclosure at concentrations varying from 0.017 - 1,000 nM for two to four hours at 37 °C. Incubations were terminated by removal of the assay buffer and addition of lysis buffer provided with the assay kit. Samples were processed according to the manufacturer’s instructions. The Alpha signal from the respective phosphoproteins was measured in duplicate using a microplate reader (Envision, Perkin-Elmer or Spectramax M5, Molecular Devices). Inhibitor concentration response curves were analyzed using normalized IC 50 regression curve fitting with control based normalization.

[00743] As an example, measured IC 50 values for selected compounds are reported below:

[00744] As an example, measured pIC 50 values for selected compounds are reported below:

Note:

Equivalents

[00745] While the present disclosure has been described in conjunction with the specific embodiments set forth above, many alternatives, modifications and other variations thereof will be apparent to those of ordinary skill in the art. All such alternatives, modifications and variations are intended to fall within the spirit and scope of the present disclosure.